force microscopy - the eyethe-eye.eu/public/books/medical/texts/force microscopy - applicatio… ·...

303

Upload: others

Post on 02-Oct-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United
Page 2: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

FORCE MICROSCOPY

Page 3: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

FORCE MICROSCOPYAPPLICATIONS IN BIOLOGYAND MEDICINE

Edited by

BHANU P. JENA PhDDepartment of PhysiologyWayne State University School of MedicineDetroit, Michigan

J. K. HEINRICH HORBER PhDDepartment of PhysicsUniversity of BristolBristol, United Kingdom

A JOHN WILEY & SONS, INC., PUBLICATION

Page 4: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

Copyright 2006 by John Wiley & Sons, Inc. All rights reserved.

Published by John Wiley & Sons, Inc., Hoboken, New Jersey.Published simultaneously in Canada.

No part of this publication may be reproduced, stored in a retrieval system, or transmitted in any form or by anymeans, electronic, mechanical, photocopying, recording, scanning, or otherwise, except as permitted underSection 107 or 108 of the 1976 United States Copyright Act, without either the prior written permission of thePublisher, or authorization through payment of the appropriate per-copy fee to the Copyright Clearance Center,Inc., 222 Rosewood Drive, Danvers, MA 01923, (978) 750-8400, fax (978) 750-4470, or on the web atwww.copyright.com. Requests to the Publisher for permission should be addressed to the Permissions Department,John Wiley & Sons, Inc., 111 River Street, Hoboken, NJ 07030, (201) 748-6011, fax (201) 748-6008, or online athttp://www.wiley.com/go/permission.

Limit of Liability/Disclaimer of Warranty: While the publisher and author have used their best efforts in preparingthis book, they make no representations or warranties with respect to the accuracy or completeness of the contentsof this book and specifically disclaim any implied warranties of merchantability or fitness for a particular purpose.No warranty may be created or extended by sales representatives or written sales materials. The advice andstrategies contained herein may not be suitable for your situation. You should consult with a professional whereappropriate. Neither the publisher nor author shall be liable for any loss of profit or any other commercialdamages, including but not limited to special, incidental, consequential, or other damages.

For general information on our other products and services or for technical support, please contact our CustomerCare Department within the United States at (800) 762-2974, outside the United States at (317) 572-3993 orfax (317) 572-4002.

Wiley also publishes its books in a variety of electronic formats. Some content that appears in print may not beavailable in electronic formats. For more information about Wiley products, visit our web site at www.wiley.com.

Library of Congress Cataloging-in-Publication Data:

Force microscopy : applications in biology and medicine / [edited by] Bhanu P. Jena, J.K.Heinrich Horber.

p. cm.Includes bibliographical references and index.ISBN-13: 978-0-471-39628-4ISBN-10: 0-471-39628-11. Medical electronics. 2. Scanning force microscopy. 3. Scanning probe microscopy.

4. Nanotechnology. I. Jena, Bhanu P. II. Horber, J. K. Heinrich.

R856.A2F675 2006610.28—dc22

2005058118

Printed in the United States of America

10 9 8 7 6 5 4 3 2 1

Page 5: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

CONTENTS

Preface vii

Contributors ix

Chapter 1. Porosome: The Universal Secretory Machinery in Cells 1

Bhanu P. Jena

Chapter 2. Molecular Mechanism of SNARE-Induced Membrane Fusion 25

Bhanu P. Jena

Chapter 3. Molecular Mechanism of Secretory Vesicle Content ExpulsionDuring Cell Secretion 37

Bhanu P. Jena

Chapter 4. Fusion Pores in Growth-Hormone-Secreting Cells of thePituitary Gland: An AFM Study 49

Lloyd L. Anderson and Bhanu P. Jena

Chapter 5. Properties of Microbial Cell Surfaces Examined by AtomicForce Microscopy 69

Yves F. Dufrene

Chapter 6. Scanning Probe Microscopy of Plant Cell Wall and ItsConstituents 95

Ksenija Radotic, Miodrag Micic, and Milorad Jeremic

Chapter 7. Cellular Interactions of Nano Drug Delivery Systems 113

Rangaramanujam M. Kannan, Omathanu Pillai Perumal,and Sujatha Kannan

v

Page 6: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

vi CONTENTS

Chapter 8. Adapting AFM Techniques for Studies on Living Cells 137

J. K. Heinrich Horber

Chapter 9. Intermolecular Forces of Leukocyte Adhesion Molecules 159

Xiaohui Zhang and Vincent T. Moy

Chapter 10. Mechanisms of Avidity Modulation in Leukocyte AdhesionStudied by AFM 169

Ewa P. Wojcikiewicz and Vincent T. Moy

Chapter 11. Resolving the Thickness and Micromechanical Propertiesof Lipid Bilayers and Vesicles Using AFM 181

Guangzhao Mao and Xuemei Liang

Chapter 12. Imaging Soft Surfaces by SFM 201

Andreas Janke and Tilo Pompe

Chapter 13. High-Speed Atomic Force Microscopy of Biomoleculesin Motion 221

Tilman E. Schaffer

Chapter 14. Atomic Force Microscopy in Cytogenetics 249

S. Thalhammer and W. M. Heckl

Chapter 15. Atomic Force Microscopy in the Study of MacromolecularInteractions in Hemostasis and Thrombosis: Utility forInvestigation of the Antiphospholipid Syndrome 267

William J. Montigny, Anthony S. Quinn, Xiao-Xuan Wu, Edwin G. Bovill,Jacob H. Rand, and Douglas J. Taatjes

Index 287

Page 7: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

PREFACE

Throughout history, the development of newimaging tools has provided new insights intoour perceptions of the living world and pro-foundly impacted human health and disease.The invention of the light microscope almost300 years ago was the first catalyst, pro-pelling us into an era of modern biology andmedicine. Using the light microscope, a giantstep into the gates of biology and medicinewas made with the discovery of the unit oflife, the cell. The structure and morphologyof normal and diseased cells, and of disease-causing microorganisms, were revealed forthe first time using the light microscope.Then in 1938, with the birth of the elec-tron microscope (EM), a new scientific erabegan. Through the 1950s, a number of sub-cellular organelles were discovered and theirfunctions determined using the EM. Viruses,the new life forms, were identified andobserved for the first time and were impli-cated in diseases ranging from the commoncold to autoimmune disease (AIDS). Despitethe capability of the EM to image biolog-ical samples at near-nanometer resolution,sample processing resulting in morphologicalalterations remained a major concern. Thenin the mid-1980s, scanning probe microscopyevolved, extending our perception of the liv-ing world even further, to the near-atomicrealm. One such scanning probe microscope,the atomic force microscope (AFM), hashelped overcome the limitations of both lightand electron microscopy, enabling three-dimensional determination of the structureand dynamics of single biomolecules andlive cells, at near-angstrom resolution. This

unique capability of the AFM, in combina-tion with conventional tools and techniques,has finally provided an understanding of thecell, subcellular organelles, and biomolecularstructure–function, as never before.

Secretion and membrane fusion are funda-mental cellular processes regulating intracel-lular transport, plasma membrane recycling,cell division, sexual reproduction, acid secre-tion, histamine release, and the release ofenzymes, hormones, and neurotransmitters,to name just a few. Therefore, defects insecretion and membrane fusion lead to dia-betes, Alzheimer’s disease, Parkinson’s dis-ease, and a host of other diseases. The firstthree chapters by Jena describe the discov-ery of the molecular machinery and mecha-nism of cell secretion and membrane fusionin cells. These discoveries were made usingthe combined approaches of AFM, EM, x-raydiffraction, photon-correlation spectroscopy,molecular biology, biochemistry, and elec-trophysiology. The secretory machinery theporosome described in the first chapter, wasfirst discovered in live secretory pancreaticacinar cells, using the AFM. Similarly, theAFM has been instrumental in the discov-ery of the porosome in numerous secretorycells as described in Chapter 1, includingthe growth-hormone-secreting cell of thepituitary gland, described in Chapter 4 byAnderson and Jena. In Chapter 5, Dufrenedescribes probing the structural and physicalproperties of microbial cell surfaces usingAFM. In Chapter 6, Radotic et al. reviewthe current state-of-the-art of scanning probemicroscopic characterization of higher plantcell wall and its components. The spatial and

vii

Page 8: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

viii PREFACE

temporal control of the delivery of drugs atthe site of action on a cellular or subcellulartarget is hugely important in the treatmentof diseases. In recent years, nanostructuredassemblies offer great promise. In Chapter 7,Kannan et al. provide case studies and dis-cuss the chemistry of engineered dendrimers,as multifunctional nanodrug delivery par-ticles. An overview of AFM techniquesfor studies on living cells is beautifullysummarized in Chapter 8. Besides imag-ing, force microscopy has frequently beenused to understand inter- and intramolec-ular interactions. In Chapter 9, Zhang andMoy describe how AFM can be used toprobe the intermolecular forces of leuko-cyte adhesion molecules. Leukocytes circu-lating in the blood vessels need to exitthe bloodstream to enter specific tissues orareas of inflammation. This find-tuned pro-cess is mediated by the interactions betweenleukocyte adhesion molecules and their adhe-sive partners expressed on the inner surfaceof blood vessel. If this adhesive processis not under proper control, it could leadto severe physiological disorders such asasthma and atherosclerosis. Similarly, inChapter 10, Wojcikiewicz and Moy exam-ine protein–protein interactions using the

AFM. Study of the structure, morphology,and stability of lipid bilayers and vesi-cles is important to the understanding ofdrug delivery, gene therapy, and biosensordesign. In Chapter 11 by Mao and Liang, themicromechanical properties of lipid bilayersand vesicles are examined using the AFM.Finally, in the last four chapters by Jankeand Pompe (Chapter 12), Schaffer (Chapter13), Thalhammer and Heckl (Chapter 14),and Montigny et al. (Chapter 15), the devel-opment of new approaches in the use offorce microscopy in biology and medicine isdiscussed.

The examples provided in this book on theutility of force microscopy in biology andmedicine, which by no means are exhaus-tive, give the reader some perspective onthe power and scope of this new scientifictool. The development of the photonic forcemicroscope discussed in Chapter 8, alongwith the use of force microscopy in com-bination with EM, electrophysiology, bio-chemistry, and so on (Chapter 1–3), has hadenormous impact in our study of cellular andphysiological processes. Nonetheless, we arejust beginning to realize the enormous bene-fits of force microscopy to fields of biologyand medicine.

Bhanu P. Jena

J. K. Heinrich Horber

Page 9: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

CONTRIBUTORS

Lloyd L. Anderson, Department of Ani-mal Science, College of Agriculture, andDepartment of Biomedical Sciences, Col-lege of Veterinary Medicine, Iowa StateUniversity, Ames, Iowa

Edwin G. Bovill, Department of Pathology,College of Medicine, University of Ver-mont, Burlington, Vermont

Yves F. Dufrene, Unite de chimie des inter-faces, Universite catholique de Louvain,Croix du Sud 2/18, B-1348 Louvain-la-Neuve, Belgium

W. M. Heckl, Department fur Geo-und Um-weltwissenschaften, University of Mun-chen, Theresienstr.41, Munich, Germany

J. K. Heinrich Horber, Department ofPhysics, University of Bristol, Bristol,United Kingdom

Andreas Janke, Leibriz-Institut fur Poly-merforschung Dresden eV., Hohe Str. 6,01005 Dresden, Germany

Bhanu P. Jena, Department of Physiol-ogy, Wayne State University School ofMedicine, Detroit, Michigan

Milorad Jeremic, Faculty of PhysicalChemistry, University of Belgrade, Stu-dentski Trg 16, 11000 Belgrade, Serbiaand Montenegro

Rangaramanujam M. Kannan, Depart-ment of Chemical Engineering and Mate-rial Science, and Biomedical Engineer-ing, Wayne State University, Detroit,Michigan

Sujatha Kannan, Department of CriticalCare Medicine, Children’s Hospital ofMichigan, Detroit, Michigan

Xuemei Liang, Department of ChemicalEngineering and Materials Science, WayneState University, 5050 Anthony WayneDrive, Detroit, Michigan

Guangzhao Mao, Department of Chemi-cal Engineering and Materials Science,Wayne State University, 5050 AnthonyWayne Drive, Detroit, Michigan

Miodrag Micic, MP Biomedicals, Inc., 15Morgan, Irvine, California, 92618-2005

William J. Montigny, Microscopy ImagingCenter, College of Medicine, Universityof Vermont, Bulington, Vermont

Vincent T. Moy, Department of Phys-iology and Biophysics, University ofMiami Miller School of Medicine, Miami,Florida

Omathanu Pillai Perumal, Department ofPharmaceutical Sciences, South DakotaState University, College of Pharmacy,Brookings, South Dakota

Tilo Pompe, Leibriz-Institut fur Polymer-forschung Dresden eV., Hohe Str. 6,01005 Dresden, Germany

Anthony S. Quinn, Department of Pathol-ogy and Microscopy Imaging Center, Col-lege of Medicine, University of Vermont,Burlington, Vermont

Ksenija Radotic, Center for Multidisci-plinary Studies, University of Belgrade,Kneza Viseslava 1, 11000 Belgrade, Ser-bia and Montenegro

Jacob H. Rand, Department of Pathology,Montefiore Medical Center, Albert Ein-stein College of Medicine, Bronx, NewYork

ix

Page 10: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

x CONTRIBUTORS

Tilman E. Schaffer, Center for Nanotech-nology and Institute of Physics, West-falische Wilhelms-Universitat Munster,Gievenbecker Weg 11, 48149 Munster,Germany

Douglas J. Taatjes, Department of Pathol-ogy and Microscopy Imaging Center, Col-lege of Medicine, University of Vermont,Burlington, Vermont

S. Thalhammer, Department fur Geo-undUmweltwissenschaften, University ofMunchen, Theresienstr.41, Munich Ger-many; GSF—National Center for Envi-ronment and Health, Institute of

Radiation Protection, Ingolstadter Land-strasse 1, 85764 Neuherberg, Germany

Ewa P. Wojcikiewicz, Department of Phys-iology and Biophysics, University ofMiami Miller School of Medicine, Miami,Florida

Xiao-Xuan Wu, Department of Pathology,Montefiore Medical Center, Albert Ein-stein College of Medicine, Bronx, NewYork

Xiaohui Zhang, Department of Physiologyand Biophysics, University of MiamiSchool of Medicine, Miami, Florida

Page 11: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

CHAPTER 1

POROSOME: THE UNIVERSAL SECRETORYMACHINERY IN CELLSBHANU P. JENADepartment of Physiology, Wayne State University School of Medicine, Detroit, Michigan

1.1. INTRODUCTION

Secretion and membrane fusion are funda-mental cellular processes regulating endo-plasmic reticulum (ER)–Golgi transport,plasma membrane recycling, cell division,sexual reproduction, acid secretion, and therelease of enzymes, hormones, and neuro-transmitters, to name just a few. It is there-fore no surprise that defects in secretion andmembrane fusion give rise to diseases suchas diabetes, Alzheimer’s, Parkinson’s, acutegastroduodenal diseases, gastroesophagealreflux disease, intestinal infections due toinhibition of gastric acid secretion, biliarydiseases resulting from malfunction of secre-tion from hepatocytes, polycystic ovariandisease as a result of altered gonadotropinsecretion, and Gitelman disease associatedwith growth hormone deficiency and distur-bances in vasopressin secretion. Understand-ing cellular secretion and membrane fusionnot only helps to advance our understand-ing of these vital cellular and physiologicalprocesses, but also helps in the develop-ment of drugs to ameliorate secretory defects,provides insight into our understanding ofcellular entry and exit of viruses and otherpathogens, and helps in the developmentof smart drug delivery systems. Therefore,

secretion and membrane fusion play animportant role in health and disease. Stud-ies (Abu-Hamdah et al., 2004; Anderson,2004; Cho et al., 2002a–f, 2004; Horberand Miles, 2003; Jena, 1997, 2002–2004;Jena et al., 1997, 2003; Jeremic et al., 2003,2004a,b; Kelly et al., 2004; Schneider et al.,1997) in the last decade demonstrate thatmembrane-bound secretory vesicles dockand transiently fuse at the base of specializedplasma membrane structures called poro-somes or fusion pores, to expel vesicular con-tents. These studies further demonstrate thatduring secretion, secretory vesicles swell,enabling the expulsion of intravesicularcontents through porosomes (Abu-Hamdahet al., 2004; Cho et al., 2002f; Jena et al.,1997; Kelly et al., 2004). With these find-ings (Abu-Hamdah et al., 2004; Anderson,2004; Cho et al., 2002a–f, 2004; Horberand Miles, 2003; Jena, 1997, 2002–2004;Jena et al., 1997, 2003; Jeremic et al., 2003,2004a,b; Kelly et al., 2004; Schneider et al.,1997) a new understanding of cell secre-tion has emerged and confirmed by a num-ber of laboratories (Aravanis et al., 2003;Fix et al., 2004; Lee et al., 2004; Taraskaet al., 2003; Thorn et al., 2004; Tojima et al.,2000).

Force Microscopy: Applications in Biology and Medicine, edited by Bhanu P. Jena and J.K. Heinrich Horber.Copyright 2006 John Wiley & Sons, Inc.

1

Page 12: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

2 POROSOME: THE UNIVERSAL SECRETORY MACHINERY IN CELLS

Throughout history, the development ofnew imaging tools has provided new insightsinto our perceptions of the living world andhas profoundly impacted human health. Theinvention of the light microscope almost300 years ago was the first catalyst, pro-pelling us into the era of modern biologyand medicine. Using the light microscope, agiant step into the gates of modern medicinewas made by the discovery of the unit oflife, the cell. The structure and morphologyof normal and diseased cells and of disease-causing microorganisms were revealed forthe first time using the light microscope.Then in 1938, with the birth of the elec-tron microscope (EM), dawned a new erain biology and medicine. Through the mid-1940s and 1950s, a number of subcellularorganelles were discovered and their func-tions determined using the EM. Viruses,the new life forms, were discovered andobserved for the first time and were impli-cated in diseases ranging from the com-mon cold to acquired immune deficiencysyndrome (AIDS). Despite the capabilityof the EM to image biological samples atnear-nanometer resolution, sample process-ing (fixation, dehydration, staining) results inmorphological alterations and was a majorconcern. Then in the mid-1980s, scanningprobe microscopy evolved (Binnig et al.,1986; Horber and Miles, 2003), furtherextending our perception of the living worldto the near atomic realm. One such scanningprobe microscope, the atomic force micro-scope (AFM), has helped overcome bothlimitations of light and electron microscopy,enabling determination of the structure anddynamics of single biomolecules and livecells in 3D, at near-angstrom resolution. Thisunique capability of the AFM has givenrise to a new discipline of “nanobioscience,”heralding a new era in biology and medicine.Using AFM in combination with conven-tional tools and techniques, this past decadehas witnessed advances in our understand-ing of cell secretion (Abu-Hamdah et al.,2004; Anderson, 2004; Cho et al., 2002a–f,

2004; Horber and Miles, 2003; Jena, 1997,2002–2004; Jena et al., 1997, 2003; Jeremicet al., 2003, 2004a,b; Kelly et al., 2004;Schneider et al., 1997) and membrane fusion(Cho et al., 2002d; Jeremic et al., 2004a,b;Weber et al., 1998), as noted earlier in thechapter.

The resolving power of the light micro-scope is dependent on the wavelength of thelight used; therefore, 250–300 nm in lateralresolution, and much less in depth resolu-tion, can be achieved at best. The porosomeor fusion pore in live secretory cells are cup-shaped structures, measuring 100–150 nm atits opening and 15–30 nm in relative depthin the exocrine pancreas, and just 10 nm atthe presynaptic membrane of the nerve ter-minal. As a result, it had evaded visual detec-tion until its discovery using the AFM (Choet al., 2002a–c, 2003, 2004; Jeremic et al.,2003; Schneider et al., 1997). The develop-ment of the AFM (Binnig et al., 1986) hasenabled the imaging of live cells in physio-logical buffer at nanometer to subnanometerresolution. In AFM, a probe tip microfab-ricated from silicon or silicon nitride andmounted on a cantilever spring is used toscan the surface of the sample at a con-stant force. Either the probe or the samplecan be precisely moved in a raster patternusing an xyz piezo tube to scan the sur-face of the sample (Fig. 1.1). The deflectionof the cantilever measured optically is usedto generate an isoforce relief of the sam-ple (Alexander et al., 1989). Force is thusused to image surface profiles of objects bythe AFM, allowing imaging of live cells andsubcellular structures submerged in physio-logical buffer solutions. To image live cells,the scanning probe of the AFM operates inphysiological buffers and may do so undertwo modes: contact or tapping. In the con-tact mode, the probe is in direct contactwith the sample surface as it scans at a con-stant vertical force. Although high-resolutionAFM images can be obtained in this modeof AFM operation, sample height informa-tion generated may not be accurate since

Page 13: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

METHODS 3

Laser SourceMonitor

Computer

Fluid Chamber

OscillatingCantilever

Position SensitivePhotosensor

Figure 1.1. Schematic diagram depicting key components of an atomic force microscope.

the vertical scanning force may depress thesoft cell. However, information on the vis-coelastic properties of the cell and the springconstant of the cantilever enables measure-ment of the cell height. In tapping modeon the other hand, the cantilever resonatesand the tip makes brief contacts with thesample. In the tapping mode in fluid, lateralforces are virtually negligible. It is thereforeimportant that the topology of living cellsbe obtained using both contact and tappingmodes of AFM operation in fluid. The scan-ning rate of the tip over the sample also playsan important role on the quality of the image.Since cells are soft samples, a high scan-ning rate would influence its shape. Hence,a slow tip movement over the cell wouldbe ideal and results in minimal distortionand better image resolution. Rapid cellularevents may be further monitored by usingsection analysis. To examine isolated cells bythe AFM, freshly cleaved mica coated withCel-Tak have also been used with great suc-cess (Cho et al., 2002a–c; Jena et al., 2003;Jeremic et al., 2003; Schneider et al., 1997).Also, to obtain optimal resolution, the con-tents of the bathing medium as well as thecell surface to be scanned should be devoidof any debris.

1.2. METHODS

1.2.1. Isolation of PancreaticAcinar Cells

Acinar cells for secretion experiments,light microscopy, atomic force microscopy(AFM), and electron microscopy (EM) wereisolated using minor modification of apublished procedure. For each experiment,a male Sprague–Dawley rat weighing80–100 g was euthanized by CO2 inhalation.The pancreas was dissected and dicedinto 0.5-mm3 pieces with a razor blade,mildly agitated for 10 min at 37◦C ina siliconized glass tube with 5 ml ofoxygenated buffer A (98 mM NaCl, 4.8 mMKCl, 2 mM CaCl2, 1.2 mM MgCl2, 0.1%bovine serum albumin, 0.01% soybeantrypsin inhibitor, 25 mM Hepes, pH 7.4)containing 1000 units of collagenase.The suspension of acini was filteredthrough a 224-µm Spectra-Mesh (SpectrumLaboratory Products, Rancho Dominguez,CA) polyethylene filter to remove largeclumps of acini and undissociated tissue. Theacini were washed six times, 50 ml per wash,with ice-cold buffer A. Isolated rat pancreaticacini and acinar cells were plated onCell-Tak-coated (Collaborative BiomedicalProducts, Bedford, MA) glass coverslips.Two to three hours after plating, cells were

Page 14: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

4 POROSOME: THE UNIVERSAL SECRETORY MACHINERY IN CELLS

imaged with the AFM before and duringstimulation of secretion. Isolated acinar cellsand hemi-acinar preparations were used inthe study because fusion of secretory vesiclesat the PM in these cells occurs at the apicalregion facing the acinar lumen.

1.2.2. Pancreatic Plasma MembranePreparation

Rat pancreatic PM fractions were iso-lated using a modification of a publishedmethod. Male Sprague–Dawley rats weigh-ing 70–100 g were euthanized by CO2

inhalation. Pancreas were removed andplaced in ice-cold phosphate-buffered saline(PBS), pH 7.5. Adipose tissue was removedand the pancreas were diced into 0.5-mm3

pieces using a razor blade in a few dropsof homogenization buffer A (1.25 M sucrose,0.01% trypsin inhibitor, and 25 mM Hepes,pH 6.5). The diced tissue was homogenizedin 15% (w/v) ice-cold homogenization bufferA using four strokes at maximum speedof a motor-driven pestle (Wheaton over-head stirrer). One-and-a-half milliliters of thehomogenate was layered over a 125-µl cush-ion of 2 M sucrose and 500 µl of 0.3 Msucrose was layered onto the homogenate inBeckman centrifuge tubes. After centrifuga-tion at 145,000 × g for 90 min in a SorvallAH-650 rotor, the material banding betweenthe 1.2 and 0.3 M sucrose interface wascollected and the protein concentration wasdetermined. For each experiment, fresh PMwas prepared and used the same day in allAFM experiments.

1.2.3. Isolation of Synaptosomes,Synaptosomal Membrane andSynaptic Vesicles

Synaptosomes, synaptosomal membrane andsynaptic vesicles were prepared from ratbrains (Jeong et al., 1998; Thoidis et al.,1998). Whole rat brain from Sprague–Dawley rats (100–150 g) was isolated andplaced in ice-cold buffered sucrose solu-tion (5 mM Hepes pH 7.4, 0.32 M sucrose)

supplemented with protease inhibitor cock-tail (Sigma, St. Louis, MO) and homoge-nized using Teflon-glass homogenizer (8–10strokes). The total homogenate was cen-trifuged for 3 min at 2500 × g. The super-natant fraction was further centrifuged for15 min at 14,500 × g, and the resultant pelletwas resuspended in buffered sucrose solu-tion, which was loaded onto 3–10–23%Percoll gradients. After centrifugation at28,000 × g for 6 min, the enriched synapto-somal fraction was collected at the 10–23%Percoll gradient interface. To isolate synapticvesicles and synaptosomal membrane (32),isolated synaptosomes were diluted with 9vol of ice-cold H2O (hypotonic lysis ofsynaptosomes to release synaptic vesicles)and immediately homogenized with threestrokes in Dounce homogenizer, followed bya 30-min incubation on ice. The homogenatewas centrifuged for 20 min at 25,500 × g,and the resultant pellet (enriched synaptoso-mal membrane preparation) and supernatant(enriched synaptic vesicles preparation) wereused in our studies.

1.2.4. Preparation of Lipid Membraneon Mica and Porosome Reconstitution

To prepare lipid membrane on mica forAFM studies, freshly cleaved mica diskswere placed in a fluid chamber. Two hundredmicroliters of the bilayer bath solution,containing 140 mM NaCl, 10 mM HEPES,and 1 mM CaCl2, was placed at the center ofthe cleaved mica disk. Ten microliters of thebrain lipid vesicles was added to the abovebath solution. The mixture was then allowedto incubate for 60 min at room temperature,before washing (×10), using 100-µl bathsolution/wash. The lipid membrane on micawas imaged by the AFM before and after theaddition of immunoisolated porosomes.

1.2.5. Atomic Force Microscopy

“Pits” and fusion pores at the PM in livepancreatic acinar secreting cells in PBS pH

Page 15: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

METHODS 5

7.5 were imaged with the AFM (BioscopeIII, Digital Instruments) using both contactand tapping modes. All images presentedin this manuscript were obtained in the“tapping” mode in fluid, using silicon nitridetips with a spring constant of 0.06 N·m−1 and an imaging force of <200 pN.Images were obtained at line frequencies of1 Hz, with 512 lines per image and constantimage gains. Topographical dimensions of“pits” and fusion pores at the cell PMwere analyzed using the software nanoscopeIIIa4.43r8 supplied by Digital Instruments.

1.2.6. ImmunoAFM on Live Cells

Immunogold localization in live pancreaticacinar cells was assessed after a 5-minstimulation of secretion with 10 µM of thesecretagogue, mastoparan. After stimulationof secretion, the live pancreatic acinar cellsin buffer were exposed to at 1:200 dilutionof α-amylase-specific antibody (BiomedaCorp., Foster City, CA) and 30 nm of goldconjugated secondary antibody for 1 min andwere washed in PBS before AFM imagingin PBS at room temperature. “Pits” andfusion pores within, at the apical end of livepancreatic acinar cells in PBS pH 7.5, wereimaged by the AFM (Bioscope III, DigitalInstruments) using both contact and tappingmode. All images presented were obtainedin the “tapping” mode in fluid, using siliconnitride tips as described previously.

1.2.7. ImmunoAFM on Fixed Cells

After stimulation of secretion with 10 µMmastoparan, the live pancreatic acinar cellswere fixed for 30 min using ice-cold 2.5%paraformaldehyde in PBS. Cells were thenwashed in PBS, followed by labeling with1:200 dilution of α-amylase-specific anti-body (Biomeda Corp.) and 10 nm of goldconjugated secondary antibody for 15 min,fixed, washed in PBS, and imaged in PBS atroom temperature using the AFM.

1.2.8. Isolation of Zymogen Granules

ZGs were isolated by using a modifica-tion of the method of our published proce-dure. Male Sprague–Dawley rats weighing80–100 g were euthanized by CO2 inhala-tion for each ZG preparation. The pancreaswas dissected and diced into 0.5-mm3 pieces.The diced pancreas was suspended in 15%(w/v) ice-cold homogenization buffer (0.3 Msucrose, 25 mM Hepes, pH 6.5, 1 mM ben-zamidine, 0.01% soybean trypsin inhibitor)and homogenized with a Teflon glass homog-enizer. The resultant homogenate was cen-trifuged for 5 min at 300 × g at 4◦C to obtaina supernatant fraction. One volume of thesupernatant fraction was mixed with 2 volof a Percoll–Sucrose–Hepes buffer (0.3 Msucrose, 25 mM Hepes, pH 6.5, 86% Per-coll, 0.01% soybean trypsin inhibitor) andcentrifuged for 30 min at 16,400 × g at4◦C. Pure ZGs were obtained as a loosewhite pellet at the bottom of the centrifugetube.

1.2.9. Transmission ElectronMicroscopy

Isolated rat pancreatic acini and ZGs werefixed in 2.5% buffered paraformaldehyde(PFA) for 30 min, and the pellets wereembedded in Unicryl resin and were sec-tioned at 40–70 nm. Thin sections weretransferred to coated specimen TEM grids,dried in the presence of uranyl acetate andmethyl cellulose, and examined in a trans-mission electron microscope.

1.2.10. Immunoprecipitation andWestern Blot Analysis

Immunoblot analysis was performed on pan-creatic PM and total homogenate fractions.Protein in the fractions was estimated by theBradford method (Bradford, 1976). Pancre-atic fractions were boiled in Laemmli reduc-ing sample preparation buffer (Laemmli,1970) for 5 min, cooled, and used for SDS-PAGE. PM proteins were resolved in a 12.5%

Page 16: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

6 POROSOME: THE UNIVERSAL SECRETORY MACHINERY IN CELLS

SDS-PAGE and electrotransferred to 0.2-µmnitrocellulose sheets for immunoblot analy-sis with a SNAP-23 specific antibody. Thenitrocellulose was incubated for 1 h at roomtemperature in blocking buffer (5% non-fat milk in PBS containing 0.1% Triton X-100 and 0.02% NaN3), and immunoblot-ted for 2 h at room temperature with theSNAP-23 antibody (ABR, Golden, CO). Theprimary antibodies were used at a dilu-tion of 1:10,000 in blocking buffer. Theimmunoblotted nitrocellulose sheets werewashed in PBS containing 0.1% Triton X-100 and 0.02% NaN3 and were incubated for1 h at room temperature in HRP-conjugatedsecondary antibody at a dilution of 1:2,000in blocking buffer. The immunoblots werethen washed in the PBS buffer, processed forenhanced chemiluminescence, and exposedto X-OMAT-AR film. To isolate the fusioncomplex for immunoblot analysis, SNAP-23 specific antibody conjugated to pro-tein A-sepharose was used. One gram oftotal pancreatic homogenate solubilized inTriton/Lubrol solubilization buffer (0.5%Lubrol; 1 mM benzamidine; 5 mM ATP;5 mM EDTA; 0.5% Triton X-100, in PBS)supplemented with protease inhibitor mix(Sigma, St. Louis, MO) was used. SNAP-23 antibody conjugated to the protein A-sepharose was incubated with the solubilizedhomogenate for 1 h at room temperaturefollowed by washing with wash buffer(500 mM NaCl, 10 mM TRIS, 2 mM EDTA,pH = 7.5). The immunoprecipitated sampleattached to the immuno-sepharose beads wasincubated in Laemmli sample preparationbuffer—prior to 12.5% SDS-PAGE, electro-transfer to nitrocellulose, and immunoblotanalysis—using specific antibodies to actin(Sigma), fodrin (Santa Cruz BiotechnologyInc., Santa Cruz, CA), vimentin (Sigma,St. Louis, MO), syntaxin 2 (Alomone Labs,Jerusalem, Israel), Ca2+-β 3 (Alomone Labs),and Ca2+-α 1c (Alomone Labs).

1.3. POROSOME: A NEWCELLULAR STRUCTURE

Earlier electrophysiological studies on mastcells suggested the existence of fusion poresat the cell plasma membrane (PM), whichbecame continuous with the secretory vesiclemembrane following stimulation of secretion(Monck et al., 1995). AFM has confirmedthe existence of the fusion pore or porosomeas permanent structures at the cell plasmamembrane and has revealed its morphol-ogy and dynamics in the exocrine pancreas(Cho et al., 2002a; Jena et al., 2003; Jeremicet al., 2003; Schneider et al., 1997), neuroen-docrine cells (Cho et al., 2002b,c), and neu-rons (Cho et al., 2004), at near-nanometerresolution and in real time.

Isolated live pancreatic acinar cells inphysiological buffer, when imaged with theAFM (Cho et al., 2002a; Jena et al., 2003;Jeremic et al., 2003; Schneider et al., 1997),reveal at the apical PM a group of circu-lar “pits” measuring 0.4–1.2 µm in diam-eter which contain smaller ‘depressions’(Fig. 1.2). Each depression averages between100 and 150 nm in diameter, and typically3–4 depressions are located within a pit.The basolateral membrane of acinar cells isdevoid of either pits or depressions. High-resolution AFM images of depressions inlive cells further reveal a cone-shaped mor-phology. The depth of each depression conemeasures 15–30 nm. Similarly, growth hor-mone (GH)-secreting cells of the pituitarygland and chromaffin cells, β cells of theexocrine pancreas, mast cells, and neuronspossess depressions at their PM, suggestingtheir universal presence in secretory cells.Exposure of pancreatic acinar cells to a sec-retagogue (mastoparan) results in a time-dependent increase (20–35%) in depressiondiameter, followed by a return to restingsize on completion of secretion (Cho et al.,2002a; Jena et al., 2003; Jeremic et al., 2003;Schneider et al., 1997) (Fig. 1.3). No demon-strable change in pit size is detected follow-ing stimulation of secretion (Schneider et al.,

Page 17: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

POROSOME: A NEW CELLULAR STRUCTURE 7

Figure 1.2. (A) On the far left is an AFM micrograph depicting “pits” and “depressions” within, at the plasmamembrane in live pancreatic acinar cells. On the right is a schematic drawing depicting depressions, at the cellplasma membrane, where membrane-bound secretory vesicles dock and fuse to release vesicular contents (Schneideret al., 1997). (B) Electron micrograph depicting a porosome close to a microvilli (MV) at the apical plasma membrane(PM) of a pancreatic acinar cell. Note association of the porosome membrane and the zymogen granule membrane(ZGM) of a docked zymogen granule (ZG), the membrane-bound secretory vesicle of exocrine pancreas. Also across section of the ring at the mouth of the porosome is seen.

Page 18: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

8 POROSOME: THE UNIVERSAL SECRETORY MACHINERY IN CELLS

Figure 1.3. Dynamics of depressions following stimulation of secretion. The top panel shows a number ofdepressions within a pit in a live pancreatic acinar cell. The scan line across three depressions in the top panelis represented graphically in the middle panel and defines the diameter and relative depth of the depressions; themiddle depressions are represented by arrowheads. The bottom panel represents percent of total cellular amylaserelease in the presence and absence of the secretagogue Mas 7. Notice an increase in the diameter and depth ofdepressions, correlating with an increase in total cellular amylase release at 5 min after stimulation of secretion.At 30 min after stimulation of secretion, there is a decrease in diameter and depth of depressions, with no furtherincrease in amylase release over the 5-min time point. No significant increase in amylase secretion or depressionsdiameter were observed in resting acini or those exposed to the nonstimulatory mastoparan analog Mas 17 (Jena,2002; Schneider et al., 1997).

1997). Enlargement of depression diameterand an increase in its relative depth afterexposure to secretagogues correlated withincreased secretion. Conversely, exposure ofpancreatic acinar cells to cytochalasin B, afungal toxin that inhibits actin polymeriza-tion, results in a 15–20% decrease in depres-sion size and a consequent 50–60% lossin secretion (Schneider et al., 1997). Resultsfrom these studies suggested depressions tobe the fusion pores in pancreatic acinar cells.Furthermore, these studies demonstrate theinvolvement of actin in regulation of both thestructure and function of depressions. Anal-ogous to pancreatic acinar cells, examinationof resting GH-secreting cells of the pitu-itary (Cho et al., 2002b) and chromaffin cellsof the adrenal medulla (Cho et al., 2002c)

also reveal the presence of pits and depres-sions at the cell PM (Fig. 1.4). The presenceof porosomes in neurons, in β cells of theendocrine pancreas, and in mast cells havealso been demonstrated (Figs. 1.4–1.8) (Choet al., 2004; Jena, 2004). Depressions in rest-ing GH cells measure 154 ± 4.5 nm (mean ±SE) in diameter. Exposure of GH cells toa secretagogue results in a 40% increasein depression diameter (215 ± 4.6 nm; p <

0.01) but no appreciable change in pitsize. The enlargement of depression diam-eter during secretion and the known effectthat actin depolymerizing agents decreasedepression size and inhibit secretion (Schnei-der et al., 1997) suggested depressions tobe the fusion pores. However, a moredirect demonstration that porosomes arefunctional supramolecular complexes came

Page 19: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

POROSOME: A NEW CELLULAR STRUCTURE 9

A B

C D

Figure 1.4. AFM micrograph of depressions or porosomes or fusion pores in the live secretory cell of the exocrinepancreas (A, B), the growth hormone (GH)-secreting cell of the pituitary (C), and in the chromaffin cell (D). Notethe “pit” (white arrowheads at the margin) with four depressions (arrowhead at 12 o’ clock). A high-resolution AFMmicrograph of one porosome is shown in part B. Bars = 40 nm for parts A and B. Similarly, AFM micrographs ofporosomes in β cell of the endocrine pancreas and mast cell have also been demonstrated.

Figure 1.5. Porosomes or fusion pores in β cells of the endocrine pancreas. (A) AFM micrograph of a pit withthree porosomes (arrowheads) at the PM in a live β cell. (B) Section analysis through a porosome at rest (top trace),during secretion (bottom trace), and following completion of secretion (middle trace). (C) Exposure of β cells tothe actin depolymerizing agent cytochalasin B results in decreased “porosome” size (not shown), accompanied by aloss in insulin secretion, as detected by immunoblot analysis of the cell incubation medium.

Page 20: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

10 POROSOME: THE UNIVERSAL SECRETORY MACHINERY IN CELLS

Figure 1.6. Electron micrograph of porosomes in neurons. (A) Electron micrograph of a synaptosome demonstratingthe presence of 40- to 50-nm synaptic vesicles. (B–D) Electron micrograph of neuronal porosomes, which are 10- to15-nm cup-shaped structures at the presynaptic membrane (arrowhead), where synaptic vesicles transiently dock andfuse to release vesicular contents. (E) Atomic force micrograph of a fusion pore or porosome at the nerve terminal ina live synaptosome. (F) Atomic force micrograph of isolated neuronal porosome, reconstituted into lipid membrane.

from immuno-AFM studies demonstratingthe specific localization of secretory prod-ucts at the porosomes, following stimula-tion of secretion (Figs. 1.9–1.12) (Jeremicet al., 2003). ZGs fused with the porosome-reconstituted bilayer, as demonstrated by

an increase in capacitance and conductanceand in a time-dependent release of the ZGenzyme amylase from cis to the trans com-partment of the bilayer chamber. Amylase isdetected using immunoblot analysis of thebuffer in the cis and trans chambers, using

Page 21: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

POROSOME: A NEW CELLULAR STRUCTURE 11

Resting Stimulated Amylase Ab-gold Fixed Amylase Ab-gold

0−75

750

0.2 0.4µm

0.6 0.8 0 0.2 0.4µm

0.6 0.8 0 0.2 0.4µm

0.6 0.8

nm−7

575

0nm

−75

750

nm

A B C

0.51.0

1.5

2.0µm

D

Figure 1.7. Depressions are fusion pores or porosomes. Porosomes dilate to allow expulsion of intra vesicularcontents. (A, B) AFM micrographs and section analysis of a pit and two out of the four fusion pores or porosomes,demonstrating enlargement following stimulation of secretion. (C) Exposure of live cells to gold conjugated-amylaseantibody (Ab) results in specific localization of immuno-gold to the porosome opening. (arrowhead) Amylase is oneof the proteins within secretory vesicles of the exocrine pancreas. (D) AFM micrograph of a fixed pancreatic acinarcell, demonstrating a pit and porosomes within labeled with amylaze-immunogold. Arrowheads point to immunogoldclusters (Cho et al., 2002a) at the pit and porosomes within.

a previously characterized amylase specificantibody (Cho et al., 2002a). As observedin immunoblot assays of isolated porosomes,chloride channel activities are also detectedwithin the reconstituted porosome complex.Furthermore, the chloride channel inhibitorDIDS was found to inhibit current activ-ity in the porosome-reconstituted bilayer.In summary, these studies demonstrate thatthe porosome in the exocrine pancreas isa 100- to 150-nm-diameter supramolecularcup-shaped lipoprotein basket at the cell PM,where membrane-bound secretory vesiclesdock and fuse to release vesicular contents.Similar studies have now been performedin neurons, demonstrating both the struc-tural (Fig. 1.6E,F) and functional reconstitu-tion of the isolated neuronal porosome com-plex. The biochemical composition of theneuronal porosome has also been determined(Cho et al., 2004).

Similar to the isolation of porosomesfrom the exocrine pancreas, neuronal poro-somes were immunoisolated from detergent-solubilized synaptosome preparations, usinga SNAP-25 specific antibody. Electrophoreticresolution of the immunoisolates reveal thepresence of 12 distinct protein bands, as

determined by sypro protein staining ofthe resolved complex (Fig. 1.13A). Fur-thermore, electrotransfer of the resolvedporosomal complex onto nitrocellulose mem-brane, followed by immunoblot analy-sis using various antibodies, revealed 9proteins. In agreement with earlier find-ings, SNAP-25, the P/Q-type calciumchannel, actin, syntaxin-1, synaptotagmin-1, vimentin, the N -ethylmaleimide-sensitivefactor (NSF), the chloride channel CLC-3,and the alpha subunit of the heterotrimericGTP-binding Go were identified to con-stitute part of the complex. To testwhether the complete porosome complexwas immunoisolated, the immunoisolatewas reconstituted into lipid membraneprepared using brain dioleoylphosphatidyl-choline (DOPC) and dioleylphosphatidylser-ine (DOPS) in a ratio of 7:3. At lowresolution, the AFM reveals the immunoiso-lates to arrange in L- or V-shaped struc-tures (Fig. 1.13B, red arrowheads), whichat higher resolution demonstrates the pres-ence of porosomes in patches (Fig. 1.13C,D,green arrowhead), similar to what is observedat the presynaptic membrane in intact synap-tosomes. Further imaging the reconstituted

Page 22: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

12 POROSOME: THE UNIVERSAL SECRETORY MACHINERY IN CELLS

Figure 1.8. Morphology of the cytosolic side of the porosome revealed in AFM studies on isolated pancreaticplasma membrane (PM) preparations. (A) AFM micrograph of isolated PM preparation reveals the cytosolic end ofa pit with inverted cup-shaped structures, the porosome. Note the 600 nm in diameter ZG at the left-hand corner ofthe pit. (B) Higher magnification of the same pit showing clearly the 4 to 5 porosomes within. (C) The cytosolicend of a single porosome is depicted in this AFM micrograph. (D) Immunoblot analysis of 10 µg and 20 µg ofpancreatic PM preparations, using SNAP-23 antibody, demonstrates a single 23-kDa immunoreactive band. (E, F)The cytosolic side of the PM demonstrating the presence of a pit with a number of porosomes within, shown priorto (E) and following addition of the SNAP-23 antibody (F) Note the increase in height of the porosome cone baserevealed by section analysis (bottom panel), demonstrating localization of SNAP-23 antibody at the base of theporosome (Jeremic et al., 2003).

immunoisolate at greater resolutions usingthe AFM shows the presence of 8- to 10-nm porosomes (Fig. 1.13E). As observedin electron and AFM micrographs of thepresynaptic membrane in synaptosomes, theimmunoisolated and lipid-reconstituted poro-some reveals the presence of an approxi-mately 2 nm in diameter central plug. Thesestudies confirm the complete isolation andstructural reconstitution of the neuronal poro-some in artificial lipid bilayers. To under-stand the structure of the porosome at the

cytosolic side of the presynaptic membrane,isolated synaptosomal membrane prepara-tions were imaged by the AFM. Thesestudies reveal the architecture of the cytoso-lic part of porosomes. In synaptosomes,40- to 50-nm synaptic vesicles are arrangedin ribbons, as seen in the electron micro-graphs (longitudinal section, Fig. 1.14A, orin cross section Fig. 1.14B). Similarly, whenthe cytosolic domain of isolated synaptoso-mal membrane preparations were analyzedby the AFM, 40- to 50-nm synaptic vesicles

Page 23: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

POROSOME: A NEW CELLULAR STRUCTURE 13

Figure 1.9. SNAP-23 associated proteins in pancreatic acinar cells. Total pancreatic homogenate was immunopre-cipitated using the SNAP-23 specific antibody. The precipitated material was resolved using 12.5% SDS-PAGE,electrotransferred to nitrocellulose membrane and then probed using antibodies to a number of proteins. Associationof SNAP-23 with syntaxin2, with cytoskeletal proteins actin, α-fodrin, and vimentin, and calcium channels β3 andα1c, together with the SNARE regulatory protein NSF, is demonstrated (arrowheads). Lanes showing more than onearrowhead suggest presence of isomers or possible proteolytic degradation of the specific protein (Jena et al., 2003).

were also found to be arranged in ribbons(Fig. 1.14C,D), thus confirming EM observa-tions. On close examination (Fig. 1.14D–F)of docked synaptic vesicles (blue arrow-heads), synaptic vesicles are found attachedto the base of the 8- to 10-nm porosomes (redarrowheads). It is possible to see both theporosome and the attached synaptic vesicle,since while imaging with the AFM, synap-tic vesicles can be gently pushed away fromtheir docked sites to reveal the porosomelying beneath them (Fig. 1.14C–F). Addi-tionally, bare porosomes with no synapticvesicles attached are also found at the cytoso-lic side of synaptosomal membrane prepara-tions (Fig. 1.14D).

Calcium, target membrane proteinsSNAP-25 and syntaxin (t-SNARE), andsecretory vesicle-associated membrane pro-tein (v-SNARE) are the minimal machinery

involved in fusion of opposing bilayers(Jeremic et al., 2004a,b). NSF is an ATPasethat is suggested to disassemble the t-/v-SNARE complex in the presence of ATP(Jeong et al., 1998). To test this hypothe-sis and to further confirm and determine themorphology of neuronal porosomes, synap-tosomal membrane preparations with dockedsynaptic vesicles were imaged using theAFM in the presence and absence of ATP(Fig. 1.15A). As hypothesized, addition of50 µM ATP resulted in t-/v-SNARE dis-assembly and the release of docked vesi-cles (blue arrowhead) at the porosome patch(Fig. 1.15B,C, red arrowhead). At higherresolution, the base of the porosome isclearly revealed (Fig. 1.15D). At increasedimaging forces (300–500 pN instead of<200 pN), porosome patches (Fig. 1.15E,red arrowhead) and individual porosomes

Page 24: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

14 POROSOME: THE UNIVERSAL SECRETORY MACHINERY IN CELLS

Figure 1.10. Negatively stained electron micrograph and atomic force micrograph of the immunoisolated porosomecomplex. (A) Negatively stained electron micrograph of an immunoisolated porosome complex from solubilizedpancreatic plasma membrane preparations, using a SNAP-23 specific antibody. Note the three rings and the 10 spokesthat originate from the inner smallest ring. This structure represents the protein backbone of the porosome complex,since the three rings and the vertical spikes are observed in electron micrographs of cells and porosome co-isolatedwith ZGs. Bar = 30 nm. (B) Electron micrograph of the fusion pore complex, cut out from (A). (C) Outline of thestructure presented for clarity. (D–F) Atomic force micrograph of the isolated pore complex in near physiologicalbuffer. Bar = 30 nm. Note the structural similarity of the complex, imaged both by EM (G) and AFM (H). The EMand AFM micrographs are superimposable (I).

Figure 1.11. Electron micrographs of reconstituted porosome or fusion pore complex in liposomes, showing acup-shaped basket-like morphology. (A) A 500-nm vesicle with an incorporated porosome is shown. Note thespokes in the complex. (B–D) The reconstituted complex at greater magnification is shown. Bar represents 100 nm.

Page 25: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

POROSOME: A NEW CELLULAR STRUCTURE 15

Bilayer

+ −

EPC 9 Amplifier

400

200pF pA

0

1.0

0.5nA

00 5

msec10

0 3min

Cis Trans0 30 min

6 9

10

5

0

ZG Addition

CapacitanceCurrent

Cl−Cl− Channel

Blocker

Pore ComplexPore Complex + 20 µM DIDS

A

B

C

Figure 1.12. Lipid bilayer-reconstituted porosome complex is functional. (A) Schematic drawing of the EPC9 bilayersetup for electrophysiological measurements. (B) Zymogen granules (ZGs) added to the cis compartment of the bilayerchamber fuse with the reconstituted porosomes, as demonstrated by an increase in capacitance and current activities,and a concomitant time-dependent release of amylase (a major ZG content) to the trans compartment of the bilayerchamber. The movement of amylase from the cis to the trans side of the chamber was determined by immunoblotanalysis of the contents in the cis and the trans chamber over time. (C) As demonstrated by immunoblot analysis ofthe immunoisolated complex, electrical measurements in the presence and absence of chloride ion channel blockerDIDS demonstrate the presence of chloride channels in association with the complex, and its role in porosomefunction.

Page 26: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

16 POROSOME: THE UNIVERSAL SECRETORY MACHINERY IN CELLS

A

B C

D E

Figure 1.13. Composition of the neuronal porosome and its reconstitution in lipid membrane. (A) Proteinsimmunoisolated from detergent-solubilized synaptosomal membrane preparation, using a SNAP-25 specific antibody.Immunoisolates when resolved by SDS-PAGE, along with the resolved proteins in the gel stained using Syprodye, reveals 12 specific bands (•), suggesting the presence of at least 12 proteins in the complex, not includingthe heavy chain (*) of the SNAP-25 antibody. When the resolved proteins were electrotransferred to nitrocellulosemembrane and probed with various antibodies, calcium channel P/Q, actin, Gαo, syntaxin-1 (Syn1), synaptotagmin-1(Syt1), NSF, vimentin, and the chloride channel CLC-3 were identified (Bradford, 1976; Laemmli, 1970). (B–E)Atomic force micrographs at different resolution of the reconstituted immunoisolate in lipid membrane. (B) At lowmagnification, the immunoisolated complex arrange in L- or V-shaped structures (white arrowheads). (C, D) Withinthe V-shaped structures, patches (arrowheads) of porosomes are found. (E) Each reconstituted porosome is almostidentical to the porosome observed in intact synaptosomes. The central plug is clearly seen. This AFM micrographdemonstrates the presence of two porosomes (arrowhead), although only half of the second porosome is in view.

Page 27: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

MOLECULAR UNDERSTANDING OF CELL SECRETION 17

(Fig. 1.15F,G) were further defined in greaterstructural detail. At 5–8 A resolution, eightperipheral knobs at the porosomal opening(Fig. 1.15F–H, yellow arrowheads) and acentral plug (green arrowhead) at the basewere revealed. These studies further providea direct demonstration of synaptic vesiclesdocking at these sites and confirming themto be porosomes, where synaptic vesiclesdock and transiently fuse to release neuro-transmitters (Aravanis et al., 2003). Hencesynaptic vesicles are able to fuse transientlyand successively at porosomes in the presy-naptic membrane without loss of identity, asreported in earlier studies (Aravanis et al.,2003).

To be able to assess the functionalityof the reconstituted porosome preparations(Figs. 1.13E and 1.16A), an electrophysio-logical bilayer setup was used (Fig. 1.16B).Membrane capacitance was continuallymonitored throughout these experiments.Following reconstitution of the bilayer mem-brane with porosomes, isolated synaptic vesi-cles were added to the cis compartmentof the bilayer chamber. A large numberof synaptic vesicles fused at the bilayer,as demonstrated by the significant step-wise increases in the membrane capacitance(Fig. 1.16C,D). As expected from results inFig. 15B, addition of 50 µM ATP allowedt-/v-SNARE disassembly and the release ofdocked vesicles, resulting in the return ofthe bilayers membrane capacitance to restinglevels (Fig. 1.16C,D). Addition of recombi-nant NSF had no further effect on mem-brane capacitance. Thus, the associated NSFat the t-/v-SNARE complex is adequatefor complete disassembly of the SNAREcomplex for release of synaptic vesiclesfollowing transient fusion and the comple-tion of a round of neurotransmitter release.To further biochemically assess the releaseof docked synaptic vesicles following ATPtreatment, synaptosomal membrane prepa-rations were exposed to 50 µM ATP, andthe supernatant fraction was assessed forsynaptic vesicles by monitoring levels of the

synaptic vesicle proteins SV2 and VAMP-2(Fig. 1.16E). Our study demonstrates thatboth SV2 and VAMP-2 proteins are enrichedin supernatant fractions following exposureof isolated synaptosomal membrane to ATP(Fig. 1.16E). Thus, AFM, electrophysiologi-cal measurements, and immunoanalysis con-firmed the dissociation of porosome-dockedsynaptic vesicles following ATP exposure.As previously suggested (Jena, 1997, 2002),such a mechanism may allow for the multipletransient docking-fusion and release cyclesthat synaptic vesicles may undergo duringneurotransmission, without loss of vesicleidentity (Aravanis et al., 2003). The neuronalporosome, although an order of magnitudesmaller than those in the exocrine pancreasor in neuroendocrine cells, possesses manysimilarities both in structure and composi-tion. Thus, nature has designed the porosomeas a general secretory machinery, but hasfine-tuned it to suit various secretory pro-cesses in different cells. Hence, porosomesize may be a form of such fine-tuning. It iswell known that smaller vesicles fuse moreefficiently than larger ones, and hence thecurvature of both the secretory vesicle andthe porosome base would dictate the effi-cacy and potency of vesicle fusion at thecell plasma membrane. For example, becauseneurons are fast secretory cells, they pos-sess small (40–50 nm) secretory vesicles andporosome bases (2–4 nm) for rapid and effi-cient fusion. In contrast, a slow secretory celllike the exocrine pancreas possesses largersecretory vesicles (1000 nm in diameter) thatfuse at porosomes having bases that measure20–30 nm in diameter.

1.4. MOLECULAR UNDERSTANDINGOF CELL SECRETION

Fusion pores or porosomes as permanentstructures at the cell plasma membraneare present in all secretory cells exam-ined, ranging from exocrine, endocrine,and neuroendocrine cells to neurons, where

Page 28: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

18 POROSOME: THE UNIVERSAL SECRETORY MACHINERY IN CELLS

A

C D

E F

B

Figure 1.14

Page 29: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

MOLECULAR UNDERSTANDING OF CELL SECRETION 19

membrane-bound secretory vesicles dockand transiently fuse to expel vesicular con-tents. Porosomes in pancreatic acinar or GH-secreting cells are cone-shaped structuresat the plasma membrane, with a 100- to150-nm-diameter opening. Membrane-boundsecretory vesicles ranging in size from 0.2to 1.2 µm in diameter dock and fuse atporosomes to release vesicular contents. Fol-lowing fusion of secretory vesicles at poro-somes, only a 20–35% increase in porosomediameter is demonstrated. It is therefore rea-sonable to conclude that secretory vesicles“transiently” dock and fuse at the site. Incontrast to accepted belief, if secretory vesi-cles were to completely incorporate at poro-somes, the PM structure would distend muchwider than what is observed. Furthermore, ifsecretory vesicles were to completely fuseat the plasma membrane, there would be aloss in vesicle number following secretion.Examination of secretory vesicles withincells before and after secretion demonstratesthat the total number of secretory vesiclesremains unchanged following secretion (Choet al., 2002e; Lee et al., 2004). However, thenumber of empty and partially empty vesi-cles increases significantly, supporting theoccurrence of transient fusion. Earlier studieson mast cells also demonstrated an increasein the number of spent and partially spentvesicles following stimulation of secretion,without any demonstrable increase in cellsize. Similarly, secretory granules are recap-tured largely intact after stimulated exocy-tosis in cultured endocrine cells (Taraskaet al., 2003). Other support in evidence of

transient fusion is the presence of neurotrans-mitter transporters at the synaptic vesiclemembrane. These vesicle-associated trans-porters would be of little use if vesicleswere to fuse completely at the plasma mem-brane to be compensatorily endocytosed ata later time. In further support, a recentstudy reports that single synaptic vesiclesfuse transiently and successively without lossof vesicle identity (Aravanis et al., 2003).Although the fusion of secretory vesicles atthe cell plasma membrane occurs transiently,complete incorporation of membrane at thecell plasma membrane would occur whencells need to incorporate signaling moleculeslike receptors, second messengers, or ionchannels. Similarly, total fusion would occurintracellularly, where during protein trans-port and maturation, vesicles derived fromthe cis Golgi would completely fuse withthe trans Golgi apparatus. The discovery ofthe porosome, along with an understand-ing of the molecular mechanism of mem-brane fusion and the swelling of secretoryvesicles required for expulsion of vesicu-lar contents, provides an understanding ofsecretion and membrane fusion in cells atthe molecular level. These findings haveprompted many laboratories to work in thearea and further confirm these findings. Thus,the porosome is a supramolecular structureuniversally present in secretory cells, fromthe exocrine pancreas to the neurons, andin the endocrine to neuroendocrine cells,where membrane-bound secretory vesiclestransiently dock and fuse to expel intravesic-ular contents. Hence, the secretory process in

Figure 1.14. Arrangement of synaptic vesicles and porosomes at the presynaptic membrane. (A) Electron micrographof rat brain synaptosome demonstrating the ribbon-like arrangement (inset) of 40- to 50-nm synaptic vesicles. (B)Cross section of such a ribbon (inset) reveals the interaction between the synaptic vesicles. (C, D) Examination ofthe presynaptic membrane from the cytosolic side (inside out) using AFM, confirmed such a ribbon arrangementof docked synaptic vesicles (white arrowheads) at porosomes (grey arrowheads). Bare porosomes (lacking dockedsynaptic vesicles) are also seen. The AFM micrograph in part c is a 2-D image, and the one in part d is a 3-Dimage. (E, F) AFM micrograph of a docked synaptic vesicle at a porosome. During imaging using the AFM, theinteraction of the cantilever tip with the sample sometimes resulted in pushing away the docked synaptic vesicle,enough to expose the porosome lying beneath. AFM section analysis further reveals the size of synaptic vesicles(white section line and arrowheads) and porosomes (grey section line and arrowheads).

Page 30: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

20 POROSOME: THE UNIVERSAL SECRETORY MACHINERY IN CELLS

A B

C D

E F

Figure 1.15. AFM micrographs revealing the dynamics of docked synaptic vesicles at porosomes and the porosomearchitecture at 4–5 A resolution. (A) AFM micrograph of five docked synaptic vesicle at porosomes. (B) Additionof 50 µM ATP dislodges two synaptic vesicles at the lower left, and exposing the porosome patches. This alsodemonstrates that a single synaptic vesicle may dock at more than one porosome complex. (C–G) AFM micrographsobtained at higher imaging forces (300–500 pN rather than <200 pN) reveal porosomes architecture at greaterdetail. (C) AFM micrograph of one of the porosome patches where a synaptic vesicle was docked prior to ATPexposure. (D) Base of a single porosome. (E) High-force AFM micrograph of the cytosolic face of the presynapticmembrane, demonstrating the ribbon arrangement of porosome patches (grey arrowhead) and docked synaptic vesicles(white arrowheads). Note how the spherical synaptic vesicles are compressed and flattened at higher imaging forces.(F, G) At such higher imaging forces, porosomes reveal the presence of eight globular structures (white arrowhead)surrounding a central plug (grey arrowhead), as demonstrated in the (H) schematic diagram.

Page 31: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

MOLECULAR UNDERSTANDING OF CELL SECRETION 21

G H

Figure 1.15. (continued)

A

C

D E

B

Figure 1.16. Functional reconstitution of immunoisolated nuronal porosomes. (A) Schematic representation ofa porosome at the presynaptic membrane, with a docked synaptic vesicle (SV) at its base. (B) Schematicdrawing of an EPC9 electrophysiological bilayers apparatus, to continually monitor changes in the capacitanceof porosome-reconstituted membrane, when synaptic vesicles are introduced into the cis bilayers chamber followedby ATP and purified recombinant NSF protein. (C) Schematic representation of SV docking at the base of aporosome, fusing to release its contents, and disengaging in the presence of ATM. (D) Capacitance measurements ofporosome-reconstituted bilayers support the experiment in Fig. 1.15B and the schematic diagram in part c. Exposureof the reconstituted bilayers to SVs results in a dramatic increase in membrane capacitance, which drops to baselinefollowing exposure to 50 µM ATP. Recombinant NSF has no further effect (n = 6). (E) Similarly, in agreement,exposure of isolated synaptosomal membrane preparations to 50 µM ATP results in the release of SVs from themembrane into the incubation medium, as demonstrated by immunoblot analysis of the incubating medium usingthe SV-specific protein antibodies, SV2 and VAMP-2.

Page 32: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

22 POROSOME: THE UNIVERSAL SECRETORY MACHINERY IN CELLS

cells is a highly regulated event, orchestratedby a number of ions and biomolecules.

ACKNOWLEDGMENT

Supported by Grants DK-56212 and NS-39918 from the National Institutes of Health(BPJ).

REFERENCES

Abu-Hamdah, R., Cho, W. J., Cho, S. J., Jeremic,A., Kelly, M., Ilie, A. E., and Jena, B. P.(2004). Regulation of the water channelaquaporin-1: Isolation and reconstitution of theregulatory complex. Cell Biol. Int. 28:7–17(published on-line 2003).

Alexander, S., Hellemans, L., Marti, O., Schneir,J., Elings, V., and Hansma, P. K. (1989). Anatomic resolution atomic force microscopeimplemented using an optical lever. J. Appl.Phys. 65:164–167.

Anderson, L. L. (2004). Discovery of a newcellular structure—the porosome: Elucidationof the molecular mechanism of secretion. CellBiol. Int. 28:3–5.

Aravanis, A. M., Pyle, J. L., and Tsien, R. W.(2003). Single synaptic vesicles fusing tran-siently and successively without loss of iden-tity. Nature 423:643–647.

Bennett, V. (1990). Spectrin-based membraneskeleton: A multipotential adaptor betweenplasma membrane and cytoplasm. Physiol. Rev.70:1029–1065.

Binnig, G., Quate, C. F., and Gerber, C. H.(1986). Atomic force microscope. Phys. Rev.Lett. 56:930–933.

Bradford, M. M. (1976). A rapid and sensitivemethod for the quantitation of microgramquantities of protein utilizing the princi-ple of protein-dye binding. Anal. Biochem72:248–254.

Cho,S. J., Quinn, A. S., Stromer, M. H., Dash, S.,Cho, J., Taatjes, D. J., and Jena, B. P. (2002a).Structure and dynamics of the fusion pore inlive cells. Cell Biol. Int. 26:35–42.

Cho, S. J., Jeftinija, K., Glavaski, A., Jeftinija, S.,Jena, B. P., and Anderson, L. L. (2002b).

Structure and dynamics of the fusion poresin live GH-secreting cells revealed usingatomic force microscopy. Endocrinology 143:1144–1148.

Cho, S. J., Wakade, A., Pappas, G. D., and Jena,B. P. (2002c). New structure involved intransient membrane fusion and exocytosis. Ann.New York Acad. Sci. 971:254–256.

Cho, S. J., Kelly, M., Rognlien, K. T., Cho, J.,Hoerber, J. K. H., and Jena, B. P. (2002d).SNAREs in opposing bilayers interact ina circular array to form conducting pores.Biophys. J. 83:2522–2527.

Cho, S. J., Cho, J., and Jena, B. P. (2002e).The number of secretory vesicles remainsunchanged following exocytosis. Cell Biol. Int.26:29–33.

Cho, S. J., Sattar, A. K., Jeong, E-H., Satchi, M.,Cho, J. A., Dash, S., Mayes, M. S., Stromer,M. H., and Jena, B. P. (2002f ). Aquaporin 1regulates GTP-induced rapid gating of water insecretory vesicles. Proc. Natl. Acad. Sci. USA99:4720–4724.

Cho, W. J., Jeremic, A., Rognlien, K. T., Zhva-nia, M. G., Lazrishvili, I., Tamar, B., andJena, B. P. (2004). Structure, isolation, compo-sition and reconstitution of the neuronal fusionpore. Cell Biol. Int. 28:699–708 (published on-line August 25, 2004).

Faigle, W., Colucci-Guyon, E., Louvard, D.,Amigorena, S., and Galli, T. (2000). Vimentinfilaments in fibroblasts are a reservoirfor SNAP-23, a component of the mem-brane fusion machinery. Mol. Biol. Cell.11:3485–3494.

Fix, M., Melia, T. J., Jaiswal, J. K., Rappoport,J. Z., You, D., Sollner, T. H., Rothman, J. E.,and Simon, S. M. (2004). Imaging singlemembrane fusion events mediated by SNAREproteins. Proc. Natl. Acad. Sci. USA 101:7311–7316.

Gaisano, H. Y., Sheu, L., Wong, P. P., Klip, A.,and Trimble, W. S. (1997). SNAP-23 islocated in the basolateral plasma membraneof rat pancreatic acinar cells. FEBS Lett.414:298–302.

Goodson, H. V., Valetti, C., and Kreis, T. E.(1997). Motors and membrane traffic. Curr.Opin. Cell Biol. 9:18–28.

Page 33: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 23

Horber, J. K. H., and Miles, M. J. (2003). Scan-ning probe evolution in biology. Science302:1002–1005.

Jena, B. P. (1997). Exocytotic Fusion: Total ortransient. Cell Biol. Int. 21:257–259.

Jena, B. P. (2002). Fusion pore in live cells. NIPS17:219–222.

Jena, B. P. (2003). Fusion Pore: Structure anddynamics. J. Endo. 176:169–174.

Jena, B. P. (2004). Discovery of the Porosome:Revealing the molecular mechanism of secre-tion and membrane fusion in cells. J. Cell Mol.Med. 8:1–21.

Jena, B. P., Schneider, S. W., Geibel, J. P., Web-ster, P., Oberleithner, H., and Sritharan, K. C.(1997). Gi regulation of secretory vesicleswelling examined by atomic force microscopy.Proc. Natl. Acad. Sci. USA 94:13317–13322.

Jena, B. P., Cho, S. J., Jeremic, A., Stromer,M. H., and Abu-Hamdah, R. (2003). Structureand composition of the fusion pore. Biophys. J.84:1337–1343.

Jeong, E.-H., Webster, P., Khuong, C. Q., Sat-tar, A. K. M. A., Satchi, M., and Jena, B. P.(1998). The native membrane fusion machineryin cells. Cell Biol. Int. 22:657–670.

Jeremic, A., Kelly, M., Cho, S. J., Stromer,M. H., and Jena, B. P. (2003). Reconstitutedfusion pore. Biophys. J. 85:2035–2043.

Jeremic, A., Kelly, M., Cho, W. J., Cho, S. J.,Horber, J. K. H., and Jena, B. P. (2004a).Calcium drives fusion of SNARE-apposedbilayers. Cell Biol. Int. 28:19–31 (publishedon-line 2003).

Jeremic, A., Cho, W. J., and Jena, B. P. (2004b).Membrane fusion: What may transpire at theatomic level. J. Biol. Phys. Chem. 4:139–142.

Kelly, M., Cho, W. J., Jeremic, A., Abu-Hamdah,R., and Jena, B. P. (2004). Vesicle swellingregulates content expulsion during secretion.Cell Biol. Int. 28:709–716 (published on-lineAugust 25, 2004).

Laemmli, U. K. (1970). Cleavage of structuralproteins during the assembly of the head ofbacteriophage T4. Nature. 227:680–685.

Lee, J. S., Mayes, M. S., Stromer, M. H., Scanes,C. G., Jeftinija, S., and Anderson, L. L. (2004).

Number of secretory vesicles in growth hor-mone cells of the pituitary remains unchangedafter secretion. Exp. Biol. Med. 229:291–302.

Monck, J. R., Oberhauser, A. F., and Fernan-dez, J. M. (1995). The exocytotic fusion poreinterface: A model of the site of neurotransmit-ter release. Mol. Membr. Biol. 12:151–156.

Nakano, M., Nogami, S., Sato, S., Terano, A.,and Shirataki, H. (2001). Interaction of syn-taxin with α-fodrin, a major component of thesubmembranous cytoskeleton. Biochem. Bio-phys. Res. Commun. 288:468–475.

Ohyama, A., Komiya, Y., and Igarashi, M.(2001). Globular tail of myosin-V is boundto vamp/synaptobrevin. Biochem. Biophys. Res.Commun. 280:988–991.

Schneider, S. W., Sritharan, K. C., Geibel, J. P.,Oberleithner, H., and Jena, B. P. (1997). Sur-face dynamics in living acinar cells imagedby atomic force microscopy: Identification ofplasma membrane structures involved in exocy-tosis. Proc. Natl. Acad. Sci. USA 94:316–321.

Taraska, J. W., Perrais, D., Ohara-Imaizumi, M.,Nagamatsu, S., and Almers, W. (2003). Secre-tory granules are recaptured largely intact afterstimulated exocytosis in cultured endocrinecells. Proc. Natl. Acad. Sci. USA 100:2070–2075.

Thoidis, G., Chen, P., Pushkin, A. V., Vallega, G.,Leeman, S. E., Fine, R. E., and Kandror, K. V.(1998). Two distinct populations of synaptic-like vesicles from rat brain. Proc. Natl. Acad.Sci. USA. 95:183–188.

Thorn, P., Fogarty, K. E., and Parker, I. (2004).Zymogen granule exocytosis is characterizedby long fusion pore openings and preservationof vesicle lipid identity. Proc. Natl. Acad. Sci.USA 101:6774–6779.

Tojima,T., Yamane, Y., Takagi, H., Takeshita, T.,Sugiyama, T., Haga, H., Kawabata, K., Ushiki,T., Abe, K., Yoshioka, T., and Ito, E. (2000).Three-dimensional characterization of interiorstructures of exocytotic apertures of nerve cellsusing atomic force microscopy. Neuroscience101:471–481.

Weber, T., Zemelman, B. V., McNew, J. A.,Westerman, B., Gmachi, M., Parlati, F.,Sollner, T. H., and Rothman, J. E. (1998).SNAREpins: Minimal machinery for mem-brane fusion. Cell 92:759–772.

Page 34: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

CHAPTER 2

MOLECULAR MECHANISMOF SNARE-INDUCED MEMBRANE FUSIONBHANU P. JENADepartment of Physiology, Wayne State University School of Medicine, Detroit, Michigan

2.1. INTRODUCTION

Cell secretion involves the sequential inter-action of proteins in opposing bilayers. Theclassical concept of fusion is a three-stepprocess of cell excitation, docking, andfusion, in which docking may occur beforecell excitation. Membrane fusion in cellshas been known to be mediated via solu-ble N -ethylmaleimide-sensitive factor (NSF)-attachment protein receptors (SNAREs)(Weber et al., 1998). SNAREs are classifiedas v-SNARE and t-SNAREs, depending ontheir primary location either in secretory vesi-cle (v-) membrane or in target (t-) membranes(Rothman, 1994). Studies demonstrate that t-and v-SNARE-reconstituted lipid vesicles canfuse with one another, suggesting SNAREsto be the minimal membrane fusion machin-ery (Weber et al., 1988). The structure of theSNARE complex formed by interacting native(Jeong et al., 1998), or recombinant (Han-son et al., 1997; Sutton et al., 1998) t- andv-SNAREs, has been examined using electronmicroscopy (Jeong et al., 1998; Hanson et al.,1997) and x-ray crystallography (Sutton et al.,1998). However, the molecular mechanismof SNARE-induced membrane fusion was

unknown. Studies in the past 4–5 years, usingrecombinant SNARE proteins reconstitutedin lipid membrane, have for the first timeunraveled the molecular mechanism of mem-brane fusion in cells (Cho et al., 2002; Jeremicet al., 2004a,b). These discoveries were madeutilizing new approaches of atomic forcemicroscopy (AFM), in combination with con-ventional techniques like electrophysiology,electron microscopy (EM), x-ray diffraction,light scattering, and photon correlation spec-troscopy.

2.2. t-/v-SNAREs IN APPOSINGBILAYERS INTERACT IN A CIRCULARARRAY TO FORM CONDUCTINGCHANNELS

Although the partial structure (so-called coredomain) of the SNARE complex formedby interacting soluble t- and v-SNAREshad been examined using x-ray crystal-lography (Sutton et al., 1998), the struc-ture and arrangement of the t-/v-SNAREcomplex formed when full-length t-SNAREand v-SNARE present in opposing bilay-ers meet, was unknown. Examination of

Force Microscopy: Applications in Biology and Medicine, edited by Bhanu P. Jena and J.K. Heinrich Horber.Copyright 2006 John Wiley & Sons, Inc.

25

Page 35: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

26 MOLECULAR MECHANISM OF SNARE-INDUCED MEMBRANE FUSION

the structure and arrangement of purifiedrecombinant t- and v-SNAREs in artificiallipid bilayers, using AFM, has solved thisriddle. To further evaluate the establish-ment of SNARE-induced continuity betweenopposing bilayers, conductance and capac-itance of membranes in the presence andabsence of SNARE proteins was examined(Cho et al., 2002). If pore structures wereto form by direct addition of SNAREs toa single membrane, an increase in con-ductance would be observed due to themovement of ions through the pore. Todetermine the interaction between t- andv-SNAREs present in opposing bilayers,v-SNARE-reconstituted artificial lipid vesi-cles were exposed to t-SNARE-reconstituted

lipid membranes. The structure and arrange-ment of the SNARE complex formed as aresult, and any changes in capacitance orconductance were recorded using AFM andan EPC9 electrophysiological bilayer appara-tus (Cho et al., 2002). Results from the studydemonstrate that t-SNAREs and v-SNARE,when present in opposing bilayers, interactin a circular array, and in the presence ofCa2+, form conducting channels (Cho et al.,2002). The interaction of t-/v-SNARE pro-teins to form a conducting pore or chan-nel was found to be strictly dependent onthe presence of t-SNAREs and v-SNAREin opposing bilayers. Addition of puri-fied recombinant v-SNARE to a t-SNARE-reconstituted lipid membrane increased only

50100

150200

250nm

X 50 nm/divZ 5 nm/div

50100

150200

250

−5.0

5.0

0

nm

X 50 nm/divZ 5 nm/div

nm

0 100 nm

tSNAREs

A

E F

B C D

tSNAREs VAMP

2 MIN

10 p

A

Figure 2.1. AFM micrographs and force plots of mica and lipid surface and of SNAREs on lipid membrane.(A) AFM performed on freshly cleaved mica (left) and on lipid membrane formed on the same mica surface (right),demonstrating differences in the force–distance curves. Note the curvilinear shape exhibited in the force–distancecurves of the lipid surface in contrast to mica. Three-dimensional AFM micrographs of neuronal t-SNAREs depositedon the lipid membrane (B), and after the addition of v-SNARE (C). (D) Section analysis of the SNARE complexin parts B and C is depicted. Note that the smaller curve belonging to the t-SNARE complex in part B is markedlyenlarged after addition of v-SNARE. Artificial bilayer lipid membranes are nonconducting either in the presenceor absence of SNAREs (E, F). Current–time traces of bilayer membranes containing proteins involved in dockingand fusion of synaptic vesicles while the membranes are held at −60 mV (current/reference voltage). (E) Whent-SNAREs are added to the planar lipid bilayer containing the synaptic vesicle protein, VAMP-2, no occurrenceof current spike for fusion event at the bilayer membrane is observed (n = 7). (F) Similarly, no current spike isobserved when t-SNAREs (syntaxin 1A-1 and SNAP25) are added to the cis side of a bilayer chamber, followingwith VAMP-2. Increasing the concentration of t-SNAREs and VAMP-2 protein.

Page 36: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

AT THE ATOMIC LEVEL, HOW DOES Ca2+ PARTICIPATE IN MEMBRANE FUSION? 27

the size of the globular t-SNARE oligomerwithout influencing the electrical propertiesof the membrane (Cho et al., 2002). How-ever, when v-SNARE vesicles are added tot-SNARE reconstituted membrane, SNAREsassembles in a ring pattern (Figs. 2.1 and 2.2)and a stepwise increase in capacitance andconductance is observed (Fig. 2.3). Thus, t-and v-SNAREs are required to reside inopposing bilayers to allow appropriate t-/v-SNARE interactions leading to the estab-lishment of continuity between the opposingbilayers, only in the presence of calcium(Cho et al., 2002).

2.3. Ca2+ AND SNAREs ARE THEMINIMAL FUSION MACHINERYIN CELLS

Studies using SNARE-reconstituted lipo-somes and bilayers (Jeremic et al., 2004a)demonstrate a low fusion rate (τ = 16 min)between t- and v-SNARE-reconstituted lipo-somes in the absence of Ca2+; they alsoshow that exposure of t-/v-SNARE lipo-somes to Ca2+ drives vesicle fusion on anear-physiological relevant timescale (τ ∼10 s), demonstrating an essential role of Ca2+in membrane fusion. The Ca2+ effect onmembrane fusion in SNARE-reconstitutedliposomes is downstream of SNAREs, sug-gesting a regulatory role for Ca2+-bindingproteins in membrane fusion in the physio-logical state (Jeremic et al., 2004a). It is fur-ther demonstrated from these studies that inthe physiological state in cells, both SNAREsand Ca2+ operate as the minimal fusionmachinery (Figs. 2.4 and 2.5) (Jeremic et al.,2004a). Native and synthetic vesicles exhibita significant negative surface charge primar-ily due to the polar phosphate head groups.These polar head groups produce a repul-sive force, preventing aggregation and fusionof apposing vesicles. SNAREs bring oppos-ing bilayers closer to within a distance of2–3 A (Fig. 2.6), allowing Ca2+ to bridgethem (Jeremic et al., 2004a,b). The boundCa2+ then leads to the expulsion of water

between the bilayers at the bridging site,allowing lipid mixing and membrane fusion.Hence SNAREs, besides bringing opposingbilayers closer, dictate the site and size ofthe fusion area during cell secretion. Thesize of the t-/v-SNARE complex forming thechannel is dictated by the curvature of theopposing membranes, hence it depends onthe size of t-/v-SNARE-reconstituted vesi-cles. The smaller the vesicles, the smaller thechannel formed (unpublished observation).

2.4. AT THE ATOMIC LEVEL,HOW DOES Ca2+ PARTICIPATEIN MEMBRANE FUSION?

This important question has been resolvedby membrane fusion studies utilizing t-/v-SNARE reconstituted vesicles and mem-brane bilayers (Jeremic et al., 2004b). Cal-cium ion [Ca2+] is essential to life’s pro-cesses, and is present in every living cell.Ca2+ participates in diverse cellular pro-cesses, such as metabolism, secretion, pro-liferation, muscle contraction, cell adhesion,learning, and memory. Although calciumis present in abundance within cells, it iswell sequestered and is available only ondemand. Upon certain cellular stimulus forinstance, Ca2+ concentration at specific loca-tions (i.e., nano-environment) within the cellis elevated by several orders of magnitudewithin a brief period (some in < 1 ms). Thisprompt mobilization of Ca2+ is essential formany physiological processes, such as therelease of neurotransmitters or cell signaling.A unique set of chemical and physical prop-erties of the Ca2+ ion makes it ideal for per-forming these biochemical reactions. Ca2+ion exists in its hydrated state within cells.The properties of hydrated calcium havebeen extensively studied using x-ray diffrac-tion, neutron scattering, in combinationwith molecular dynamics simulations (Bakoet al., 2002; Chialvo and Simonson, 2003;Licheri et al., 1976; Schwenk et al., 2001).The molecular dynamic simulations include

Page 37: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

28 MOLECULAR MECHANISM OF SNARE-INDUCED MEMBRANE FUSION

nm

050

−50

0 1.00 2.00 nm

nm

050

−50

0 1.00 2.00nm

A

C D

B

11

2

2

3 µm

0.5

1.5

µm

Figure 2.2. Pore-like structures are formed when t-SNAREs and v-SNARE in opposing bilayers interact.(A) Unfused v-SNARE vesicles on t-SNARE reconstituted lipid membrane. (B) Dislodgement or fusion ofv-SNARE-reconstituted vesicles with a t-SNARE-reconstituted lipid membrane exhibits formation of pore-likestructures due to the interaction of v- and t-SNAREs in a circular array. The size of the pores range between50 and 150 nm (B–D). Several 3D AFM amplitude images of SNAREs arranged in a circular array (C) and someat higher resolution (D), illustrating a pore-like structure at the center is depicted. Scale bar is 100 nm. Recombinantt-SNAREs and v-SNARE in opposing bilayers drive membrane fusion. (E) When t-SNARE vesicles were exposedto v-SNARE-reconstituted bilayers, vesicles fused. Vesicles containing nystatin/ergosterol and t-SNAREs were addedto the cis side of the bilayer chamber. Fusion of t-SNARE containing vesicles with the membrane was observed ascurrent spikes that collapse as the nystatin spreads into the bilayer membrane. To determine membrane stability, thetransmembrane gradient of KCl was increased, allowing gradient-driven fusion of nystatin-associated vesicles.

three-body corrections compared with ab ini-tio quantum mechanics/molecular mechan-ics molecular dynamics simulations. Firstprinciples molecular dynamics has also beenused to investigate the structural, vibrational,and energetic properties of [Ca(H2O)n]2+clusters and the hydration shell of calcium

ion. These studies demonstrate that hydratedcalcium [Ca(H2O)n]2+ has more than oneshell around the Ca2+, with the first hydra-tion shell around the Ca2+ having sixwater molecules in an octahedral arrange-ment (Chialvo and Simonson, 2003). In stud-ies using light scattering and x-ray diffraction

Page 38: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

AT THE ATOMIC LEVEL, HOW DOES Ca2+ PARTICIPATE IN MEMBRANE FUSION? 29

E

tSnare Vesicles KCl

10 p

A

2 min

Fusion Events

Figure 2.2. (continued)

+ −

BilayerEPC9 Amplifier

400

200

00 2 4 minC

apac

itanc

e (p

F)

Con

duct

ance

(nS

)

Lipid Vesicles KCl nS

pF

A

B

Figure 2.3. Opposing bilayers containing t- and v-SNAREs, respectively, interact in a circular array to formconducting pores. (A) Schematic diagram of the bilayer–electrophysiology setup. (B) Lipid-vesicle-containingnystatin channels and both vesicles and membrane bilayer without SNAREs demonstrate no significant changesin capacitance and conductance. Initial increase in conductance and capacitance may be due to vesicle–membraneattachment. To demonstrate membrane stability (both bilayer membrane and vesicles), the transmembrane gradientof KCl was increased to allow gradient-driven fusion and a concomitant increase of conductance and capacitance.(C) When t-SNARE vesicles were added to a v-SNARE membrane support, the SNAREs in opposing bilayers formeda ring pattern, thereby forming pores (as seen in the AFM micrograph on the extreme right); stepwise increases incapacitance and conductance (−60-mV holding potential) were seen. Docking and fusion of the vesicle at the bilayermembrane opens vesicle-associated nystatin channels and SNARE-induced pore formation, allowing conductanceof ions from cis to the trans side of the bilayer membrane. Then further addition of KCl to induce gradient-drivenfusion resulted in little or no further increase in conductance and capacitance, demonstrating that docked vesicleshave already fused.

Page 39: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

30 MOLECULAR MECHANISM OF SNARE-INDUCED MEMBRANE FUSION

400

200

00 2 4 minC

apac

itanc

e (p

F)

Con

duct

ance

(nS

) tSNARELipid Vesicles

KClnS

pF

Pore

C

Figure 2.3. (continued)

of SNARE-reconstituted liposomes, it wasdemonstrated that fusion proceeds only whenCa2+ ions are available between the t- and v-SNARE-apposed bilayers (Fig. 2.7) (Jeremicet al., 2004b).

To monitor interaction(s) between Ca2+ions and phosphate on the lipid membranehead groups, the x-ray diffraction methodhas been utilized (Jeremic et al., 2004a,b).This experimental approach for monitoringinterbilayers contacts essentially requires thepresence of (i) highly concentrated lipid sus-pensions (>10 mM) favoring a multitude ofintervesicular contacts and (ii) fully hydratedliposomes, where vesicles have total free-dom to interact with each other in solution,hence establishing confined hydrated areasbetween adjacent bilayers. This small fluidspace could arise from interbilayer hydro-gen bond formation through water molecules(McIntosh, 2000) in addition to bridgingforces contributed by trans-SNARE com-plex formation (Cho et al., 2002; Jeremicet al., 2004a). When these two conditions aremet, liposomes diffract as shown (Fig. 2.6).Mixing of t- and v-SNARE liposomes inthe absence of Ca2+ leads to a diffuse andasymmetric diffractogram (depicted by thegray trace in Fig. 2.6), a typical character-istic of short-range ordering in a liquid sys-tem. In contrast, mixing the t-SNARE and

v-SNARE liposomes in the presence of Ca2+leads to a more structured diffractogram(depicted by the black trace in Fig 2.6) withan approximately 12% increase in x-ray scat-tering intensity, pointing to an increase in thenumber of contacts between apposing bilay-ers established presumably by calcium–PObridges, as previously suggested (Portiset al., 1979). The ordering effect of Ca2+ oninterbilayer contacts observed in x-ray stud-ies (Jeremic et al., 2004b) is in good agree-ment with recent light, AFM, and spectro-scopic studies, suggesting close apposition ofPO lipid head groups in the presence of Ca2+ions followed by formation of Ca2+ –PObridges between adjacent bilayers (Jeremicet al., 2004a; Laroche et al., 1991). X-raystudy shows that effect of Ca2+ on bilayerorientation and interbilayer contacts is mostprominent in the area of 3 A, with theappearance of an additional peak (shoulder)at 2.8 A (depicted by the arrow in Fig 2.6),both of which are within the ionic radius ofCa2+ (Jeremic et al., 2004b). These studiessuggest that the ionic radius of Ca2+ mayplay an important role in membrane fusion.But there remained a major spatial prob-lem, which was recently resolved (Jeremicet al., 2004b). As discussed earlier, calciumions [Ca2+] exist in their hydrated statewithin cells. Hydrated calcium [Ca(H2O)n]2+

Page 40: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

AT THE ATOMIC LEVEL, HOW DOES Ca2+ PARTICIPATE IN MEMBRANE FUSION? 31

A

B

Figure 2.4. Fluorimetric fusion assays demonstrate theability of Ca2+ to induce rapid lipid mixing of plain(AV) and SNARE-associated vesicles. Addition of5 mM Ca2+ to liposomal solution significantly increasesthe fusion of plain and SNARE-associated vesicles(+P<0.05, Student t-test between AV and AV + Ca2+or t-/v-SNARE-AV + Ca2+, n = 5). Note the inabilityof SNAREs in the absence of Ca2+ to significantlyinduce vesicle fusion (P > 0.1, Student t-test betweenAV and t-/v-SNARE-AV, n = 5). Incorporation oft-/v-SNAREs at the vesicles membrane increases theoverall yield but does not alter the rate of Ca2+-inducedmembrane fusion (A). The graph depicts the first-orderkinetics of SNAREs vesicle fusion in the presence andabsence of Ca2+ (B).

has more than one shell around the Ca2+,with the first hydration shell having sixwater molecules in an octahedral arrange-ment (Chialvo and Simonson, 2003), andmeasuring >6 A (Fig. 2.7). Studies revealthat for hydrated Ca2+ ion, depending onits coordination number, the nearest averageneighbor Ca2+ –O and Ca2+ –H distances

are at r ∼ 2.54 A and r ∼ 3.2 A in thefirst hydration shell, respectively. How then,would a hydrated calcium ion measuring>6 A fit between the 2.8- to 3-A space estab-lished by t-/v-SNAREs, between the appos-ing vesicle bilayers? One possibility wouldbe that calcium has to be present in thebuffer solution when t-SNARE vesicles andv-SNARE vesicles meet. If t- and v-SNAREvesicles are allowed to mix in a calcium-freebuffer, prior to the addition of calcium, nofusion should occur, since the 2.8- to 3-Aspace would not be able to accommodate thehydrated calcium. This hypothesis was tested(Jeremic et al., 2004b) and was found to beindeed correct. Light scattering experiments(Fig. 2.7) were performed on t-SNARE- andv-SNARE-reconstituted phospholipids vesi-cles, in the presence and absence of cal-cium and in the presence of NSF + ATP.NSF (N -ethylmalemide-sensitive factor) isan ATPase that is known to disassemble thet-/v-SNARE complex. Using light scatteringmeasurements, aggregation and membranefusion of lipid vesicles can be monitoredon the second timescale (Jeremic et al.,2004a; Wilschut et al., 1980). The initialrapid increase in intensity of light scatter-ing was found to be initiated on exposure oft-SNARE vesicles to v-SNARE vesicles insolution, followed by a slow decay of lightscattering (Fig. 2.7) due to vesicle–vesicleinteraction and settling. These studies showthat if t-SNARE vesicles and v-SNARE vesi-cles are allowed to interact prior to cal-cium addition (depicted by arrow, Fig. 2.7),no significant change in light scattering isobserved (there is no significant decrease inscattering, attributed to little fusion betweenthe vesicle suspension). On the contrary,when calcium is present in the buffer solu-tion prior to addition of the t-SNARE andv-SNARE vesicles, there is a marked drop inlight scattering, as a result of vesicle aggre-gation and fusion (Fig. 2.7). However, in thepresence of NSF-ATP in the assay buffercontaining calcium, a significant inhibition

Page 41: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

32 MOLECULAR MECHANISM OF SNARE-INDUCED MEMBRANE FUSION

A

B

C

Figure 2.5. Conductance and capacitance measurements of SNARE-reconstituted lipid bilayers. The EPC9-electro-physiological setup is shown (A). In the presence of 5 mM EGTA, t-SNARE-associated vesicles containing nystatinchannels at their membrane interact with v-SNARE-reconstituted lipid bilayer without fusing (B). Note no change inconductance or capacitance following exposure of SNARE-associated lipid vesicles to the bilayer. The vesicles fuse,however, when 3 mM KCl is applied, demonstrating fusion of docked vesicles and presence of an intact bilayer (B).The arrowhead indicates when the stirring is switched on to mix the addition. In the presence of 1 mM CaCl2, thet-SNARE-associated vesicles fuse with the v-SNARE-reconstituted bilayer as depicted in a consequent increase inconductance and capacitance. Since a large majority of docked vesicles have fused, addition of 3 mM KCl has nofurther effect (C). Traces B and C are representative profiles from one of five separate experiments.

Page 42: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

ACKNOWLEDGMENT 33

100 t-/v- SNARE Vesicles

t-/v- SNARE Vesicles + Ca2+

80

60

40

20

023 28 33

I (ar

bita

ry u

nits

)

38 43 48

4.28 3.5 3.0 2.62 2.33 2.1 Å

2 θ (°)

Figure 2.6. Wide-angle x-ray diffraction patterns of interacting SNARE-vesicles. Representative diffraction profilesfrom one of four separate experiments using t- and v-SNARE-reconstituted lipid vesicles, in the presence or absenceof 5 mM Ca2+, is shown. Arrows mark appearance of new peaks in the x-ray diffractogram, following addition ofcalcium.

in aggregation and fusion of proteoliposomesis observed (Fig. 2.7). NSF, in the absenceof ATP, has no effect on the light scatter-ing properties of the vesicle mixture. Theseresults demonstrate that NSF-ATP disassem-bles the SNARE complex, thereby reduc-ing the number of interacting vesicles insolution. In addition, disassembly of trans-SNARE complex will then leave apposedbilayers widely separated, out of reach forthe formation of Ca2+ –PO bridges, prevent-ing membrane fusion (Fig. 2.7). Similarly, ifthe restricted area between adjacent bilayersdelineated by the circular arrangement of thet-/v-SNARE complex (Cho et al., 2002) ispreformed, then hydrated Ca2+ ions are toolarge (Fig. 2.7) to be accommodated betweenbilayers and, hence, subsequent addition ofCa2+ would have no effect (Fig 2.7). How-ever, when t-SNARE vesicles interact with v-SNARE vesicles in the presence of Ca2+, thet-/v-SNARE complex formed allow forma-tion of calcium–phosphate bridges betweenopposing bilayers, leading to the expulsion

of water around the Ca2+ ion to enablelipid mixing and membrane fusion (Fig. 2.7).Thus, x-ray and light scattering studies(Jeremic et al., 2004b) demonstrate that cal-cium bridging of the apposing bilayers isrequired to enable membrane fusion. Thiscalcium bridging of apposing bilayers allowsfor the release of water from the hydratedCa2+ ion, leading to bilayers destabiliza-tion and membrane fusion. In addition, thebinding of calcium to the phosphate headgroups of the apposing bilayers may also dis-place the loosely coordinated water at the POgroups, further contributing to the destabi-lization of the lipid bilayer, leading to mem-brane fusion.

ACKNOWLEDGMENT

Supported by Grants DK-56212 and NS-39918 from the National Institutes of Health(BPJ).

Page 43: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

120

100 80 60 40 20 0

010

020

0tim

e (s

ec)

300

400

140

t-S

NA

RE

vesi

cle

v-S

NA

RE

vesi

cle

2.9Å

SN

AR

EC

ompl

ex

~ 6

.0Å

2.56

Å

0.9Å

Ca

OH

NS

FA

DPPi

AT

P

+ N

SF

OO

O

P

P

O

O

O

O

O

ME

MB

RA

NE

FU

SIO

N

AC D

B

I (a. u.)

t-/v

- S

NA

RE

Ves

icle

sC

a2+ +

t-/v

- S

NA

RE

Ves

icle

sC

a2+ +

NS

F +

AT

P +

t-/v

- S

NA

RE

Ves

icle

s

Fig

ure

2.7.

Lig

htsc

atte

ring

profi

les

ofSN

AR

E-a

ssoc

iate

dve

sicl

ein

tera

ctio

ns.(

A,B

)A

dditi

onof

t-SN

AR

Ean

dv-

SNA

RE

vesi

cles

inca

lciu

m-f

ree

buff

erle

adto

sign

ifica

ntin

crea

sein

light

scat

teri

ng.

Subs

eque

ntad

ditio

nof

5m

MC

a2+(m

arke

dby

arro

whe

ad)

does

not

have

any

sign

ifica

ntef

fect

onlig

htsc

atte

ring

( �).

(A,

C)

Inth

epr

esen

ceof

NSF

-AT

P(1

µg/

ml)

inas

say

buff

erco

ntai

ning

5m

MC

a2+,s

igni

fican

tlyin

hibi

ted

vesi

cle

aggr

egat

ion

and

fusi

on( �

).(A

,D)

Whe

nth

eas

say

buff

erw

assu

pple

men

ted

with

5m

MC

a2+,

prio

rto

addi

tion

oft-

and

v-SN

AR

Eve

sicl

es,

itle

dto

afo

ur-f

old

decr

ease

inlig

htsc

atte

ring

inte

nsity

due

toC

a2+-i

nduc

edag

greg

atio

nan

dfu

sion

oft-

/v-S

NA

RE

appo

sed

vesi

cles

(©).

Lig

htsc

atte

ring

profi

les

show

nar

ere

pres

enta

tives

offo

urse

para

teex

peri

men

ts.

34

Page 44: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 35

REFERENCES

Bako, I., Hutter, J., and Palinkas, G. (2002).Car–Parrinello molecular dynamics simulationof the hydrated calcium ion. J. Chem. Phys.117:9838–9843.

Chialvo, A. A., and Simonson, J. M. (2003). Thestructure of CaCl2 aqueous solutions over awide range of concentration. Interpretation ofdiffraction experiments via molecular simula-tion. J. Chem. Phys. 119:8052–8061.

Cho, S. J., Kelly, M., Rognlien, K. T., Cho, J.,Hoerber, J. K. H., and Jena, B. P. (2002).SNAREs in opposing bilayers interact in a cir-cular array to form conducting pores. Biophys.J. 83:2522–2527.

Hanson, P. I., Roth, R., Morisaki, H., Jahn, R.,and Heuser, J. E. (1997). Structure and con-formational changes in NSF and its mem-brane receptor complexes visualized by quick-freeze/deep-etch electron microscopy. Cell 90:523–535.

Jeong, E.-H., Webster, P., Khuong, C. Q., Sat-tar, A. K. M. A., Satchi, M., and Jena, B. P.(1998). The native membrane fusion machineryin cells. Cell Biol. Int. 22:657–670.

Jeremic, A., Kelly, M., Cho, W. J., Cho, S. J.,Horber, J. K. H., and Jena, B. P. (2004a). Cal-cium drives fusion of SNARE-apposed bilay-ers. Cell. Biol. Int. 28:19–31.

Jeremic, A., Cho, W. J., and Jena, B. P. (2004b).Membrane fusion: What may transpire atthe atomic level. J. Biol. Phys. Chem. 4:139–142.

Laroche, G., Dufourc, E. J., Dufoureq, J., andPezolet, M. (1991). Structure and dynamicsof dimyristoylphosphatidic acid/calcium com-plex by 2H NMR, infrared, spectroscopiesand small-angle x-ray diffraction. Biochemistry30:3105–3114.

Licheri, G., Piccaluga, G., and Pinna, G. (1976).X-ray diffraction study of the average solutespecies in CaCl2 aqueous solutions. J. Chem.Phys. 64:2437–2446.

McIntosh, T. J. (2000). Short-range interactionsbetween lipid bilayers measured by X-ray dif-fraction. Curr. Opin. Struct. Biol. 10: 481–485.

Portis, A., Newton, C., Pangborn, W., and Papa-hadjopoulos, D. (1979). Studies on the mech-anism of membrane fusion: Evidence for anintermembrane Ca2+ –phospholipid complex,synergism with Mg2+, and inhibition by spec-trin. Biochemistry 18:780–790.

Rothman, J. E. (1994). Mechanism of intra-cellular protein transport. Nature 372: 55–63.

Schwenk, C. F., Loeffler, H. H., and Rode, B. M.(2001). Molecular dynamics simulations ofCa2+ in water: Comparison of a classi-cal simulation including three-body correc-tions and Born–Oppenheimer ab initio anddensity functional theory quantum mechani-cal/molecular mechanics simulations. J. Chem.Phys. 115:10808–10813.

Sutton, R. B., Fasshauer, D., Jahn, R., and Brun-ger, A. T. (1998). Crystal structure of a SNAREcomplex involved in synaptic exocytosis at2.4 A resolution. Nature 395:347–353.

Weber, T., Zemelman, B. V., McNew, J. A.,Westerman, B., Gmachl, M., Parlati, F., Soll-ner, T. H. and Rothman, J. E. (1998). SNARE-pins: Minimal machinery for membrane fusion.Cell 92:759–772.

Wilschut, J., Duzgunes, N., Fraley, R., and Papa-hadjopoulos, D. (1980). Studies on the mech-anism of membrane fusion: Kinetics of cal-cium ion induced fusion of phosphatidylserinevesicles followed by a new assay for mix-ing of aqueous vesicle content. Biochemistry19:6011–6021.

Page 45: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

CHAPTER 3

MOLECULAR MECHANISM OF SECRETORYVESICLE CONTENT EXPULSION DURING CELLSECRETIONBHANU P. JENADepartment of Physiology, Wayne State University School of Medicine, Detroit, Michigan

3.1. INTRODUCTION

In the last decade, the molecular mechanismof vesicle swelling (Abu-Hamdah et al.,2004; Cho et al., 2002c; Jena et al., 1997)and its involvement in the regulated expul-sion of vesicular contents (Kelly et al.,2004) has been determined. Secretory vesi-cle swelling is critical for secretion (Alvarezde Toledo et al., 1993; Curran and Brod-wick, 1991; Monck et al., 1991; Sattar et al.,2002), however, the underlying mechanismof vesicle swelling was largely unknownuntil recently (Abu-Hamdah et al., 2004;Cho et al., 2002; Jena et al., 1997). In mastcells, an increase in secretory vesicle volumeafter stimulation of secretion has previ-ously been suggested from electrophysiolog-ical measurements (Fernandez et al., 1991).However, direct evidence of secretory vesi-cle swelling in live cells was first demon-strated in pancreatic acinar cells using theAFM (Fig. 3.1) (Cho et al., 2002a). Isolatedzymogen granules (ZGs), the membrane-bound secretory vesicles in exocrine pan-creas and parotid glands, possess Cl−- andATP-sensitive, K+-selective ion channels atthe vesicle membrane, whose activities have

been implicated in vesicle swelling (Jenaet al., 1997). Additionally, secretion of ZGcontents in permeabilized pancreatic aci-nar cells requires the presence of both K+and Cl− ions. In vitro ZG-pancreatic plasmamembrane fusion assays further demonstratepotentiation of fusion in the presence of GTP(Sattar et al., 2002). Gαi protein has beenimplicated in the regulation of both K+ andCl− ion channels in a number of tissues.Analogous to the regulation of K+ and Cl−ion channels at the cell plasma membrane,their regulation at the ZG membrane by aGαi3 protein was demonstrated (Jena et al.,1997). Isolated ZGs from exocrine pancreasswell rapidly in response to GTP (Fig. 3.2)(Jena et al., 1997). These studies suggestedthe involvement of rapid water entry intoZGs following exposure to GTP. There-fore, when the possible involvement of waterchannels or aquaporins in ZG swelling wasexplored (Cho et al., 2002c), results from thestudy demonstrate the presence of aquaporin-1 (AQP1) at the ZG membranes and its par-ticipation in GTP-mediated water entry andswelling (Fig. 3.3) (Cho et al., 2002c). Tofurther understand the molecular mechanism

Force Microscopy: Applications in Biology and Medicine, edited by Bhanu P. Jena and J.K. Heinrich Horber.Copyright 2006 John Wiley & Sons, Inc.

37

Page 46: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

38 MOLECULAR MECHANISM OF SECRETORY VESICLE CONTENT EXPULSION DURING CELL SECRETION

Figure 3.1. AFM micrograph or live pancreatic acinar cell demonstrating the size of ZGs within the cell, in resting(A) and following stimulation of secretion (B). Note the increase in size of the same granules immediately followinga secretory stimuli (section analysis of two such ZGs are shown). There is no loss of secretory vesicles followingcompletion of secretion.

of secretory vesicle swelling, the regulationof AQP1 in the ZG was determined (Abu-Hamdah et al., 2004). Detergent-solubilizedZGs immunoprecipitated with monoclonalAQP-1 antibody co-isolates AQP-1, PLA2,Gαi3, potassium channel IRK-8, and thechloride channel ClC-2 (Abu-Hamdah et al.,2004). Exposure of ZGs to either thepotassium channel blocker glyburide orthe PLA2 inhibitor ONO-RS-082 blockedGTP-induced ZG swelling. RBC, known topossess AQP-1 at the plasma membrane,also swell on exposure to the Gαi-agonistmastoparan and respond similarly to ONO-RS-082 and glyburide, as do ZGs. Addition-ally, liposomes reconstituted with the AQP-1immunoisolated complex from solubilizedZGs, also swell in response to GTP. Gly-buride or ONO-RS-082 abolished the GTPeffect in reconstituted liposomes. Further-more, immunoisolate-reconstituted planarlipid membrane demonstrate conductance,which is sensitive to glyburide and toan AQP-1 specific antibody. These resultsdemonstrate a Gαi3-PLA2-mediated pathway

and potassium channel involvement inAQP-1 regulation (Fig. 3.4) (Abu-Hamdahet al., 2004), contributing to our understand-ing of the molecular mechanism of ZGswelling. Studies in both slow and fast secre-tory cells (neurons) further reveal that secre-tory vesicle swelling is a requirement for theregulated expulsion of intravesicular contentsduring cell secretion (Kelly et al., 2004).

3.2. VESICLE SWELLING IN A SLOWSECRETORY CELL (PANCREATICACINAR) IS REQUIRED ININTRAVESICULAR CONTENTEXPULSION DURING SECRETION

Pancreatic acinar cells (Fig. 3.5A) have beenused in studies that demonstrate that vesicleswelling is a requirement for the expulsionof intravesicular contents during cell secre-tion (Kelly et al., 2004). Isolated live pancre-atic acinar cells in near-physiological bufferimaged using the atomic force microscope(AFM) at higher force (200–300 pN) is able

Page 47: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

VESICLE SWELLING IN A SLOW SECRETORY CELL (PANCREATIC ACINAR) 39

Figure 3.2. Increase in size of isolated ZGs in the presence of GTP. (A–C) Two-dimensional AFM images ofzymogen granules (ZG), the membrane-bound secretory vesicles of the exocrine pancreas, after exposure to 20 µMGTP at time 0 min (A), 5 min (B), and 10 min (C). (D–F) The same granules are shown in three dimensions at 0,5, and 10 min, respectively, following exposure to GTP. (G–I) The GTP-induced increase in size of another groupof ZGs observed by confocal microscopy is shown at time 0, 5, and 10 min following GTP addition. (Bar represents1 µm.) Values represent one of three representative experiments.

to reveal the structure and dynamics of ZGs,the membrane-bound secretory vesicles ofthe exocrine pancreas, lying immediatelybelow the apical plasma membrane (PM)of the cell (Fig. 3.5B). Within 2.5 min ofexposure to a physiological secretory stim-ulus (1 µM carbamylcholine), the majorityof ZGs within cells were found to swell(Fig. 3.5C), followed by a decrease in ZGsize (Fig. 3.5D) by which time most of therelease of secretory products from withinZGs had occurred (Fig. 3.5E). These studies(Kelly et al., 2004) reveal, for the first time inlive cells, intracellular swelling of secretoryvesicles following stimulation of secretion

and their deflation following partial dischargeof vesicular contents. Measurements of intra-cellular ZG size further reveal that differentvesicles swell differently, following a secre-tory stimulus. For example, the ZG markedby the red arrowhead swelled to show a23–25% increase in diameter, in contrastto the green arrowhead-marked ZG whichincreased by only 10–11% (Fig. 3.5B,C).This differential swelling among ZGs withinthe same cell may explain why follow-ing stimulation of secretion, some intracel-lular ZGs demonstrate the presence of lessvesicular content than others, and hence have

Page 48: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

40 MOLECULAR MECHANISM OF SECRETORY VESICLE CONTENT EXPULSION DURING CELL SECRETION

Figure 3.3. AQP1-specific antibody binds to the ZG membrane and blocks water entry. (A) Immunoblot assaydemonstrating the presence of AQP1 antibody in SLO-permeabilized ZG. Lanes: 1, AQP1 antibody alone; 2,nonpermeable ZG exposed to antibody; 3, permeable ZG exposed to AQP1 antibody. Immunoelectron micrographsof intact ZGs exposed to AQP1 antibody demonstrate little labeling (B, C). (Bar = 200 nm.) Contrarily, SLO-treatedZG demonstrate intense gold labeling at the luminal side of the ZG membrane (D, E). AQP1 regulates GTP-inducedwater entry in ZG. (F) Schematic diagram of ZG membrane depicting AQP1-specific antibody binding to the carboxyldomain of AQP1 at the intragranular side to block water gating. (G,H, K) AQP1 antibody introduced into ZG blocksGTP-induced water entry and swelling (from G to H, after GTP exposure). (I–K) However, only vehicle introducedinto ZG retains the stimulatory effect of GTP (from I to J, after GTP exposure).

discharged more of their contents (Cho et al.,2002a; Kelly et al., 2004).

To determine precisely the role of swellingin vesicle–plasma membrane fusion and inthe expulsion of intravesicular contents, anelectrophysiological ZG-reconstituted lipidbilayer fusion assay (Cho et al., 2002b;Jeremic et al., 2003) has been employed.

Figure 3.4. Gαi-PLA2-mediated pathway and potas-sium involvement in AQP-1 regulation.

Page 49: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

VESICLE SWELLING IN A SLOW SECRETORY CELL (PANCREATIC ACINAR) 41

Figure 3.5. The swelling dynamics of ZGs in live pancreatic acinar cells. (A) Electron micrograph of pancreaticacinar cells showing the basolaterally located nucleus (N) and the apically located ZGs. The apical end of the cellfaces the acinar lumen (L). Bar = 2.5 µm. (B–D) The apical ends of live pancreatic acinar cells were imaged byAFM, showing ZGs (grey and white arrowheads) lying just below the apical plasma membrane. Exposure of the cellto a secretory stimulus using 1 µM carbamylcholine resulted in ZG swelling within 2.5 min, followed by a decreasein ZG size after 5 min. The decrease in size of ZGs after 5 min is due to the release of secretory products such asα-amylase, as demonstrated by the immunoblot assay (E).

The ZGs used in the bilayer fusion assayswere characterized for their purity and theirability to respond to a swelling stimulus. ZGswere isolated (Jena et al., 1997) and theirpurity assessed using electron microscopy(Fig. 3.6A). As previously reported (Abu-Hamdah et al., 2004; Cho et al., 2002c;Jena et al., 1997), exposure of isolated ZGs(Fig. 3.6B) to GTP resulted in ZG swelling(Fig. 3.6C). Once again, similar to whatis observed in live acinar cells (Fig. 3.5),each isolated ZG responded differently to thesame swelling stimulus (Kelly et al., 2004).For example, the red arrowhead points toa ZG whose diameter increased by 29% asopposed to the green arrowhead pointing ZGthat increased only by a modest 8%. The dif-ferential response of isolated ZGs to GTPwas further assessed by measuring changesin the volume of isolated ZGs of differ-ent sizes (Fig. 3.6D). ZGs in the exocrine

pancreas range in size from 0.2 to 1.3 µmin diameter (Jena et al., 1997). Not all ZGswere found to swell following a GTP chal-lenge (Jena et al., 1997; Kelly et al., 2004).Most ZG volume increases were between5% and 20%; however, larger increases upto 45% were observed only in ZGs rangingfrom 250 to 750 nm in diameter (Fig. 3.6D)(Kelly et al., 2004). These studies (Kellyet al., 2004) demonstrate that following stim-ulation of secretion, ZGs within pancreaticacinar cells swell, followed by (i) a releaseof intravesicular contents through porosomes(12) at the cell plasma membrane and (ii)a return to resting size on completion ofsecretion. On the contrary, isolated ZGs stayswollen following exposure to GTP, sincethere is no outlet for the release of theintravesicular contents. In acinar cells, lit-tle or no secretion is detected 2.5 min fol-lowing stimulation of secretion, although the

Page 50: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

42 MOLECULAR MECHANISM OF SECRETORY VESICLE CONTENT EXPULSION DURING CELL SECRETION

Figure 3.6. Swelling of isolated ZGs. (A) Electron micrograph of isolated ZGs demonstrating a homogeneouspreparation. Bar = 2.5 µm. (B, C) Isolated ZGs, on exposure to 20 µM GTP, swell rapidly. Note the enlargementof ZGs as determined by AFM section analysis of two vesicles (grey and white arrowheads). (D) Percent ZG volumeincrease in response to 20 µM GTP. Note how different ZGs respond to the GTP-induced swelling differently.

ZGs within them were completely swollen(Fig. 3.5C). However, at 5 min followingstimulation, ZGs deflate and the intravesic-ular α-amylase released from the acinar cellcould be detected, suggesting the involve-ment of ZG swelling in cell secretion (Kellyet al., 2004).

In electrophysiological bilayer fusionassays, immunoisolated fusion pores orporosomes from the exocrine pancreas,functionally reconstituted into the lipidmembrane of the bilayer apparatus, havebeen used in our studies (Jeremic et al.,2003). In these experiments, membraneconductance and capacitance is continuallymonitored (Fig. 3.7A). Reconstitution ofthe porosome into the lipid membraneresulted in a small increase in capacitance(Fig. 3.7B), possibly due to increase inmembrane surface area contributed by theincorporation of porosomes, ranging insize from 100 to 150 nm in diameter(Jeremic et al., 2003). Isolated ZGs whenadded to the cis compartment of sucha porosome-reconstituted bilayer chamber,

fuse at the base of porosomes in the lipidmembrane (Fig. 3.7A) and is detected asa step increase in membrane capacitance(Fig. 3.7B). However, even after 15 minof ZG addition to the cis compartment ofthe bilayer chamber, little or no release ofthe intravesicular enzyme α-amylase wasdetected in the trans compartment of thechamber (Fig. 3.7C, D). On the contrary,exposure of ZGs to 20 µM GTP inducedswelling (Abu-Hamdah et al., 2004; Choet al., 2002c; Jena et al., 1997), and itresults in both (a) the potentiation offusion and (b) a robust expulsion of α-amylase into the trans compartment ofthe bilayer chamber (Fig. 3.7C, D). Thesestudies demonstrated that during secretion,secretory vesicle swelling is required for theefficient expulsion of intravesicular contents(Kelly et al., 2004).

Within minutes or even seconds followingstimulation of secretion, empty and partiallyempty secretory vesicles accumulate withincells (Cho et al., 2002b; Lawson et al., 1975;Plattner et al., 1997). There may be two pos

Page 51: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

VESICLE SWELLING IN A SLOW SECRETORY CELL (PANCREATIC ACINAR) 43

Figure 3.7. Fusion of isolated ZGs at porosome-reconstituted bilayer and GTP-induced expulsion of α-amylase. (A)Schematic diagram of the EPC9 bilayer apparatus showing the cis and trans chambers. Isolated ZGs, when added tothe cis chamber, fuse at the bilayers-reconstituted porosome. Addition of GTP to the cis chamber induces ZG swellingand expulsion of its contents such as α-amylase to the trans bilayers chamber. (B) Capacitance traces of the lipidbilayer from three separate experiments following reconstitution of porosomes, following addition of ZGs to the cischamber, and at the 5-min time point after ZG addition. Note the small increase in membrane capacitance followingporosome reconstitution, along with a greater increase when ZGs fuse at the bilayers. (C) In a separate experiment,15 min after addition of ZGs to the cis chamber, 20 µM GTP was introduced. Note the increase in capacitance,demonstrating potentiation of ZG fusion. Flickers in current trace represent current activity. (D) Immunoblot analysisof α-amylase in the trans chamber fluid at different times following exposure to ZGs and GTP. Note the undetectablelevels of α-amylase even up to 15 min following ZG fusion at the bilayer. However, following exposure to GTP,significant amounts of α-amylase from within ZGs were expelled into the trans bilayers chamber.

Page 52: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

44 MOLECULAR MECHANISM OF SECRETORY VESICLE CONTENT EXPULSION DURING CELL SECRETION

sible explanations for such accumulation ofpartially empty vesicles. Following fusion atthe porosome, secretory vesicles may either(a) remain fused for a brief period and there-fore time would be the limiting factor forpartial release of intravesicular contents, oralternately, inadequate vesicle swelling wouldbe unable to generate the required intravesic-ular pressure for complete discharge of con-tents. Results in Fig. 3.5 suggests that it wouldbe highly unlikely that generation of partiallyempty vesicles result from brief periods ofvesicle fusion at porosomes. After additionof ZGs to the cis chamber of the bilayerapparatus, membrane capacitance continuesto increase (demonstrating the establishmentof continuity between the opposing bilay-ers), however, little or no detectable secretionoccurred even after 15 min (Fig. 3.5), sug-gesting that either variable degrees of vesicleswelling or repetitive cycles of fusion andswelling of the same vesicle, or both, mayoperate during cell secretion. Under these cir-cumstances, empty and partially empty vesi-cles could be generated within cells follow-ing secretion. To test this hypothesis, two keyparameters have been examined (Kelly et al.,2004): (i) whether the extent of swelling issame for all ZGs exposed to a certain concen-tration of GTP and (ii) whether ZG is capableof swelling to different degrees. And, if so,whether there is a correlation between extentof swelling and the quantity of intravesicu-lar contents expelled. The answer to the firstquestion is clear: Different ZGs respond tothe same stimulus differently (Fig. 3.5) (Kellyet al., 2004). As previously discussed, stud-ies (Kelly et al., 2004) reveal that differentZGs within cells or in isolation undergo dif-ferent degrees of swelling, even though theyare exposed to the same stimuli (carbamyl-choline for live pancreatic acinar cells) or GTPfor isolated ZGs (Figs. 3.5B–D, 3.6B–D).The requirement of ZG swelling for expulsionof vesicular contents has been further con-firmed, since GTP dose-dependently increasesin ZG swelling (Fig. 3.8A–C) translated into

increased secretion of the intravesicular α-amylase enzyme (Fig. 3.8D). Although higherGTP concentrations elicit an increased ZGswelling, the extent of swelling between ZGsonce again is varies (Kelly et al., 2004).

3.3. SYNAPTIC VESICLE SWELLINGIS ALSO REQUIRED FORINTRAVESICULAR CONTENTEXPULSION DURING SECRETIONIN NEURONS

To determine if a similar or an alternatemechanism is responsible for the releaseof secretory products in a fast secretorycell, synaptosomes and synaptic vesiclesfrom rat brain was used in our studies(Kelly et al., 2004). Since synaptic vesi-cle membrane is known to possess bothGi and Go proteins, it was hypothesizedthat GTP and the Gi-agonist (mastoparan)may mediate vesicle swelling (Kelly et al.,2004). To test this hypothesis, isolated synap-tosomes (Fig. 3.9A) were lysed to obtainsynaptic vesicles and synaptosomal mem-brane (Kelly et al., 2004). Isolated synapto-somal membrane, when placed on mica andimaged by the AFM in near-physiologicalbuffer, reveal on the cytosolic compartment40–50 nm in diameter synaptic vesicles stilldocked to the presynaptic membrane. Sim-ilar to ZGs, exposure of synaptic vesi-cles (Fig. 3.9B) to 20 µM GTP (Fig. 3.9C)resulted in an increase in synaptic vesi-cle swelling. However, exposure to Ca2+resulted in the transient fusion of synap-tic vesicles at the presynaptic membrane,expulsion of intravesicular contents, and theconsequent decrease in size of the synapticvesicle (Fig. 3.9D,E). In Fig. 3.9B–D, theblue arrowhead points to a synaptic vesi-cle undergoing this process. Additionally,as observed in ZGs of the exocrine pan-creas, not all synaptic vesicles were found toswell; if they did, they swelled to differentextents even though they had been exposedto the same stimulus (Kelly et al., 2004).

Page 53: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

ACKNOWLEDGMENT 45

Figure 3.8. The extent of ZG swelling is directly proportional to the amount of intravesicular contents released.(A) AFM micrographs showing the GTP dose-dependent increase in swelling of isolated ZGs. (B) Note the AFMsection analysis of a single ZG (grey arrowhead), showing the height and relative width at resting (control, greyoutline), following exposure to 5 µM GTP (middle white outline) and 10 µM GTP (outer white outline). (C) Graphdemonstrating the GTP dose-dependent percent increase in ZG volume. Data are expressed as mean ± SEM. (D)Immunoblot analysis of α-amylase in the trans chamber fluid of the bilayers chamber following exposure to differentdoses of GTP. Note the GTP dose-dependent increase in α-amylase release from within ZGs fused at the cis side ofthe reconstituted bilayer.

This differential response of synaptic vesicleswithin the same nerve ending may dictateand regulate the potency and efficacy of neu-rotransmitter release at the nerve terminal.To further confirm synaptic vesicle swellingand determine swelling rate, light scatter-ing experiments have been performed. Lightscattering studies demonstrate a mastoparan-dose-dependent increase in synaptic vesicleswelling (Fig. 7.9F). Mastoparan (20 µM)induces a time-dependent (in seconds)increase in synaptic vesicle swelling(Fig. 7.9G), as opposed to the control peptide(Mast-17) (Kelly et al., 2004).

These studies demonstrate that followingstimulation of secretion, ZGs (the membrane-bound secretory vesicles in exocrine pan-creas) swell. Different ZGs swell differ-ently, and the extent of their swelling dic-tates the amount of intravesicular contentsto be expelled. ZG swelling is therefore a

requirement for the expulsion of vesicularcontents in the exocrine pancreas. Similar toZGs, synaptic vesicles also swell, enablingthe expulsion of neurotransmitters at thenerve terminal. This mechanism of vesicularexpulsion during cell secretion may explainwhy partially empty vesicles are generatedin secretory cells (Cho et al., 2002b; Lawsonet al., 1975; Plattner et al., 1997) followingsecretion. The presence of empty secretoryvesicles could result from multiple rounds offusion–swelling–expulsion, which a vesiclemay undergo during the secretory process.These results reflect the precise and regulatednature of cell secretion, from the exocrinepancreas to neurons.

ACKNOWLEDGMENT

This work was supported by NIH grantsDK56212 and NS39918 (B.P.J).

Page 54: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

46 MOLECULAR MECHANISM OF SECRETORY VESICLE CONTENT EXPULSION DURING CELL SECRETION

Figure 3.9. Synaptic vesicles swell in response to GTP and mastoparan, and vesicle swelling is required forneurotransmitter release. (A) Electron micrographs of brain synaptosomes, demonstrating the presence of 40- to50-nm synaptic vesicles within. Bar = 200 nm. (B) AFM micrographs of synaptosomal membrane, demonstrating thepresence of 40- to 50-nm synaptic vesicles docked to the cytosolic face of the presynaptic membrane. (C) Exposureof the synaptic vesicles to 20 µM GTP results in vesicle swelling (arrowhead). (D, E) Further addition of calciumresults in the transient fusion of the synaptic vesicles at porosomes in the presynaptic membrane of the nerve terminal,along with expulsion of intravesicular contents. Note the decrease in size of the synaptic vesicle following contentexpulsion. (F) Light scattering assays on isolated synaptic vesicles demonstrate the mastoparan dose-dependentincrease in vesicle swelling (n = 5), and they further confirm the AFM results. (G) Exposure of isolated synapticvesicles to 20 µM mastoparan demonstrates a time-dependent (in seconds) increase in their swelling. Note that thecontrol peptide mast-17 has little or no effect on synaptic vesicles.

REFERENCES

Abu-Hamdah, R., Cho, W.-J., Cho, S.-J., Jeremic,A., Kelly, M., Ilie, A. E., and Jena, B. P.

(2004). Regulation of the water channel aqua-porin-1: Isolation and reconstitution of theregulatory complex. Cell Biol. Int. 28:7–17(published on-line 2003).

Alvarez de Toledo, G., Fernandez-Chacon, R.,and Fernandez, J. M. (1993). Release of secre-tory products during transient vesicle fusion.Nature (London) 363:554–558.

Cho, S.-J., Cho, J., and Jena, B. P. (2002a).The number of secretory vesicles remainsunchanged following exocytosis. Cell Biol. Int.26:29–33.

Cho, S. J., Kelly, M., Rognlien, K. T., Cho, J.,Hoerber, J. K. H., and Jena, B. P. (2002b).SNAREs in opposing bilayers interact in acircular array to form conducting pores. Bio-phys. J. 83:2522–2527.

Cho, S. J., Sattar, A. K., Jeong, E. H., Satchi, M.,Cho, J. A., Dash, S., Mayes, M. S., Stromer,

M. H., and Jena, B. P. (2002c). Aquaporin 1regulates GTP-induced rapid gating of water insecretory vesicles. Proc. Natl. Acad. Sci. USA99:4720–4724.

Curran, M. J., and Brodwick, M. S. (1991). Ioniccontrol of the size of the vesicle matrix of beigemouse mast cells. J. Gen. Physiol. 98:771–790.

Fernandez, J. M., Villalon, M., and Verdugo, P.(1991). Reversible condensation of the mastcell secretory products in vitro. Biophys. J.59:1022–1027.

Jena, B. P., Schneider, S. W., Geibel, J. P., Web-ster, P., Oberleithner, H., and Sritharan, K. C.(1997). Gi regulation of secretory vesicleswelling examined by atomic force microscopy.Proc. Natl. Acad. Sci. USA 94:13317–13322.

Jeremic, A., Kelly, M., Cho, S. J., Stromer,M. H., and Jena, B. P. Reconstituted fusionpore. Biophys. J. 85:2035–2043.

Kelly, M., Cho, W. J., Jeremic, A., Abu-Hamdah,R., and Jena, B. P. (2004). Vesicle swelling

regulates content expulsion during secretion.Cell Biol. Int. 28:709–716.

Page 55: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 47

Lawson, D., Fewtrell, C., Gomperts, B., and Raff,M. C. (1975). Anti-immunoglobulin-induced

histamine secretion by rat peritoneal mast cellsstudied by immunoferritin electron microscopy.J. Exp. Med. 142:391–401.

Monck, J. R., Oberhauser, A. F., Alvarez deToledo, G., and Fernandez, J. M. (1991). Isswelling of the secretory granule matrix theforce that dilates the exocytotic fusion pore?Biophys. J. 59:39–47.

Plattner, H., Artalejo, A. R., and Neher, E. (1997).Ultrastructural organization of bovine chromaf-fin cell cortex—analysis by cryofixation andmorphometry of aspects pertinent to exocytosis.J. Cell Biol. 139:1709–1717.

Sattar, A. K. M., Boinpally, R., Stromer, M. H.,and Jena, B. P. (2002) Gαi3 in pancreatic zymo-gen granule participates in vesicular fusion. J.Biochem. 131:815–820.

Page 56: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

CHAPTER 4

FUSION PORES INGROWTH-HORMONE-SECRETING CELLS OFTHE PITUITARY GLAND: AN AFM STUDYLLOYD L. ANDERSONDepartment of Animal Science, College of Agriculture, and Department of Biomedical Sciences,College of Veterinary Medicine, Iowa State University, Ames, Iowa

BHANU P. JENADepartment of Physiology, Wayne State University School of Medicine, Detroit, Michigan

4.1. INTRODUCTION

This will briefly review the physiologyof growth hormone (GH) secretion in vivofocused on an animal model, the pig. Thereis increasing knowledge of the subcellu-lar events leading to GH release. Thisincludes signal transduction mechanismsand the physical nature of GH secretorygranules fusing transiently with the cellmembrane at the nanoscale. Hypothalamichormones that influence GH release andsynthesis with particular attention to otherpeptides as well as GH-releasing hormone(GHRH), somatostatin (SRIF), and the natu-ral GH-secretagogue, ghrelin, will be consid-ered. The immunohistochemical distributionand differential immunoreactivity of soma-totrophs in porcine anterior pituitary acrossages may reflect changes in the numberof GH vesicles per cell or heterogeneityof somatotrophs. Finally, a major focus onthe cell biology for controlled GH secretion

involving movement of secretory granulesand fusion pores and secretory pits of soma-totrophs utilizing atomic force microscopy(AFM) and transmission electron microscopy(TEM) will be examined.

4.2. NEUROENDOCRINEREGULATION OF GH SECRETIONIN VIVO

A series of stimulatory and inhibitory releas-ing hormones of hypothalamic and peripheralorigins controls the release of growth hormone(GH) from somatotrophs. Until recently, theconsensus was of two hypothalamic releas-ing hormones for GH, respectively GH-releasing hormone (GHRH; stimulatory GH)and somatostatin (SRIF; inhibitory) with GHrelease and synthesis reflecting a balancebetween these.

In vivo approaches such as stalk section-ing and hypothalamic deafferentation provide

Force Microscopy: Applications in Biology and Medicine, edited by Bhanu P. Jena and J.K. Heinrich Horber.Copyright 2006 John Wiley & Sons, Inc.

49

Page 57: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

50 FUSION PORES IN GROWTH-HORMONE-SECRETING CELLS OF THE PITUITARY GLAND: AN AFM STUDY

A

B

Figure 4.1. Adult pig and its pituitary gland. (A) Por-cine pituitary gland (∼250 mg), resting on a dime,is located at the base of the brain and connected tothe hypothalamus by an hypophyseal stalk. (B) AdultHampshire male pig (∼120-kg body weight).

strong evidence for the physiological controlof GH release. The pituitary gland in theadult pig weighs about 250 mg (Fig. 4.1).The anterior lobe of the gland containssomatotrophs that release GH into periph-eral blood in a pulsatile pattern (Fig. 4.2).After hypophyseal stalk transection (HST),the pulsatile GH secretion pattern is oblit-erated (Klindt et al., 1983). Control of GHsecretion was investigated in pigs by compar-ing the effects of anterior (AHD), complete(CHD), and posterior (PHD) hypothalamicdeafferentation with sham-operated controls(SOC) (Molina et al., 1986). A camera lucidadrawing of the sagittal view of the porcinethalamus and hypothalamus with a depic-tion of the hypothalamic areas isolated bythe knife is shown in Fig. 4.3. Mean serumconcentrations of GH after AHD, CHD,and PHD were reduced (P < 0.01) when

compared with SOC gilts (Fig. 4.4). Fur-thermore, episodic GH release evident inSOC animals was obliterated after hypotha-lamic deafferentation. Thus, maintenance ofepisodic GH secretion depends upon its neu-ral connections traversing the anterior andposterior aspects of the hypothalamus inthe pig.

There is at least one other hypothalamicreleasing hormone for GH, with the identifi-cation by Kojima and colleagues of ghrelinas the natural ligand for the GH secretagogue(GHS) receptor (GHS-R; Kojima et al.,1999). GH secretion is influenced by pep-tides/proteins produced in the hypothalamusand in the periphery, including gonadotropin-releasing hormone (GnRH), insulin-likegrowth factor 1 (IGF-1), leptin, pituitaryadenylate cyclase activating polypeptide(PACAP), and thyrotropin-releasing hor-mone (TRH). There is increasing knowl-edge of the subcellular events leading toGH release. This includes signal transductionmechanisms and the physical nature of GHsecretory granules fusing transiently with thecell membrane.

4.3. HYPOTHALAMIC HORMONESCONTROLLING GH RELEASE ANDSYNTHESIS

4.3.1. Growth-Hormone-ReleasingFactor (GHRH)

The major function of GHRH, released fromneurosecretory terminals in the median emi-nence, is to stimulate the release and syn-thesis of GH. GHRH is expressed in thearcuate nucleus of the hypothalamus togetherwith other tissues—for example, intestine,gonads, immune tissues, and the placenta.GHRH-immunoreactive (-IR) neurons havebeen identified in coronal and sagittal frozensections of bovine and porcine hypotha-lami (Leshin et al., 1994). Rounded bipolarGHRH-IR perikarya are localized in ventro-lateral regions of the arcuate nucleus (ARC) inboth species. GHRH-immunoreactive fibers

Page 58: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

HYPOTHALAMIC HORMONES CONTROLLING GH RELEASE AND SYNTHESIS 51

2310−UC

9002−UC

2396− SOC

2383−SOC

2371−HST

2211−HST

2

0

4

2

0

4

2

0

4

2

0

0

0

+3

DAYS AFTER HST

ng p

GH

/ml S

ER

UM

+4 +5 +6 +7 +8

1

1

Figure 4.2. Sequential profiles of peripheral serum concentrations of GH in two gilts from each treatment group(UC, unoperated control: SOC, sham operated control: HST, hypophyseal stalk transection) during a 120-h periodfrom Day +3 to Day +8. HST or SOC was performed on Day 0. Four-digit numbers designate individual gilts.Symbols indicate unoperated control (•), sham-operated control (◦), and hypophyseal (�) animals. (Reprinted fromProceedings of the Society for Experimental Biology and Medicine, Vol. 172, Klindt, J., Ford, J. J., Beradinelli, G.,and Anderson, L. L. Growth hormone secretion after hypophyseal stalk transection in pigs, pp. 503–513. Copyright1983, with permission from the Society for Experimental Biology and Medicine, Maywood, NJ.)

projected ventrally into the median eminence(ME) in both species.

Signal transduction involves Gs-protein,adenylate cyclase (isoform II and/or IV), cy-clic 3′, 5′-adenosine monophosphate (cAMP),

and protein kinase A. There is a cAMPresponse element (CRE) upstream from thecoding region of the GH gene. GHRHincreases GH expression (e.g., cattle, Silver-man et al., 1988; chickens, Vasilatos-Younken

Page 59: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

52 FUSION PORES IN GROWTH-HORMONE-SECRETING CELLS OF THE PITUITARY GLAND: AN AFM STUDY

MI

F

PV

DM

VM

MT

MBP

AN

PS

OC

Figure 4.3. A camera lucida drawing of the sagittal view of the porcine thalamus and hypothalamus with a depictionof the areas isolated by the knife. Interrupted lines define the arc for position of anterior and posterior knife cuts. Themammillary bodies (MB), mammillothalamic tract (MT), fornix (F), dorsomedial nucleus (DM), ventromedial nucleus(VM), arcuate nucleus (AN), posterior nucleus (P), optic chiasm (OC), messa intermedia (MI), and pituitary stalk (PS)are indicated. ×2.7. (Reprinted from Proceedings of the Society for Experimental Biology and Medicine, Vol. 183,Molina, J. R., Klindt, J., Ford, J. J., and Anderson, L. L. Growth hormone and prolactin secretion after hypothalamicdeafferentation in pigs, pp. 163–168. Copyright 1986, with permission from the Society for Experimental Biologyand Medicine, Maywood, NJ.)

et al., 1992; Radecki et al., 1994). Intracel-lular Ca2+ concentrations are increased byGHRH. This involves both influx of Ca2+(via L- and T-type voltage-sensitive Ca2+channels) and phospholipase C hydrolysis ofphosphatidyl inositol, leading to mobilizingintracellular Ca2+ stores.

4.3.2. Somatostatin (SRIF)

Somatostatin (SRIF) is a cyclic 14- or28-amino-acid residue containing peptide.A major function of SRIF is suppress-ing the release, but not synthesis, of GHby the somatotroph (reviewed in Tannen-baum and Epelbaum, 1999). SRIF-IR neu-rons have been identified in bovine andporcine hypothalamic tissue (Leshin et al.,1994). Bipolar SRIF-IR perikarya are locatedabout the third ventricle in the periventricular

nucleus. SRIF-IR fibers project ventrally intothe ME. SRIF-IR fibers densely innervate theventromedial and arcuate (ARC) nuclei inpigs, but are not as distinguishable as in cattle(Leshin et al., 1994). Double immunostain-ing reveals a close apposition of SRIF-IRfibers with GHRH-IR perikarya in the ARCand ventromedial nuclei and apposition ofSRIF- and GHRH-IR varicosities in the ME(Leshin et al., 1994).

The SRIF receptor (sstr) is encoded byfive different genes (sstr1-5; reviewed in Tan-nenbaum and Epelbaum, 1999). The dom-inant sstr influencing GH release from thesomatotroph appears to be sstr-2 (Reedet al., 1999). Signal transduction involves G-protein-coupled reduction in L- and T-typevoltage-sensitive Ca2+ influx/channels andincreased K+ channels (reviewed in Tannen-baum and Epelbaum, 1999).

Page 60: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

HYPOTHALAMIC HORMONES CONTROLLING GH RELEASE AND SYNTHESIS 53

−60 −20 0 0 040 40 4080 80120 12060

0 1 2DAYS FROM SURGERY

TIME IN MINUTES

16

12

8

4

0

16

12

8

4

0

16

12

8

4

0

16

12

8

4

0

AHDANESTHESIA

ANDSURGERY

CHD

PHD

SOC

GR

OW

TH

HO

RM

ON

E IN

PE

RIP

HE

RA

L S

ER

UM

, ng/

ml

Figure 4.4. GH concentrations in peripheral serum in ovariectomized prepuberal gilts during anesthesia and earlyrecovery (Day 0) and 24 and 48 h after hypothalamic deafferentation (Day 1 and 2, respectively). Groups consistedof 4 AHD, 5 CHD, 4 PHD, and 4 SOC gilts. Values are expressed as means ± SEM. (Reprinted from Proceedingsof the Society for Experimental Biology and Medicine, Vol. 183, Molina, J. R., Klindt, J., Ford, J. J., and Anderson,L. L. Growth hormone and prolactin secretion after hypothalamic deafferentation in pigs, pp. 163–168. Copyright1986, with permission from the Society for Experimental Biology and Medicine, Maywood, NJ.)

4.3.3. Ghrelin

Ghrelin is a natural ligand for the GH-secretagogue receptor (GHS-R; Lee et al.,2002). It is produced by the stomach, smallintestine, and central nervous system—forexample, the hypothalamus, specificallyincluding the ARC (Kojima et al., 1999;Lee et al., 2002; Masuda et al., 2000).

Ghrelin acts by binding to the GHS receptor.The GHS-R is a seven-transmembranedomain G-protein-coupled receptor (Howardet al., 1996). Signal transduction involvesactivation of phospholipase C (via G pro-tein), generation of inositol phosphate anddiacyl glycerol, and increased intracellularCa2+ (reviewed in Bowers, 1999). Ghrelin

Page 61: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

54 FUSION PORES IN GROWTH-HORMONE-SECRETING CELLS OF THE PITUITARY GLAND: AN AFM STUDY

stimulates GH release both directly (acting atthe level of the anterior pituitary gland) andby enhancing GHRH release. The GHS-Ris expressed in various hypothalamic andthalamic nuclei, the dentate gyrus, substantialnigra, ventral tegmentum, and facial nucleiof the brainstem, thus implicating a possiblecentral role for ghrelin (Bennet et al., 1997;Guan et al., 1997; Yokote et al., 1998;Mitchell et al., 2001). For instance, ghrelinplays a critically important role in thecontrol of appetite, stimulating food intake(Lawrence et al., 2002).

Ghrelin, when injected intracerebroven-tricularly into rodents, stimulates feedingbehavior and an increase in body weight(Nakazato et al., 2001; Tschop et al., 2000).Synthetic GHSs and the endogenous hormone,ghrelin, induce immediate gene c-fos onlyin the ARC, the site that also contains neu-ropeptide Y (NPY; Dickson et al., 1993; Dick-son and Luckman, 1997; Hewson and Dick-son, 2000). Additionally, the ARC and NPYneurons possess GHS-R mRNA, and theseneurons also project to orexinand melanin-concentrating hormone (MCH)-containingneurons in the lateral hypothalamus, neuronsthat affect feed intake (Broberger et al., 1998;Horvath et al., 1999). The effects of ghre-lin on feed intake appear to be mediated viaNPY. Preadministration of the Y1 NPY recep-tor antagonist (BIB03304) blocks both (a) thestimulatory effects of intracerebroventricular(icv) injection of ghrelin or GHRP-6 on foodintake in rats and (b) the decreased body-core temperature (Shintani et al., 2001; Brownet al., 1973). Thus, NPY neurons of the ARCare likely a primary site for ghrelin increas-ing food intake.

It might be speculated that motilin mayhave analogous effects to ghrelin. Motilin isa highly conserved, 22-amino-acid polypep-tide (Brown et al., 1973) secreted by ente-rochromaffin cells of the small intestine.This peptide stimulates gastrointestinal motoractivity and seems to play a physiologi-cal role in the regulation of fasting motility

patterns (Yanaihara et al., 1990). Both IRmotilin and mRNA expression are found inthe gastrointestinal tract, brain, nerves, andother endocrine glands in several species,including monkey, man, pig, sheep, and rab-bit (Huang et al., 1998). The motilin receptor,designated motilin-R1A (MTLR1A), is a het-erotrimeric, guanosine triphosphate-binding,protein (G protein)-coupled receptor. It wasisolated from human stomach, and its aminoacid sequence was found to be 52% iden-tical to the human GHSR (Feighner et al.,1999). The high amino-acid-sequence identitybetween MTL-R1A and the GHSR suggests arole for motilin in the control of GH secretion.

4.3.4. Other Hypothalamic PeptidesInfluencing GH Release Directly

1. Thyrotropin-Releasing Hormone(TRH). The modified tripeptide, TRH,can stimulate the release of GH inmany species under some, but notunder all, physiological circumstances.

2. Pituitary Adenylate Cyclase-ActivatingPeptide (PACAP). Pituitary adenylatecyclase-activating peptide (PACAP) isa potent GH secretagogue, and haseven been proposed as the ancestralreleasing factor for GH (reviewed inSherwood et al., 2000).

3. Gonadotropin-Releasing Hormone. Inhigher vertebrates, gonadotropin-re-leasing hormone (GnRH) does notstimulate GH release. However, insome species, at specific physiologicalphases, GnRH can evoke GH secretion.

4. Leptin. The effects of leptin on GHrelease appear to be predominantlystimulatory. The presence of leptinreceptors in/on somatotropes is wellestablished (e.g., Shimon et al, 1998;Iqbal et al., 2000; Lin et al., 2000,2003).

5. Others (FMRF Amide, Corticotropin-Releasing Hormone, Galanin, andNPY). Studies, particularly in lower

Page 62: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

SIGNAL TRANSDUCTION MECHANISM SYSTEMS 55

vertebrate species, suggest that soma-totropes are promiscuous in terms ofthe peptides that will stimulate GHrelease. In some species of lower ver-tebrates, corticotropin-releasing hor-mone (CRH) has been reported tostimulate GH secretion [e.g., rep-tiles (Denver and Licht, 1990) andfish (Rousseau et al., 1999)]. Neuro-peptide Y has a direct effect of GHrelease in some species of fish (Penget al., 1993a,b). In contrast, galanincan inhibit GH release (Arvat et al.,1995; Giustina et al., 1996, 1997).Moreover, peptides related to the neu-ropeptide Phe-Met-Arg-Phe-NH2 (FM-RF amide), originally described inmollusks, stimulate GH release inamphibians. Similar peptides have beenidentified in brains of vertebrates (i.e.,rat, chicken, frog, carp) with a simi-lar RF amide motif at their C-termini(Dockray et al., 1983; Chartrel et al.,2002; Fujimoto et al., 1998; Ukenaet al., 2002). A novel hypothalamic RFamide peptide localized in the hypotha-lamus of the bullfrog has GH-releasingactivity and was designated frog GH-releasing peptide [fGRP (Koda et al.,2002)].

4.4. SIGNAL TRANSDUCTIONMECHANISM SYSTEMS

4.4.1. Adenylate Cyclase/cAMP/Protein Kinase A and PhospholipaseC-IP3-Protein Kinase C

Somatotrophs, excitable cells that exhibitspontaneous action potential, when treatedwith GHRH, depolarize the cell membranepotential and stimulate Ca2+ influx viatransmembrane Ca2+ channels, resulting inincreases in intracellular free Ca2+ ([Ca2+];Chen and Clarke, 1995; Chen, 2002). Thisinvolves both an influx of Ca2+ (via L- and

T-type voltage-sensitive Ca2+ channels) andphospholipase C hydrolysis of phosphatidylinositol, leading to mobilizing intracellularCa2+ stores (reviewed in Frohman andKineman, 1999). It is well established thatthe signal transduction for GHRH, andprobably PACAP, involves the following:

1. Binding to specific receptors in theplasma membrane of the somatotropes.

2. Activation of adenylate cyclase (iso-form II and/or IV) via the coupling pro-tein, Gs-protein cyclic 3′,5′-adenosinemonophosphate (cAMP).

3. Activation of protein kinase A. Inaddition, there is activation of phos-pholipase C-IP3-protein kinase C andvoltage-sensitive calcium channels.

Signal transduction for ghrelin and syn-thetic analogues or GHS (e.g., GHRP-6 andnonpeptidergic GHS such as L-692, 585, MK-677) involves activation of phospholipase C(via G protein), generation of inositol phos-phate and diacyl glycerol, and increased intra-cellular Ca2+ (reviewed in Bowers, 1999).Modification of the somatotroph’s electro-physiological properties leads to changes insensitivity to ghrelin or its analogues.

We have recently demonstrated, usingpharmacological blockers, that both ghrelinand a synthetic GHS can increase intracellu-lar calcium concentrations in porcine soma-totrope (Glavaski-Joksimovic et al., 2002,2003). This two-phase phenomenon involves,first, mobilization of intracellular calcium,followed by calcium entry via L chan-nels with somatotrope depolarization (seeFig. 4.5; Glavaski-Joksimovic et al., 2002).The effects of GHS are blocked by inhibitorsof adenylate cyclase and phospholipase C,supporting the involvement of both these,adenylate cyclase/cAMP/protein kinase Aand phospholipase C; in the generation ofinositol phosphate and diacyl glycerol andincreased intracellular Ca2+.

Page 63: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

56 FUSION PORES IN GROWTH-HORMONE-SECRETING CELLS OF THE PITUITARY GLAND: AN AFM STUDY

A

B

C

Time (sec)

80

100

120

140

160

180

200

L-585 10 µM

Time (sec)

0 200 400 600 800 1000 1200 1400

0 200 400 600 800 1000 1200 1400 1600 1800

0 200 400 600 800 1000 1200 1400 1600 1800

[Ca2+

] i (nM

)[C

a2+] i (

nM)

[Ca2+

] i (nM

)

80

100

h G H R H 10 µM

h G H R H 10 µM

h G H R H 10 µM

Ghrelin 1 µM

120

140

160

180

200

L-585 10 µM

Time (sec)

80

100

120

140

160

180

200

L - 585 10 µM

Ghrelin 100 nM

Figure 4.5. Inhibitory effects of ghrelin on calcium transients evoked by L-585 in isolated porcine somatotrophs.(A) Control studies on calcium transients in isolated porcine somatotropes evoked by a 2-min application of humangrowth hormone-releasing hormone (hGHRH) (10 µM) and L-585 (10 µM; n = 29). (B) Application of L-585(10 µM) 10 min after administration of ghrelin (1 µM) did not have an additive effect on [Ca2+]i in isolatedporcine somatotropes (n = 58). (C) Ghrelin (100 µM) alone did not affect [Ca2+]i , but the changes in [Ca2+]ievoked by 10 µM L-585 were almost completely blocked (n = 7). (Reprinted from Neuroendocrinology, Vol. 77,Glavaski-Joksimovic, A., Jeftinija, K., Scanes, C. G., Anderson L. L., and Jeftinija, S. Stimulatory effect of ghrelinon isolated porcine somatotropes, pp. 366–378. Copyright 2003, with permission from Karger, Basel.)

Page 64: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

IMMUNOCYTOCHEMICAL DISTRIBUTION OF SOMATOTROPHS IN PORCINE ANTERIOR PITUITARY 57

4.5. IMMUNOCYTOCHEMICALDISTRIBUTION OF SOMATOTROPHSIN PORCINE ANTERIOR PITUITARY

Growth hormone (GH) is a primary regulatorthat plays an important role in determiningbody composition to maintain a benefi-cial ratio between skeletal muscle and fat(Anderson et al., 2004). Animals deficientin GH lack normal skeletal and musculardevelopment (Ford and Anderson, 1967).At 40 days of gestation (term = 114 days),porcine fetuses have measurable concentra-tions of GH which increase 40-fold to peakvalues at about 90 days of gestation inan episodic secretion pattern (Klindt et al.,1983; Klindt and Stone, 1984). Hypophysealstalk transection in pigs and cattle obliter-ates pulsatile GH secretion; however, basalGH concentrations are sufficient for con-tinued growth (Klindt et al., 1983; Plouzeket al., 1988; Anderson et al., 1999). Imme-diately after birth, serum GH concentrationsdecline abruptly, followed by elevated hor-mone secretion 3 to 5 weeks after birth(Sun et al., 2002). These postnatal rises inserum GH secretion occur before weaning;thus dietary changes are not the cause ofthis increase (Owens et al., 1991). The objec-tive of this immunohistochemical study wasto identify the spatial distribution patternsof growth hormone secreting cells (soma-totrophs) in the newborn and prepubertalporcine pituitary.

(a) Immunohistochemistry. The immu-noperoxidase/avidin-biotin system was usedto identify immunoreactive GH cells inthe anterior pituitary. Tissue sections werewashed in 50 mM potassium phosphatebuffered saline (KPBS) and then treatedwith 0.3% hydrogen peroxide to neutralizeendogenous peroxidase activity. The sectionswere incubated for 1 h at room temper-ature with normal goat serum to blocknonspecific binding. The slides were incu-bated overnight with monkey antiporcineGH (pGH) antibody (1:500,000 for day 1

and day 42 pigs, 1:100,000 for day 100pigs). The sections were thoroughly washedand treated for 1 h with biotinylated goatantihuman IgG (1:500) and then treatedwith avidin–peroxidase compound. Peroxi-dase activity was detected by using 0.04%3,3′-diaminobenzidine tetrahydrochloride,nickel sulfate (2.5%), and 0.1% hydro-gen peroxide in 0.1 M sodium acetate for6 min to visualize immunoreactive GH cellsas a black-colored reaction product. Tis-sue sections then were dehydrated. Sectionswere mounted in acrytol (Surgipath MedicalIndustries, Graylake, IL) mounting medium.Negative control immunohistochemical testswere done by substituting normal goat serumor KPBS for the primary antisera on ran-domly selected sections of the serial sets. Nostaining was observed in any negative controlsection.

(b) Quantitative Analysis. Ten sectionsof each serial set were preliminarily exam-ined at the lowest magnification (×10 objec-tive) to confirm that the change of cell distri-bution pattern was consistent from proximalto distal levels. Three sections per pituitarygland were selected in order from proxi-mal (nearest to the brain), middle (largestpart of gland), and distal (farthest fromthe brain) levels. Anterior lobes containingsomatotrophs were divided into five radialregions (regions 1 and 5 in the lateral wingsof anterior lobe, regions 2 and 4 in theshoulder areas, and region 3 in the cen-ter) with a ×10 objective (final magnifica-tion ×125). Three different positions (posi-tion a—proximal to the intermediate lobe;position b—middle; position c—nearest theouter surface of the pituitary) were pho-tographed (unit area 30,495 µm2) along eachof the five regions with a ×40 objective (finalmagnification ×500) for quantification. Iden-tifying regions and positions within regionsprovided a method to compare the distri-bution patterns of immunoreactive soma-totrophs. Somatotrophs were counted at 15positions (5 regions × 3 positions per region)

Page 65: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

58 FUSION PORES IN GROWTH-HORMONE-SECRETING CELLS OF THE PITUITARY GLAND: AN AFM STUDY

in three sections (proximal, middle, and dis-tal levels) from each pituitary. The numberof somatotrophs in each position of the sameage group was averaged.

No differences were found among thetotal somatotrophs across the age groups (1,42, and 100 days old). A distinctive pat-tern was found in somatotroph distributionthroughout the gland in the three age groups.Characteristics of the pattern included ahigh population of somatotrophs (44 ± 1.2;mean ± standard error of the mean per30,495 µm2) in regions 1 and 5 and a lowpopulation (22 ± 1.4) in regions 2 and 4at each level (P < 0.05) (Fig. 4.6). Soma-totrophs increased 55% in region 3 fromproximal to distal levels at all ages. Soma-totrophs in region 3 at the proximal leveldecreased 33% as age increased.

The present study with immunocytochem-ical staining of somatotrophs from differentage groups required different optimal con-centrations of the primary antibody, porcineGH antibody raised in monkey. Differ-ent immunoreactive density of somatotrophs

with increasing age of the pig may reflectchanges in the number of GH vesicles orheterogeneity of the somatotrophs. Our datasuggest that there may be regional speci-ficity of cellular differentiation and transfor-mation to facilitate GH secretion in the needfor endocrine regulation during rapid grow-ing period in young pig. An imaging tech-nique for three-dimensional reconstruction ofdistribution pattern with serial sections oflarge size models would improve visualiz-ing spatial characteristics. Because the pitu-itary is an endocrine gland with a complexand heterogeneous distribution of cells whichfunctionally communicate with neighboringendocrine cells, a temporal correlation ofchanges in GH production with changes inother hypothalamic/pituitary hormones mustbe taken into account to increase our under-standing of pituitary function. The charac-teristics of spatial distribution patterns ofspecific endocrine cells in pituitary glandmay be applied to examine therapeutic strate-gies for the management of pituitary disor-ders in both physiological and pathologicalcondition.

0

20

40

60

abc

0

20

40

60

abc

0

20

40

60

1 2 3 4 5

1 2 3 4 5

1 2 3 4 5

abc

No.

of s

omat

otro

phs

per

30,4

95 µ

m2

(ii) Middle

(iii) Distal

(i) Proximal

RegionLevel

NL

IL

AL

Figure 4.6. Schematic diagram depicts a general spatial distribution pattern of porcine somatotrophs in all agegroups as indicated by gray dots (left). NL, neural lobe (posterior pituitary); IL, intermediate lobe; AL, anterior lobe(anterior pituitary). Proximal (nearest to the brain), middle (largest part of gland), and distal (farthest from the brain)levels. Graphs (left) indicated mean numbers of somatotrophs per 30,495 µm2 ± standard error of the mean.

Page 66: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

LIVE GH-SECRETING CELLS REVEALED USING ATOMIC FORCE MICROSCOPY 59

4.6. STRUCTURE AND DYNAMICOF THE FUSION PORES IN LIVEGH-SECRETING CELLS REVEALEDUSING ATOMIC FORCE MICROSCOPY

Until recently, our understanding of secretoryvesicle fusion at the cell plasma membranewas obtained from morphological, electro-physiological, and biochemical studies allsuggesting the presence of “fusion pores” atthe cell plasma membrane (Anderson, 2004).Using atomic force microscopy (AFM), thestructure and dynamics of the “fusion pore”were first revealed and examined in live pan-creatic acinar cells. In live resting pancreaticacinar cells, “pits” measuring 0.5–2 µm andcontaining 3–20 “depressions” of 100- to180-nm diameter were identified only at theapical region of these cells where membrane-bound secretory vesicles are known to dockand fuse. Following stimulation of secretion,only “depression” enlarged and returned to

resting size following completion of secre-tion. Exposure of acinar cells to cytocha-lasin B, a fungal toxin that inhibits actinpolymerization, resulted in a 50–60% lossof stimulated amylase secretion. A signif-icant decrease in depression diameter wasalso observed in acinar cells exposed to thefungal toxin. To determine if similar struc-tures are present in neuroendocrine cells, theGH-secreting cell of the pig pituitary wasstudied using AFM and transmission electronmicroscopy (TEM). Results from this studydemonstrate the presence of pits and depres-sions in GH-secreting cells of the pituitaryand their involvement in hormone release.

Reverse hemolytic plaque assay on iso-lated GH-secreting cells of the pig pituitarydemonstrated GH release following expo-sure to a nonpeptidyl GH secretagogue (L-692,585) (Fig. 4.7). Examination of restingGH cells revealed the presence of “pits”and “depressions” at the plasma membrane.

Mea

n pl

aque

are

a (µµ

m2 ×

10−2

)

0

10

20

30

40

50

Control L-585 100 nM

**

B

Per

cent

of p

laqu

e fo

rmin

g ce

lls (

%)

0

10

20

30

40

50

Control L-585 100 nM

*

A C

Tot

al s

ecre

tion

inde

x (×

10−2

)

02468

1012141618

Control L-585 100 nM

**

Figure 4.7. Light microscopy revealing the extent of growth hormone release from isolated GH cells of the pituitarygland, in a typical reverse hemolytic plaque assay. The larger the plaque (clear area), the greater the release. Notethe presence of a small plaque in a resting GH cell (A), compared to an L-692,585 stimulated cell (B).

Page 67: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

60 FUSION PORES IN GROWTH-HORMONE-SECRETING CELLS OF THE PITUITARY GLAND: AN AFM STUDY

Control + L-692,585

Control + L-692,585

+ L-692,585 GH Ab + Gold

A B

C D

E F

Figure 4.8. High-resolution atomic force microscopy(AFM) performed on resting and stimulated GH-secret-ing cells, after fixation. Note a large area of a cellsurface in resting (A) and in a stimulated (B) cell. AFMmicrographs of a “pit” with “depressions,” in restingcell (C), again clearly demonstrating the enlargementof porosomes or fusion pores following stimulationof secretion (D). Exposure of “pits” in a stimulatedcell (E) to 30-nm gold-tagged GH-antibody results inbinding of released growth hormone at the site to 30-nmgold-tagged GH-antibody (F). Note the large amountsof gold-tagged antibody binding at these sites obscuringthe porosome opening.

Depressions in resting cells measure 154 ±4.5 nm. However, following exposure ofGH cells to the secretagogue L-692,585, amarked increase in the size of “depressions”is demonstrated (Fig. 4.8). When stimulatedlive cells were exposed to 30-nm gold-taggedGH-antibody, gold particles were found todecorate “pit” and “depression” structures(Fig. 4.8). Aldehydes fixation of biologicalsamples is known to result in decreasedelasticity and increased hardness. Since force

spectroscopy is used to image objects usingthe AFM, less elastic samples are betterresolved. From studies using that pancreaticacinar cells, it was determined that 1 h treat-ment of ice-cold 2.5% paraformaldehyde inphosphate buffered saline, pH 7.5, was idealin retaining the structural integrity of “pits”and “depressions.” No detectable changeswere identified in live cells following fixa-tion. To obtain high-resolution images of thecells and of the “pits” and “depressions” andto determine the distribution of 30-nm GH-immunogold labeling, stimulated GH cellswere fixed following immunogold labeling.In conformation with our observation in liveGH cells, AFM images of the immunolabeledfixed cells demonstrate specific localizationof immunogold at “depressions” (Fig. 4.8),implicating them to be secretory sites atthe cell plasma membrane. In agreementwith earlier studies in pancreatic acinar cells,the GH-secreting neuroendocrine cells of thepituitary demonstrate the presence of “pits”and “depressions” at the plasma membrane,where secretory vesicles dock and fuse torelease vesicular contents. The presence of“pits” and “depressions” in both exocrineand neuroendocrine cells suggests that thesestructures may be universal to secretory cells,where exocytosis occurs (Fig. 4.9).

4.7. NUMBER OF SECRETORYVESICLES IN GROWTH HORMONECELLS OF THE PITUITARY REMAINSUNCHANGED AFTER SECRETION

Physiological processes such as neurotrans-mission and the secretion of enzymes or hor-mones require fusion of membrane-boundedsecretory vesicles at the cell plasma mem-brane and the consequent release of vesicu-lar contents. It has been commonly acceptedthat exocytosis requires the total incorpora-tion of secretory vesicle membrane into thecell plasma membrane for release of vesicu-lar contents; however, studies in the last de-cade (Cho et al., 2002c–e; Jena et al., 2003;

Page 68: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

NUMBER OF SECRETORY VESICLES IN GROWTH HORMONE CELLS 61

A

ActinGH

B

C

Figure 4.9. Schematic diagrams depict cross sectionalviews at the cell plasma membrane of a “pit” andwith porosomes, and a vesicle containing growthhormone attached to one. Immediately followingGH-secretagogue stimulation, the secretory vesicle con-taining hormone docks and fuses with the porosome(A) and then releases hormone (B) through the poro-some. After hormone release from the vesicle (C) theporosome opening becomes smaller and the vesicle dis-associates from it.

Jeremic et al., 2003; Lawson et al., 1975;Platter et al., 1997; Schneider et al., 1997)clearly demonstrate otherwise. Transmis-sion electron microscopy (TEM) studies onmast cells demonstrate that, after stimula-tion of secretion, several intact as well asempty and partly empty secretory vesiclesare present (Cho et al., 2002f). Quantitative

TEM on stimulated and resting bovine chro-maffin cells of the adrenal cortex showedno significant change in the number ofperipheral dense-core vesicles after stimu-lation of secretion (Plattner et al., 1997).Similarly, combined studies using atomicforce microscopy (AFM) and TEM clearlydemonstrate no change in the total num-ber of secretory vesicles following secre-tion in pancreatic acinar cells (Cho et al.,2002b). Fusion pores or depressions in pan-creatic acinar- or growth hormone (GH)-secreting cells are cone-shaped structuresat the plasma membrane, with a 100- to150-nm-diameter opening (Cho et al., 2002c;Schneider et al., 1997). Membrane-boundedsecretory vesicles range in diameter from0.2 to 1.2 µm dock and fuse at depres-sions to release vesicular contents. Afterfusion of secretory vesicles at depressions, a20–40% increase in depression diameter hasbeen demonstrated. It has therefore been con-cluded that secretory vesicles “transiently”dock and fuse at depressions (Cho et al.,2002a; Jena, 2002; Jena et al., 1997). In con-trast to accepted belief, if secretory vesicleswere to completely incorporate at depres-sions, the fusion pore would distend muchwider than observed. Additionally, if secre-tory vesicles were to completely fuse at theplasma membrane, there would be a decreasein total number of vesicles after secretion.

Although the fusion of secretory vesi-cles at the cell plasma membrane occurstransiently, complete incorporation of mem-brane at the cell plasma membrane takesplace when cells need to incorporate signal-ing molecules like receptors, second messen-gers, and ion channels. Therefore, in GH-secreting cells, transient fusion is suggestedand no change in total number of vesiclesis hypothesized. The present study has beenundertaken to test this hypothesis (Lee et al.,2004).

Control (resting) pituitary cells exposedto PBS contained more than twice asmany filled vesicles than did the stimulated

Page 69: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

62 FUSION PORES IN GROWTH-HORMONE-SECRETING CELLS OF THE PITUITARY GLAND: AN AFM STUDY

cells exposed to the GH secretagogue, L-692,585 (4.9 ± 0.21 in control, 2.3 ± 0.23in stimulated; P < 0.001). Stimulated cellscontained nearly twice as many empty vesi-cles (0.6 ± 0.13 in control, 1.2 ± 0.16 instimulated; P < 0.05) and 2.5 times morepartly empty vesicles than did control cells(1.1 ± 0.08 in control, 2.6 ± 0.12 in stimu-lated; P < 0.001). However, there was nosignificant difference in total number of vesi-cles between control and stimulated pituitarycells. The remarkable increase in number ofempty and partly empty vesicles in stimu-lated cells compared with control cells is evi-dent. The intracellular distribution of emptyor partially empty vesicles seemed random instimulated cells. Immunogold labeling withGH antibody occurs only in electron denseGH vesicles in both control and stimulatedcells. There was a complete absence ofimmunogold labeling with GH antibody inempty vesicles or other areas of the cyto-plasm. Brief exposure of cultured porcinepituitary cells to the nonpeptidyl L-692,585,ghrelin, or human GH-releasing hormone(hGHRH) evoked a marked increase inintracellular calcium [Ca2+]i concentration(Glavaski-Joksimovic et al., 2002, 2003).The result of stimulating live GH cells for90 s with L-692,585 is that filled secretory

vesicles empty very rapidly, most likelyby docking at the plasma membrane. Afterreleasing vesicular contents, the empty orpartly empty vesicles return to the cytoplas-mic compartments of the cell.

From our study on GH cells, 90-s expo-sure to 20 mM L-692,585 (GH secreta-gogue) for stimulation of GH secretion wasideal in inducing rapid and effective exo-cytosis. L-692,585 intravenously adminis-tered to pigs also causes an immediate peakrelease of GH > 80 ng/ml peripheral plasmawithin 10 min compared with pulsatile GHpeaks of 6–10 ng/ml in placebo treated con-trols (Fig. 4.10). Intracellular signal trans-duction of GH-secretagogue undergoes aphosphoinositol-protein kinase C pathwaythat induces intracellular Ca2+ accumulationand depolarization, leading to exocytosis ofGH-containing vesicles. The step increase inplasma membrane capacitance of GH cells,therefore, may result from a rapid transientfusion which is consistent with the AFMobservation that shows the presence of “pits”which contain “depressions” or fusion pores(porosomes) at the GH cell plasma mem-brane that increase markedly in diameterafter L-692,585 exposure.

The data reported here clearly showthat the total number of secretory vesicles

Figure 4.10. Electron micrograph of secretory vesicles in porcine pituitary gland. (A) Typical structure of tissueembedded in Epon-Araldite. (B) Secretory granules embedded in unicryl, sectioned and specifically labeled withanti-GH and a gold-conjugated second antibody. Note the immunogold labeled vesicles, demonstrating the presenceof growth hormone within.

Page 70: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 63

in porcine pituitary cells is not decreasedafter exocytosis. Such a decrease would berequired if the secretory vesicle membranewere to fuse with the plasma membrane (asin the generally accepted model for secre-tion). The present data are consistent with amechanism where vesicles transiently dockand fuse at the fusion pore (porosome) torelease vesicular contents after stimulationof secretion. Based on these and other sup-porting findings, transient fusion of secretoryvesicles at the porosome on the plasma mem-brane may be universal in the process ofexocytosis.

ACKNOWLEDGMENT

This work was supported by research grantUSDA NRI 2003-35206-12817 (L.L.A.) andthe Iowa Agriculture and Home EconomicsExperiment Station, Ames, Iowa; by HatchAct and State of Iowa funds; and by NIHgrants DK56212 and NS39918 (B.P.J.).

REFERENCES

Anderson, L. L. (2004). Discovery of a newcellular structure: The porosome, elucidationof the molecular mechanism of secretion. CellBiol. Int. 28:3–5.

Anderson, L. L., Hard, D. L., Trenkle, A. H., andCho, S.-J. (1999). Long term growth afterhypophyseal stalk transection and hypophysec-tomy of beef calves. Endocrinology 140:2405–2414.

Anderson, L. L., Jeftinija, S., and Scanes, C. G.(2004). Growth hormone secretion: Molec-ular and cellular mechanisms and in vivoapproaches. Exp. Biol. Med. 229:291–302.

Arvat, E., Gianotti, L., Ramunni, J, Grottoli, S.,Brossa, P. C., Bertagna, A., Camanni, F., andGhigo, E. (1995). Effect of galanin on basaland stimulated secretion of prolactin gonado-tropins, thyrotropin, adrenocorticotropin andcortisol in humans. Eur. J. Endocrinol. 133:300–304.

Bennet, P. A., Thomas, G. B., Howard, A. D.,Feighner, S. D., Van der Ploeg, L. H. T.,

Smith, R. G., and Robinson, I. C. (1997). Hy-pothalamic growth hormone secretagogue-rece-ptor (GHS-R) expression is regulated bygrowth hormone in the rat. Endocrinology138:4522–4557.

Bowers, C. Y. (1999). Growth hormone-releasingpeptides. In: Handbook of Physiology, Sec-tion 7: The Endocrine System, Vol. V: Hor-monal Control of Growth (J. L. Kostyo andH. M. Goodman, eds.), pp. 187–219, Ameri-can Physiological Society, Oxford UniversityPress, New York.

Broberger, C., DeLecea, L., Sutcliffe, J. C., andHorfelt, T. (1998). Hypocretin/orexin- and me-lanin-concentrating hormone-expressing cellsfrom distinct populations in the rodent lateralhypothalamus relationship to the neuropeptideY and agouti gene-related protein systems. J.Comp. Neurol. 402:406–474.

Brown, J. C., Cook, M. A., and Dryburgh, T.(1973). Motilin, a gastric motor activity stim-ulating polypeptide: The complete amino acidsequence. Can. J. Biochem. 51:533–537.

Chartrel, N., Dujardin, C., Leprince, J., Desrues,L., Tonon, M. C., Cellier, E., Cosette, P., Jou-enne, T., Simonnet, G., and Vaudry, H. (2002).Isolation, characterization, and distribution of anovel neuropeptide, Rana RFamide (R-RFa),in the brain of the European green frog Ranaesculenta . J. Comp. Neurol. 448:111–127.

Chen, C. (2002). The effect of two-day treatmentof primary cultured ovine somatotropes withGHRP-2 on membrane voltage-grated K+ cur-rents. Endocrinology 143:2659–2663.

Chen, C., and Clarke, I. J. (1995). Modulationof Ca2+ influx in the ovine somatotrophby growth hormone-releasing factor. Am. J.Physiol. 268:E204–E212.

Cho, S.-J., Abdus Sattar, A. K. M., Jeong, E. H.,Satchi, M., Cho, J., Dash, S., Mayes, M. S.,Stromer, M. H., and Jena B. P. (2002a). Aqua-porin 1 regulates GTP induced rapid gating ofwater in secretory vesicles. Proc. Natl. Acad.Sci. USA 99:4720–4724.

Cho, S.-J., Cho, J., and Jena, B. P. (2002b). Thenumber of secretory vesicles remains unchangedfollowing exocytosis. Cell Biol. Int. 26:29–33.

Cho, S.-J., Jeftinija, K., Glavaski, A., Jeftinija, S.,Jena, B. P., and Anderson, L. L. (2002c). Struc-ture and dynamics of the fusion pores in live

Page 71: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

64 FUSION PORES IN GROWTH-HORMONE-SECRETING CELLS OF THE PITUITARY GLAND: AN AFM STUDY

GH-secreting cells revealed using atomic forcemicroscopy. Endocrinology 143:1144–1148.

Cho, S.-J., Kelly, M., Rognlien, K. T., Cho, J.,Horber, J. K., and Jena, B. P. (2002d). SNA-REs in opposing bilayers interact in a circulararray to form conducting pores. Biophys. J.83:2522–2527.

Cho, S.-J., Quinn, A. S., Stromer, M. H., Dash,S., Cho, J., Taatjes, D. J., and Jena, B. P.(2002e). Structure and dynamics of the fusionpore in live cells. Cell Biol. Int. 26:35–42.

Cho, S.-J., Wakade, A., Pappas, G. D., and Jena,B. P. (2002f). New structure involved in tran-sient membrane fusion and exocytosis. Ann.New York Acad. Sci. 971:254–256.

Denver, R. J., and Licht, P. (1990). Modula-tion of neuropeptide-stimulated pituitary hor-mone secretion in hatching turtles. Gen. Comp.Endocrinol. 77:107–115.

Dickson, S. L., and Luckman, S. M. (1997). In-duction of c-fos messenger ribonucleic acid inneuropeptide Y and growth hormone (GH)-releasing factor neurons in the rat arcu-ate nucleus following systemic injection ofthe GH secretagogue, GH-releasing-peptide-6.Endocrinology 138:771–777.

Dickson, S. L., Ling, G., and Robinson, I. C.(1993). Systemic administration of growthhormone-releasing peptide activates hypotha-lamic arcuate neurons. Neuroscience 53:303–306.

Dockray, G. J., Reeve, J. R., Jr, Shively, J., Gay-ton, R. J., and Barnard, C. S. (1983). A novelactive pentapeptide from chicken brain iden-tified by antibodies to FMRF-amide. Nature305:328–330.

Feighner, S. D., Tan, C. P., McKee, K. K., Paly-ha, O C., Hreniuk, D. L., Pong, S. S., Austin,C. P., Figueroa, D., MacNeil, D., Cascieri,M. A., Nargund, R., Bakshi, R., Abramovitz,M., Stocco, R., Kargman, S., O’Neill, G., VanDer Ploeg, L. H. D., Evans, J., Patchett, A. A.,Smith, R. G., and Howard, A. D. (1999). Re-ceptor for motilin identified in the human gas-trointestinal system. Science 284:2184–2188.

Ford, J. J., and Anderson, L. L. (1967). Growthin immature hypophysectomized pigs. J. Endo-crinol. 37:347–348.

Frohman, L. A., and Kineman, R. D. (1999).Growth hormone-releasing hormone: Discov-ery, regulation, and actions. In: Handbook

of Physiology, Section 7: The Endocrine Sys-tem, Vol. V: Hormonal Control of Growth(J. L. Kostyo and H. M. Goodman, eds.), pp.187–219, American Physiological Society,Oxford University Press, New York.

Fujimoto, M., Takeshita, K., Wang, X., Taka-batake, I., Fujisawa, Y., Teranishi, H., Ohtani,M., Muneoka, Y., and Ohta, S. (1998). Isola-tion and characterization of a novel bioactivepeptide, Carassius Rfamide (C-Rfa), from thebrain of the Japanese crucian carp. Biochem.Biophys. Res. Commun. 242:436–440.

Giustina, A., Monfanti, C., Licini, M., Stefana,B., Ragni, G., and Turano, A. (1996). Effect ofgalanin on growth hormone (GH) response tothyrotropin releasing hormone of rat pituitaryGH-secreting adenomatous cells (GH1) in cul-ture. Life Sci. 58:83–90.

Giustina, A., Ragni, G., Bollati, A., Cozzi, R.,Licini, M., Poiesi, C., Turazzi, S., and Mon-fanti, C. (1997). Inhibitory effects of galaninon growth hormone (GH) release in cul-tured GH-secreting adenoma cells: comparativestudy with octreotide, GH-releasing hormone,and thyrotropin-releasing hormone. Metabolism46:425–430.

Glavaski-Joksimovic, A., Jeftinija, K., Jeremic,A., Anderson, L. L., and Jeftinija, S. (2002).Mechanism of action of the growth hormonesecretagogue, L-692,585, on isolated porcinesomatotropes. J. Endocrinol. 175:625–636.

Glavaski-Joksimovic, A., Jeftinija, K., Scanes,C. G., Anderson, L. L., and Jeftinija, S. (2003).Stimulatory effect of ghrelin on isolated porcinesomatotropes. Neuroendocrinology 77:366–378.

Guan, X. M., Yum, H., Palyha, O. C., McKee,K. K., Feighner, S. D., Sirinath-Singhji, D. J.,Smith, R. G., Van der Ploeg, L. H. T., andHoward, A. D. (1997). Distribution of mRNAencoding the growth hormone secretagoguereceptor in brain and peripheral tissues. BrainRes. Mol. Brain Res. 48:23–29.

Hewson, A. K., and Dickson, S. L. (2000). Sys-temic administration of ghrelin induces Fosand Egr-1 proteins in the hypothalamic arcu-ate nucleus of fasted and fed rats. J. Neuroen-docrinol. 12:1047–1049.

Horvath, T. L., Diano, S., and van Den Pol, A. N.(1999). Synaptic interaction between hypocre-tin (orexin) and neuropeptide Y cells in the

Page 72: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 65

rodent and primate hypothalamus: A novel cir-cuit implicated in metabolic and endocrine reg-ulations. J. Neurosci. 19:1072–1087.

Howard, A. D., Feighner, S. D., Cully, D. F.,Arena, J. P., Liverator, P. A., Rosenblum, C. I.,Hamelin, M., Hreniuk, D. L., Palyha, O. C.,Anderson, J., Paress, P. S., Diax, C, Dhou, M,Liu, K. K., McKee, K. K., Pong, S. S., Chaung,L. Y., Elverecht, A., Dashkevicz, M., Hea-vens, R., Rigby, M., Sirinath-Singhji, D. J.,Dean, D. C., Melillo, D. G., Patchett, A. A.,Nargund, R., Griffion, P. R., DeMaritino, J. A.,Gupta, S. K., Schaeffer, J. M., Smith, R. G., andVan der Ploeg, L. H. T. (1996). A receptor inpituitary and hypothalamus that functions ingrowth hormone release. Science 273:974–977.

Huang, Z., De Clercq, P., Depoortere, I., andPeeters, T. L. (1998). Isolation and sequenceof cDNA encoding the motilin precursor frommonkey intestine. Demonstration of the motilinprecursor in the monkey brain. FEBS Lett.435:149–152.

Iqbal, J., Pompolo, S., Considine, R. V., andClarke, I. J. (2000). Localization of leptinreceptor-like immunoreactivity in the corti-cotropes, somatotropes, and gonadotropes inthe ovine anterior pituitary. Endocrinology141:1515–1520.

Jena, B. P. (2002). Fusion pore in live cells. NewsPhysiol. Sci. 17:219–222.

Jena, B. P., Schneider, S. W., Geibel, J. P., Web-ster, P., Oberleithner, H., and Sritharan, K. C.(1997). Gi regulation of secretory vesicleswelling examined by atomic force microscopy.Proc. Natl. Acad. Sci. USA 94:13317–13322.

Jena, B. P., Cho, S.-J., Jeremic, A., Stromer,M. H., and Abu-Hamdah, R. (2003). Structureand composition of the fusion pore. Biophys. J.84:1337–1343.

Jeremic, A., Kelly, M., Cho, S.-J., Stromer,M. H., and Jena, B. P. (2003). Reconstitutedfusion pore. Biophys. J. 85:2035–2043.

Klindt, J., and Stone, R. T. (1984). Porcine growthhormone and prolactin: Concentrations in thefetus and secretory patterns in the growing pig.Growth 48:1–15.

Klindt, J., Ford, J. J., Beradinelli, G., and Ander-son, L. L. (1983). Growth hormone secretionafter hypophyseal stalk transection in pigs.Proc. Soc. Exp. Biol. Med. 172:503–513.

Koda, A., Ukena, K., Teranishi, H., Ohta, S.,Yamamoto, K., Kikuyama, S., and Tsutsui, K.(2002). A novel amphibian hypothalamic neu-ropeptide: Isolation, localization, and biologicalactivity. Endocrinology 143:411–419.

Kojima, M., Hosoda, H., Date, Y., Nakazato, M.,Matsuo, H., and Kangawa, K. (1999). Ghrelinis a growth-hormone-releasing peptide fromstomach. Nature 402:656–660.

Lawrence, C. B., Snape, A. C., Baudoin, F. M.H., and Luckman, S. M. (2002). Acute cen-tral ghrelin and GH secretagogues induce feed-ing and activate brain and appetite centers.Endocrinology 143:155–162.

Lawson, D., Fewtrell, C., Gomperts, B., and Raff,M. C. (1975). Anti-immunoglobulin-inducedhistamine secretion by rat peritoneal mast cellsstudied by immunoferritin electron microscopy.J. Exp. Med. 142:391–401.

Lee, H. M., Wahng, G., Englander, E. W., Koji-ma, M., and Greeley, G. H., Jr. (2002). Ghre-lin, a new gastrointestinal endocrine peptidethat stimulates insulin secretion: Enteric dis-tribution, ontogeny, influence of endocrine,and dietary manipulations. Endocrinology 143:185–190.

Lee, J.-S., Mayes, M. S., Stromer, M. H., Scanes,C. G., Jeftinija, S., and Anderson, L. L. (2004).Number of secretory vesicles in growth hor-mone cells of the pituitary remains unchangedafter secretion. Exp. Biol. Med. 229:632–639.

Leshin, L. S., Barb, C. R., Kiser, T. E., Ram-pacek, G. B., and Kraeling, R. R. (1994).Growth hormone-releasing hormone and soma-tostatin neurons within the porcine and bovinehypothalamus. Neuroendocrinology 59:251–264.

Lin, J. Barb, C. R., Matteri, R. L., Kraeling,R. R., Chen, S., Meinersmann, R. J., and Ram-pacek, G. B. (2000). Long from leptin recep-tor mRNA expression in the brain, pituitary,and other tissues in the pig. Domest. Anim.Endocrinol. 19:53–61.

Lin, J. Barb, C. R., Matteri, R. L., Kraeling,R. R., and Rampacek, G. B. (2003). Growthhormone releasing factor decreased long formleptin receptor expression in porcine ante-rior pituitary cells. Domest. Anim. Endocrinol.24:95–101.

Masuda, Y., Tanaka, T., Inomata, N., Ohnuma, N.,Tanaka, S., Itoh, Z., Hosoda, H., Kojima, M.,

Page 73: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

66 FUSION PORES IN GROWTH-HORMONE-SECRETING CELLS OF THE PITUITARY GLAND: AN AFM STUDY

and Kangawa, K. (2000). Ghrelin stimulatesgastric acid secretion and motility in rats.Biochem. Biophys. Res. Commun. 276:905–908.

Mitchell, V., Bouret, S., Meauvillian, J. C.,Schilling, A., Perret, M., Kordan, C., and Epel-baum, J. (2001). Comparative distribution ofmRNA encoding the growth hormone secreta-gogue-receptor (GHS-R) in Microcebus muri-nus (primate, lemurian) and rat forebrain andpituitary. J. Comp. Neurol. 429:469–489.

Molina, J. R., Klindt, J., Ford, J. J., and Ander-son, L. L. (1986). Growth hormone and pro-lactin secretion after hypothalamic deafferenta-tion in pigs. Proc. Soc. Exp. Biol. Med.183:163–168.

Nakazato, M., Murakami, N., Date, Y., Kojima,M., Matsuo, H., Kangawa, K., and Matsukura,S. (2001). A role for ghrelin in the centralregulation of feeding. Nature 409:194–198.

Owens, P. C., Campbell, R. G., Johnson, R. G.,King, R., and Ballard, F. J. (1991). Develop-mental change in growth hormone, insulin-likegrowth factors (IGF-I and IGF-II) and IGF-binding proteins in plasma of young growingpigs. J. Endocrinol. 128:439–447.

Peng, C., Chang, J. P., Yu, K. L., Wong, A. O.,Van Goor, R., Peter, R. E., and Rivier, J. E.(1993a). Neuropeptide-Y stimulates growthhormone and gonadotropin-II secretion in thegoldfish pituitary: Involvement of both presy-naptic and pituitary cell actions. Endocrinology132:1820–1829.

Peng, C., Humphries, S., Peter, R. E., Rivier, J. E.,Blomqivist, A. G., and Larhammar, D. (1993b).Actions of goldfish neuropeptide Y on the secre-tion of growth hormone and gonadotropin-IIin female goldfish. Gen. Comp. Endocrinol.90:306–317.

Plattner, H., Artalejo, A. R., and Neher, E. (1997).Ultrastructural organization of bovine chromaf-fin cell cortex—analysis by cryofixation andmorphometry of aspects pertinent to exocytosis.J. Cell Biol. 139:1709–1717.

Plouzek, C. A., Molina, J. R., Hard, D. L., Vale,W., Rivier, J., Trenkle, A., and Anderson, L. L.(1988). Effect of growth hormone-releasingfactor and somatostatin on growth hor-mone secretion in hypophysial stalk transected

beef calves. Proc. Soc. Exp. Biol. Med.189:158–167.

Radecki, S. V., Deaver, D. R., and Scanes, C. G.(1994). Triiodothyronine reduces growth hor-mone secretion and pituitary growth hormonemRNA in the chicken, in vivo and in vitro.Proc. Soc. Exp. Biol. Med. 205:340–346.

Reed, D. K, Korytko, A. I., Hipkin, R. W., We-hernberg, W. B, Schonbrunn, A., and Cut-tler, L. (1999). Pituitary somatostatin recep-tor (sst)1-5 expression during rat development:age-dependent expression of sst2. Endocrinol-ogy 140:4739–4744.

Rousseau, K., Le Belle, N, Marchelidon, J., andDufour, S. (1999). Evidence that corticotropin-releasing hormone acts as growth hormone-releasing factor in a primitive teleost, theEuropean eel (Anguilla anuilla). J. Neuroen-docrinol. 11:385–392.

Sherwood, N. M, Krueckl, S. L., and McRory,J. E. (2000). The origin and function of the

pituitary adenylate cyclase-activation polypep-tide (PACAP)/glucagons superfamily. Endocr.Rev. 21:619–670.

Shimon, I., Yan, X., Magoffin, D. A, Friedman,T. C., and Melmed, S. (1998). Intact leptinreceptor is selectively expressed in humanfetal pituitary and pituitary adenomas andsignals human fetal pituitary growth hor-mone secretion. J. Clin. Endocrinol. Metab.83:4059–4064.

Schneider, S. W., Sritharan, K. C., Geibel, J. P.,Oberleithner, H., and Jena, B. P. (1997). Sur-face dynamics in living acinar cells imagedby atomic force microscopy: Identification ofplasma membrane structures involved in exocy-tosis. Proc. Natl. Acad. Sci. USA 94:316–321.

Shintani, M., Ogawa, Y., Ebihara, K., Aiszawa-Abe, M., Miyanaga, F., Takaya, K, Hayashi, T.,Inove, G., Hosoda, K, Kojima, M., Kangawa,K., and Nakao, K. (2001). Ghrelin, an endoge-nous growth hormone secretagogue, is a novelorexigenic peptide that antagonizes leptin actionthrough activation of hypothalamic neuropep-tide Y/Y1 receptor pathway. Diabetes 50:227–232.

Silverman, B. L., Kaplan, S. L., Grumbach,M. M., and Miller, W. L. (1988). Hormonal

Page 74: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 67

regulation of growth hormone secretion andmessenger ribonucleic acid accumulation incultured bovine pituitary cells. Endocrinology122:1236–1241.

Sun, H. S., Anderson, L. L., Yu, T.-P., Kim,K.-S., Klindt, J., and Tuggle, C. K. (2002).Neonatal Meishan pigs show POU1F1 geno-type effects on plasma GH and PRL concentra-tion. Anim. Reprod. Sci. 69:223–237.

Tannenbaum, G. S., and Epelbaum, J. (1999).Somatostatin. In: Handbook of Physiology,Section 7: The Endocrine System, Vol. V: Hor-monal Control of Growth (J. L. Kostyo andH. M. Goodman, eds.), pp. 221–265. Ameri-can Physiological Society, Oxford UniversityPress, New York.

Tschop, M., Smiley, D. L., and Heiman, M. L.(2000). Ghrelin induces adiposity in rodents.Nature 407:903–913.

Ukena, K., Iwakoshi, E., Minakata, H., and Tsut-sui, K. (2002). A novel rat hypothalamic

Rfamide-related peptide identified by immuno-affinity chromatography and mass spectrome-try. FEBS Lett. 512:255–258.

Vasilatos-Younken, R., Tsao, P. H., Foster, D. N.,Smiley, D. L., Bryant, H., and Heiman, M. L.(1992). Restoration of juvenile baseline growthhormone secretion with preservation of theultradian growth hormone rhythm by continu-ous delivery of growth hormone-releasing fac-tor. J. Endocrinol. 135:371–382.

Yanaihara, N., Yanaihara, C., Mochizuki, T.,Iguchi, K, and Hoshino, M. (1990). Chemi-cal synthesis, radioimmunoassay, and distri-bution of immuno-reactivity of motilin. In:Motilin (Z. Itoh, ed.), pp. 31–46. AcademicPress, Tokyo.

Yokote, R., Sato, M., Matsubara, S., Ohye, H.,Niimi, M., Murao, K., and Takahara, J. (1998).Molecular cloning and gene expression ofgrowth hormone-releasing peptide receptor inrat tissue. Peptides 19:15–20.

Page 75: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

CHAPTER 5

PROPERTIES OF MICROBIAL CELL SURFACESEXAMINED BY ATOMIC FORCE MICROSCOPYYVES F. DUFRENEUnite de chimie des interfaces, Universite catholique de Louvain, Croix du Sud 2/18, B-1348Louvain-la-Neuve, Belgium

5.1. INTRODUCTION

During the past 40 years, the importanceof the microbial cell surface in biology,medicine, industry, and ecology has beenincreasingly recognized. Because they con-stitute the frontier between the cells andtheir environment, microbial cell walls playseveral key functions: supporting the inter-nal turgor pressure of the cell, protectingthe cytoplasm from the outer environment,imparting shape to the organism, and actingas a molecular sieve and controlling inter-facial interactions (i.e., molecular recogni-tion, cell adhesion, and aggregation). Thesebiointerfacial processes have major conse-quences, which can be either beneficial, suchas in biotechnology (wastewater treatment,bioremediation, immobilized cells in reac-tors), or detrimental, such as in industrialsystems (biofouling, contamination) and inmedicine (interactions of pathogens with ani-mal host tissues; accumulation on implantsand prosthetic devices) (Savage and Fletcher,1985). Hence, there is considerable interestin improving our current understanding ofthe functions of microbial cell surfaces.

The functions of the cell surface are directlyrelated to structure. As opposed to animal

cells, microbial cells are surrounded by thick,mechanically strong cell walls (Hancock,1991). The wall mechanical strength in eubac-teria is provided by a layer of peptidogly-can consisting of glycan chains of alternatingN -acetylglucosamine and N -acetylmuramicacid residues, which are cross-linked by shortpeptide chains. Archaebacteria possess stress-bearing wall components which may havedifferent forms: peptidoglycan-like polymers,proteinaceous sheats, crystalline glycoproteinarrays (S-layers). In gram-positive bacteria,the peptidoglycan is bound to anionic poly-mers (e.g., teichoic acid) while in gram-negative bacteria it is overlayed by anouter membrane—that is, an asymmetricalbilayer of phospholipids and lipopolysaccha-rides containing membrane proteins (e.g.,porins). For many bacterial strains, these cellwall constituents are covered by additionalsurface layers in the form of polysaccharidecapsules, surface appendages (fimbriae, pili,fibrils, flagella), or regular crystalline arrays of(glyco)proteins, referred to as S-layers. Strongcell walls are formed in yeasts and filamen-tous fungi by the aggregation of polysaccha-ride polymers. In yeasts, these are made of amicrofibrillar array of β1–3 glucan, overlaidby β1–6 glucan and mannoproteins. The walls

Force Microscopy: Applications in Biology and Medicine, edited by Bhanu P. Jena and J.K. Heinrich Horber.Copyright 2006 John Wiley & Sons, Inc.

69

Page 76: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

70 PROPERTIES OF MICROBIAL CELL SURFACES EXAMINED BY ATOMIC FORCE MICROSCOPY

of fungal hyphae consist of microfibrillarpolysaccharides, chitin or cellulose, coveredby layers of proteins and glucans. For fun-gal spores, the wall is often covered by anouter layer of regularly arranged proteina-ceous rodlets having a periodicity of about10 nm.

Understanding the functions of microbialcell surfaces requires determination of theirstructural and physical properties. Variouscharacterization methods have been devel-oped toward this goal (Beveridge and Gra-ham, 1991; Mozes et al., 1991): separationand chemical analysis of wall constituents,serological procedures, binding studies (dyes,antibodies and lectins), selective degrada-tion by enzymes, cell wall mutants, modi-fications by antibiotics, electron microscopytechniques combined with freeze-fractureand surface replica or negative staining,x-ray photoelectron spectroscopy, infraredspectroscopy, and contact-angle and elec-trophoretic mobility measurements. Thesemethods often require cell manipulation priorto examination (extraction, drying, staining)and in many cases provide averaged infor-mation obtained on a large ensemble ofcells. This indicates that nondestructive, high-resolution tools are needed to probe the micro-bial cell surface.

With atomic force microscopy (AFM)(Binnig et al., 1986), unprecedented possi-bilities are now offered to microscopiststo explore microbial surfaces: imaging thesurface ultrastructure of cell surface lay-ers under physiological conditions and withsubnanometer resolution, monitoring confor-mational changes of individual membraneproteins, examining bacterial biofilms, pro-viding high-resolution images of the sur-face structure of living cells, and followingdynamic cellular processes. But AFM is actu-ally much more than a microscope in thatit also enables forces to be probed quan-titatively. With an AFM, one can measuresurface forces and produce spatially resolvedmaps of the physical properties of the cellsurface, including surface hydrophobicity,

surface charges, adhesion, and elasticity. Itis also possible to measure the elasticityof single surface molecules in relation withfunction. These measurements provide newinsight into the molecular and cellular struc-ture–function relationships of microbial sur-faces (molecular recognition, protein folding,conformational changes, cell adhesion). Theaim of this contribution is to illustrate theseunique capabilities. The chapter is primarilyintended for biologists from various horizons(microbiology, biophysics, biomedicine) anddoes not require advanced understanding ofphysical science principles.

5.2. IMAGING SURFACE STRUCTURE

This section focuses on the use of AFMfor imaging the structure of microbial cellsurfaces. A brief description of appropriatesample preparation procedures and operatingconditions is provided, followed by a dis-cussion of imaging applications dealing withreconstituted cell surface layers and cells.

5.2.1. Methods

5.2.1.1. Substrates. An important require-ment for AFM is that the sample must beattached to a solid substrate. For micro-bial layers, mica, glass, and silicon oxideare the most widely used substrates becausethey are flat and are easy to handle, pre-pare, and clean (Muller et al., 1997b; Coltonet al., 1998). Mica is easily cleaved—forinstance, using an adhesive tape—to pro-duce clean surfaces that are flat over largeareas. Glass coverslips, however, are alwayscoated with organic contaminants and par-ticles that should be removed before use.This can be achieved by washing in con-centrated acidic solution followed by ultra-sonication in several water solutions. Insome cases, best results may be obtainedusing hydrophobic substrates, such as highlyoriented pyrolytic graphite (HOPG). Forsome applications, substrates with well-defined surface chemistries can be designed;

Page 77: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

IMAGING SURFACE STRUCTURE 71

a widely used approach is the functional-ization of gold-coated surfaces with self-assembled monolayers (SAMs) of organicalkanethiols. Finally, as we shall see below,isoporous polymer membranes are valuablesubstrates for immobilizing cells.

5.2.1.2. Immobilization. Three immobili-zation strategies have been developed forAFM of reconstituted surface layers (Pumet al., 1993; Muller et al., 1997a; Coltonet al., 1998): (i) physical adsorption fromaqueous solution in the presence of appro-priate electrolytes which has been employedsuccessfully with purple membranes, OmpFporin and aquaporin crystals; (ii) recrystal-lization on a lipid monolayer, a procedureinitially developed for Bacillus S-layers; (iii)covalent immobilization, which has beenused for the hexagonally packed intermediate(HPI) layer.

Immobilizing native, living cells is oftena challenge. Microbial cells have a well-defined shape and have no tendency to spreadover substrates. As a result, the contact areabetween the cell and substrate is very small,often leading to cell detachment by the scan-ning probe. A second problem is the verticalmotion of the sample (or probe), typicallylimited to a few micrometers, which makesit difficult to image the surface of largeobjects such as microbial cells. To circum-vent these problems, several approaches havebeen proposed, including pretreatments suchas air-drying and chemical fixation to pro-mote cell attachment. These treatments maycause significant rearrangement/denaturationof the surface molecules, thereby compro-mising the biological significance of theresults. An attractive alternative is to per-form cell immobilization by mechanical trap-ping in porous membranes (Kasas and Ikai,1995; Dufrene et al., 1999). This approachoffers two advantages; that is, it is fairlysimple and straightforward and it does notinvolve drying, coating, or chemical fixation.However, an important limitation is that itessentially works for spherical cells (some

bacteria, fungal spores, yeasts) but not forrod-like cells. Figure 5.1 illustrates how thisapproach can be used to immobilize andimage individual cells under aqueous con-ditions. The two height images clearly showmajor differences depending on the cell type:While the yeast Saccharomyces cerevisiae(Fig. 5.1A) showed a very smooth surface,the spore from the fungus Rhizopus oryzaedisplayed a rough surface morphology withlarge fiber-like protrusions (Fig. 5.1B). Thispeculiar surface morphology may play arole in determining the biological functionof fungal spores. Note that the cells aresurrounded by artifactual structures result-ing from the contact between the AFMprobe and the edges of the cell and of thepore.

5.2.1.3. Imaging Conditions. In contactmode, the most widely used imaging mode,sample topography can be measured in twoways: (1) the constant-height mode, in whichthe cantilever deflection is recorded whilethe sample is scanned horizontally, and(2) the constant-deflection mode, in whichthe sample height is adjusted to keep thedeflection of the cantilever constant using afeedback loop. The feedback output is usedto display a true “height image” (Fig. 5.1),while the error signal can also be usedto generate a so-called “deflection image.”Both height and deflection images are oftenuseful when dealing with microbial cells.Height images provide quantitative heightmeasurements, thus allowing an accuratemeasure of cell dimensions and of the surfaceroughness. Deflection images do not reflecttrue height variations, but because theyare more sensitive to fine surface detailsthey are useful for revealing the surfaceultrastructure of curved, rough samples suchas microbial cells.

Selection of an appropriate imaging envi-ronment is critical for high-resolution imag-ing because it controls the force actingbetween probe and sample. When imag-ing in air, the layer of water and contam-inant molecules on both probe and sample

Page 78: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

72 PROPERTIES OF MICROBIAL CELL SURFACES EXAMINED BY ATOMIC FORCE MICROSCOPY

A

B

1 µm

1 µm

Figure 5.1. Immobilization of single microbial cells for AFM imaging under physiological conditions. 3-D AFMheight images, under aqueous solution, showing a yeast cell (Saccharomyces cerevisiae; A) and a fungal spore(Rhizopus oryzae; B) trapped in a porous polymer membrane. The two cells showed very different surfacemorphologies: the yeast surface was very smooth, while the spore displayed a rough surface morphology withlarge fiber-like protrusions.

forms a meniscus pulling the two together.The resulting strong attractive force makeshigh-resolution imaging difficult and some-time causes sample damage. This can beeliminated by performing the imaging inaqueous solution. By selecting appropriatebuffer conditions, it is generally possibleto maintain an applied force in the rangeof 0.1–0.5 nN. For purple membrane andOmpF porin surfaces (Muller et al., 1999b),it was possible to improve the spatial res-olution by changing the pH and electrolyte

concentration, and the best results wereobtained with 150 mM KCl or NaCl (in10 mM Tris-HCl, pH 7.6).

For imaging applications, excellent resultsare obtained using cantilevers made of sili-con (Si) or silicon nitride (Si3N4). Althoughthe probe radii are in the range of 10–50 nm,topographs of flat cell surface layers havebeen acquired routinely with a resolution of1 nm. Nanoscale protrusions extending fromthe probe are thought to be responsible forthis high resolution.

Page 79: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

IMAGING SURFACE STRUCTURE 73

5.2.2. Imaging Cell Surface Layers

Microbial cell surface layers made of two-dimensional (2-D) protein crystals, includingBacillus S-layers, hexagonally packed inter-mediate (HPI) layers, purple membranes,and porin OmpF and aquaporin crystals,have proved to be excellent systems forhigh-resolution AFM imaging. What dowe learn from these images? Structuralinformation can be directly obtained to aresolution of 0.5–1 nm, under physiologicalconditions, which makes AFM a complemen-tary tool to x-ray and electron crystallogra-phy. In addition, the technique can be usedfor modifying single-membrane proteins ormonitoring conformational changes that arerelated to functions.

5.2.2.1. Bacillus S-Layers. ReconstitutedS-layers of Bacillus coagulans E38-66 andBacillus sphaericus CCM2177 imaged byAFM in buffer solution showed obliqueand square lattices, respectively, with latticeparameters in good agreement with transmis-sion electron microscopy data (Ohnesorgeet al., 1992). AFM was used to investi-gate the recrystallization of S-layer proteinsof a Bacillus stearothermophilus strain onuntreated and modified silicon surfaces (Pumand Sleytr, 1995). The oblique lattice of theS-layer was visualized to a lateral resolu-tion of about 1.5 nm. Recrystallization wasshown to occur only at hydrophobic sur-faces, the S-layer being always oriented withits more hydrophobic outer face against theinterface. Both S-layers of B. coagulans E38-66 and B. sphaericus CCM2177 were recrys-tallized on silanized silicon substrates andimaged under liquid with nanometer lateralresolution (Pum and Sleytr, 1996).

5.2.2.2. HPI Layers. Among the variouscell surface layers, the hexagonally packedintermediate (HPI) layer of Deinococcusradiodurans has been the subject of con-siderable research efforts. AFM was usedto image the surface of the HPI layer

in buffer solution with a lateral resolu-tion of 1 nm (Karrasch et al., 1994). Theimages agreed within a few Angstroms withdata obtained by electron microscopy. Morerecent studies have revealed that the innersurface of the HPI layers has a protrudingcore with a central pore exhibiting two con-formations, one with and the other without acentral plug. Individual pores were observedto switch from one state to the other, indi-cating that conformational changes can bedetected at subnanometer resolution (Mulleret al., 1996, 1997b). Remarkably, the AFMcould also be used as a nanotool to inducestructural alterations in the HPI layer (Mulleret al., 1999a). Figure 5.2A shows a high res-olution image of the inner surface of theHPI layer, consisting of hexameric cores withcentral channels that are connected by slen-der arms. As shown in Figure 5.2B, retrac-tion of the AFM stylus resulted in the zippingout of an entire hexameric complex. Thesedata illustrate the power of AFM for imag-ing cell surface layers at high resolution andfor modifying their individual constituents,providing new opportunities for studying themechanical stability of supramolecular struc-tures in relation with function.

5.2.2.3. Purple Membranes. Purplemembranes from the archeon Halobacteriumrepresent another class of well-studied micro-bial layers. These membranes contain bac-teriorhodopsin, a light-driven proton pump,in the form of highly ordered 2-D lat-tices. In 1990, the hexagonal symmetry ofpurple membranes adsorbed on mica wasalready imaged in aqueous conditions, thelattice constant being in agreement withelectron diffraction data (Butt et al., 1990).Later, conditions were established to repro-ducibly acquire topographs of purple mem-branes at subnanometer resolution (Mulleret al., 1995b), and force-induced conforma-tional changes of bacteriorhodopsin weredirectly visualized by AFM (Muller et al.,1995a, 1997b). Upon lowering the imag-ing forces from 300 pN to 100 pN, donut-shaped trimers reversibly transformed into

Page 80: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

74 PROPERTIES OF MICROBIAL CELL SURFACES EXAMINED BY ATOMIC FORCE MICROSCOPY

structures exhibiting three protrusions. Thestandard deviation of the sample height mea-sured for many independent protein units wasshown to provide quantitative information onthe flexibility of the protein domains (Mulleret al., 1998). Tapping mode AFM (TMAFM)is an interesting alternative to contact modebecause it has been shown to faithfullyrecord high-resolution images of purplemembranes and to have sufficient sensitiv-ity to contour individual peptide loops with-out detectable deformations (Moller et al.,1999). Structural changes of purple mem-brane during photobleaching in the pres-ence of hydroxylamine were monitored usingAFM (Moller et al., 2000), providing novelinsights into factors triggering purple mem-brane formation and structure. Crystals ofhalorhodopsin from the overexpressing strainHalobacterium salinarum D2 showed anorthogonal structure and the orientationof the molecules showed p42(1)2 symme-try (Persike et al., 2001). The crystal sur-face was found to display different structuresdepending on the imaging force used, indi-cating that some parts of the molecule weremore rigid but others more compressible.Helix-connecting loops in single molecules

of halorhodopsin were assigned. The imagesindicated that the large extracellular loopcovered the whole molecule and was veryflexible.

5.2.2.4. Porin Crystals. Gram-negativebacteria are protected by an outer mem-brane in which trimeric channels, the porins,facilitate the passage of small solutes. Asshown in Fig. 5.3 (left panels), topographsof porin OmpF crystals of Escherichia colireconstituted in the presence of lipids wererecorded in aqueous solution to a lateral res-olution of 1 nm and a vertical resolutionof 0.1 nm (Schabert et al., 1995). To assessthe accuracy of the AFM topographs, aver-aged surface contours were directly com-pared with the protein structure from x-ray analysis (Fig. 5.3, right panels), withexcellent agreement being obtained betweenthe subtle features of the topographs andthe models. More recently, voltage andpH-dependent conformational changes ofthe extracellular loops were demonstrated(Muller and Engel, 1999). To this end,porin membranes adsorbed to HOPG wereimaged while applying a voltage with aclosely apposed platinum wire. The observed

A B

Figure 5.2. Imaging and manipulating the HPI bacterial surface layer at the molecular level (Muller et al., 1999a).(A) AFM topograph recorded in buffer solution for the inner surface of the HPI layer of Deinococcus radiodurans;the distance between the core centers is 18 nm. (B) The same inner surface area imaged after recording a forcecurve, showing that a molecular defect the size of a hexameric HPI protein complex had been created. (Usedwith permission from Prof. A. Engel, Universitat Basel, Switzerland; copyright 1999, National Academy ofSciences, USA.)

Page 81: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

IMAGING SURFACE STRUCTURE 75

conformational changes were correlated withthe closure of the channel entrance, suggest-ing that this is a mechanism to protect thecells from drastic changes of the environ-ment.

Aquaporins are ubiquitous membranechannels in bacteria, fungi, plants and ani-mals. They are highly specific for wateror small uncharged hydrophilic solutes andare involved in osmoregulation. For E.coli aquaporin Z, AFM images revealed2-D crystals with p42(1)2 and p4 symme-try (Scheuring et al., 1999). Imaging bothcrystal types before and after cleavage ofthe N-termini allowed the cytoplasmic sur-face to be identified. Flexibility mapping andvolume calculations identified the longestloop at the extracellular surface, this loopexhibiting a reversible force-induced confor-mational change.

5.2.3. Imaging Cells

While the potential of AFM for probingliving animal cells was recognized quiteearly (Haberle et al., 1991), attempts to per-form such measurements on microbial cellshave been more limited, due in part to thedifficulties associated with the sample prepa-ration. However, recent studies have demon-strated that AFM can provide a wealth ofinformation on the microbial surface.

5.2.3.1. Microbial Biofilms. The adhe-sion of microorganisms to solid surfacesis a widespread phenomenon that playsa crucial role in the natural environment(self-purification of natural water, symbioticinteractions), in industrial processes (bio-corrosion, biofouling of industrial equip-ments, bioremediation, cells in bioreactors),

A

B

10 nm

Figure 5.3. Visualizing reconstituted porin surfaces at high resolution (Schabert et al., 1995). AFM topographsof the periplasmic (A, left panel) and extracellular (B, left panel) surface of 2-D crystals of the channel formingprotein porin OmpF from Escherichia coli reconstituted in the presence of lipids. Insets: averages calculated from 25translationally aligned subframes. Comparison with the protein structure from x-ray analysis (right panels); zebra-likecontours mark zones of identical altitude, with a height difference between contours of 0.1 nm. (Used with permissionfrom Prof. A. Engel, Universitat Basel, Switzerland; copyright 1995, American Association for the Advancement ofScience.)

Page 82: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

76 PROPERTIES OF MICROBIAL CELL SURFACES EXAMINED BY ATOMIC FORCE MICROSCOPY

and in medicine (accumulation on bioma-terials, infections). Despite their great sig-nificance, the mechanisms of cell adhesionand biofilm formation remain poorly under-stood at the molecular level. In recent years,AFM has helped to shed new light on thesemechanisms.

The instrument has proven useful in bio-corrosion studies. While imaging the topog-raphy of hydrated bacterial biofilms on acopper surface (Bremer et al., 1992), bacte-rial cells were shown to be associated withpits on the copper surface, supporting pre-vious studies related to the pitting corrosionof copper. In order to elucidate the processof stainless steel corrosion, AFM was usedto observe bacteria and bacterial exopoly-mers in their hydrated forms (Steele et al.,1994). AFM was shown to allow estimationof the width and height of bacterial cells,estimation of the thickness and width ofexopolymeric capsule and bacterial flagella,and characterization of substrate roughness,including measurements of depth and diame-ter of individual corrosion pits (Beech et al.,1996). Corrosion, biofilm formation, and theadhesion of different, corrosion-enhancingmicroorganisms to different surfaces (iron,copper, pyrite) were studied in aqueous envi-ronment by AFM (Telegdi et al., 1998).

TMAFM in liquids was used to investi-gate the effect of iron coatings on the interac-tions of Shewanella putrefaciens with silicaglass surfaces (Grantham and Dove, 1996).The results suggested that adhesion, motility,and iron surface chemistry are interrelatedin environments where Fe-reducing microor-ganisms are present. Structures of sizescomparable to large proteins were resolvedon Pseudomonas putida surfaces as wellas flagella trapped within biofilms (Gunninget al., 1996). Biofilms formed by Pseu-domonas sp. on hematite and goethite min-erals were examined in the dried state usingTMAFM (Forsythe et al., 1998).

Unsaturated biofilms of Pseudomonasputida (i.e., biofilms grown in humid air)

were analyzed by AFM to determine sur-face morphology, roughness, and adhesionforces in the outer and basal cell layersof fresh and desiccated biofilms (Auerbachet al., 2000). In sharp contrast to the effectsof drying on biofilms grown in fluid, dryingcaused little change in morphology, rough-ness, or adhesion forces in these unsaturatedbiofilms. The extracellular polymeric sub-stances (EPS) formed “mesostructures” thatwere much larger than the discrete polymersof glycolipids and proteins that have beenpreviously characterized on the outer surfaceof these gram-negative bacteria.

With the aim to gain insight into thesupramolecular organization of bacterial EPS,AFM was used to probe, under aqueousconditions, the nanoscale morphology andmolecular interactions of polystyrene sub-strates after adhesion of the gram-negativebacterium Azospirillum brasilense (van derAa and Dufrene, 2002). After cell adhe-sion under favorable conditions, topographicimages revealed that the substrate surface wascovered by a continuous layer of adsorbedsubstances, ∼2 nm thick, from which supra-molecular aggregates were protruding. Cor-relations were found between the AFM dataand the cell adhesion behavior, providing evi-dence for the involvement of EPS in the adhe-sion process.

5.2.3.2. Cell Surface Nanostructures.Immobilization of single microbial cells—for example, using porous membranes (Fig.5.1)—makes it possible to record high-resolution images of the surface. As anexample, Fig. 5.4C shows a high-resolutionimage under aqueous solution of the surfaceof a spore of the fungus Aspergillus oryzae.The surface was covered with a crystalline-like array of rodlets, 10 ± 1 nm in diame-ter, consistent with electron microscopy data.Chemical analysis and enzymatic digestionhave shown rodlet layers to be essentiallymade of proteins. Regular arrays of rodletsare known to cover the surface of spores ofmany fungi, including ascomycetes, basid-iomycetes, and zygomycetes, and are thought

Page 83: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

IMAGING SURFACE STRUCTURE 77

A

C D

B

germination

10 µm

100 nm 200 nm

Figure 5.4. Imaging cell surface nanostructure: changes upon germination. Optical micrographs of fungal sporesfrom Aspergillus oryzae in the dormant state (A) and after 7 h of incubation in agitated liquid medium (B). AFMdeflection images recorded in aqueous solution for the surface of dormant (C) and germinating (D) spores revealingthat, upon germination, the crystalline rodlet layer changed into a layer of soft material.

to play different biological functions such asspore dissemination and protection.

By contrast, S. cerevisiae cells showed aremarkably smooth (root-mean-square rough-ness ∼1 A over 200-nm × 200-nm areas)and uniform nanoscale surface morphology,consistent with the presence of glucan andmannoproteins (Fig. 5.5A). For bacteria, itis worth emphasizing that high-resolutionimaging in contact mode is often challeng-ing due to sample deformation. By wayof example, high-resolution AFM imagesof Lactococcus lactis bacteria showed asponge-like network of small holes together

with grooves attributed to strong probe–cellinteractions (Boonaert et al., 2002). In futureresearch, the use of dynamic imaging modesmay help to circumvent this kind of problems.

Appendages (fimbriae, fibrils, flagella)belong to a class of surface structuresoften found on bacterial cells which mayplay various important functions includingcell adhesion, motility, and conjugation.Electron microscopy is a well-establishedtechnique to visualize appendages at highresolution. AFM may bring complementaryinformation by allowing direct observationin the hydrated state. For instance, fibrillar

Page 84: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

78 PROPERTIES OF MICROBIAL CELL SURFACES EXAMINED BY ATOMIC FORCE MICROSCOPY

50 nm

2 nm

0 nm

8 nm

0 nm

+ Cu2+

A B

Figure 5.5. Imaging cell surface nanostructure: Influence of external agents. High-resolution AFM height imagesrecorded in 1 mM KNO3 solution for the surface of the yeast Saccharomyces cerevisiae prior to (A) and after(B) exposure to copper (10 mM Cu2+, 10 min). Clearly, addition of copper caused a significant increase of surfaceroughness, from 1 A to 7 A. Note that the AFM probe was functionalized with COOH groups.

structures were detected on the cell surfaceof Streptococcus salivarius HB, while S.salivarius HBC12 appeared to have a baldcell surface, in agreement with electronmicroscopy data (van der Mei et al., 2000).However, due to the mobility and softnessof these structures, the image quality waspoor, emphasizing the need to use alternativepreparation procedures (isolation, drying)and imaging modes (dynamic modes).

5.2.3.3. Physiological Changes. One ap-plication of particular interest is the ability tostudy, under native conditions, the changes ofcell surface structure associated with physi-ological processes. An example of such pro-cess is the germination of fungal spores. Asillustrated in Fig. 5.4A and 5.4B, incubat-ing A. oryzae spores in liquid medium fora few hours lead to their germination and,in turn, to their aggregation, a process thatis of both natural and biotechnological sig-nificance. High-resolution images (Fig. 5.4Cand 5.4D) showed that dramatic changes ofthe spore surface ultrastructure occurred upongermination, with the rodlet layer changinginto a layer of soft material (van der Aaet al., 2001). On close examination, this mate-rial showed streaks oriented in the scanning

direction, suggesting that soft, loosely boundmaterial was interacting strongly with thescanning probe. These direct observations arein good agreement with previous structuraland chemical studies which showed that ger-mination of Aspergillus spores results in thedisruption of the proteinaceous rodlet layerand reveals inner spore walls which are essen-tially composed of polysaccharides.

Another example is the growth of yeastscells. Using a soft, deformable immobiliza-tion matrix made of agar gel, the growth pro-cesses of S. cerevisiae cells were investigatedin situ and micrometer-size bud scars result-ing from the detachment of budding daughtercells were visualized (Gad and Ikai, 1995).

5.2.3.4. Effect of External Agents. Thepossibility to directly visualize the effectof external agents such as solvents, ions,chemicals, enzymes, and antibiotics on thestructure of microbial cells opens the doorto new applications in biotechnology andbiomedicine—for example, for the rapidscreening of microorganisms and for therationale design of new antimicrobial drugs.The action of penicillin on the morpholgyof Bacillus subtilis cells was investigated by

Page 85: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

MEASURING PHYSICAL PROPERTIES 79

AFM in air (Kasas et al., 1994). Bacteriagrown in the presence of the antibioticshowed dramatic changes: Modification incell length was first noted, followed byrelease of material into the environment.Changes in the morphology of E. coliinduced by cefodizime were demonstratedusing AFM in air (Braga and Ricci, 1998).Holes and rough patches were observedat the cell surface after treatment withsuprainhibitory concentrations of cefodiz-ime, while filamentation was induced bysubinhibitory concentrations. In the sameway, morphological alterations in Helicobac-ter pylori were visualized after exposure torokitamycin (Braga and Ricci, 2000). Vari-ous changes were noted (i.e., cell enlarge-ment, cell clustering, increased roughness),depending on the antibiotic concentration.The effect of vancomycin, a glycopeptideantibiotic that kills gram-positive bacteriaby interfering with the synthesis of peptido-glycan, was also investigated (Boyle-Vavraet al., 2000). The fine structural detail pro-vided by AFM extended previous topo-graphic studies of vancomycin-susceptibleand vancomycin-resistant Staphylococcusaureus by showing novel morphologicalchanges occurring in the cell surface.

The effect of the denaturant LiCl onthe surface of Lactobacillus helveticus cellswas investigated by AFM (Sokolov et al.,1996). While images with a lateral reso-lution of 2 nm were recorded on nativecells, exposure to LiCl changed the sur-face morphology into a pattern of holes.The influence of various chemicals (Tween20, heparin, disodium tetraborate, sodiumpyrophosphate, lysozyme, and EDTA) onthe surface topography of bacterial cellswas investigated using TMAFM (Camesanoet al., 2000); most treatments were found toincrease the surface roughness, and a changeof cell shape was often noted.

Bacteria, fungi and yeasts are capableof removing heavy metals from aqueoussolution in substantial amounts. Conse-quently, microbial biomass represents a

new, attractive technology for the treatmentof wastewaters (Kapoor and Viraraghavan,1995). Despite the vast amount of litera-ture available on biosorption processes, lit-tle is known at the molecular level dueto the lack of appropriate tools. Figure 5.5shows that the nanoscale morphology of S.cerevisiae cells changed dramatically uponaddition of copper (10 mM for 10 min),the surface roughness increasing from 1 Ato 7 A (root-mean-square roughness over200-nm × 200-nm areas). Copper bindingwas further evidenced by force measure-ments, with significant adhesion being mea-sured between treated cells and negativelycharged probes. These data are consistentwith biosorption studies showing that S. cere-visiae cells remove copper by a two-stepprocess consisting of an initial rapid surfacebinding of copper ions, followed by a slowerintracellular uptake (Kapoor and Viraragha-van, 1995).

Accordingly, these experiments demon-strate that AFM has a great potential inbiotechnology and biomedicine studies toprobe the interactions between cell surfacesand external agents at the molecular level. Aunique advantage over classical microscopiesis that subtle changes can be directly visual-ized at the cell surface without any pretreat-ment (drying, fixation).

5.3. MEASURING PHYSICALPROPERTIES

A key advantage of AFM over classicalmicroscopies is that it can measure forcesand produce quantitative, spatially resolvedmaps of physical properties. Furthermore, thevery high sensitivity of force measurementsmakes it possible to stretch single moleculesto learn about their elastic behavior and con-formational transitions. This section intro-duces the principles and interpretation offorce measurements by AFM and their appli-cations to microbial cell surfaces.

Page 86: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

80 PROPERTIES OF MICROBIAL CELL SURFACES EXAMINED BY ATOMIC FORCE MICROSCOPY

5.3.1. Principles of ForceMeasurements

5.3.1.1. Force–Distance Curves. Howdoes AFM measure forces? By monitoring,at a given x, y location, the cantilever deflec-tion, expressed as a voltage, as a function ofthe vertical displacement of the piezoelectricscanner (z), a raw force–distance curve isconstructed (Butt et al., 1995). Using theslope of the retraction force curve in theregion where probe and sample are in con-tact, the photodiode voltage can be convertedinto a cantilever deflection (d). The can-tilever deflection is then converted into aforce (F ) using Hooke’s law: F = −k × d ,where k is the cantilever spring constant. Thecurve can be corrected by plotting F as afunction of (z − d). The zero separation dis-tance is then determined as the position of thevertical linear parts of the curve in the con-tact region. In addition to single force curves,spatially resolved mapping can be performedby recording arrays of force curves in thex, y plane (Heinz and Hoh, 1999).

The different parts of a force–distancecurve can provide a wealth of informa-tion (Butt et al., 1995; Dufrene et al., 2001).At large probe–sample separation distances,the force experienced by the probe is null.As the probe approaches the surface, thecantilever may bend upwards due to repul-sive forces until it jumps into contact whenthe gradient of attractive forces exceeds thespring constant plus the gradient of repul-sive forces. The approach portion of theforce–distance curve can be used to mea-sure surface forces, including van der Waalsand electrostatic, solvation, hydration andsteric/bridging forces. When the force isincreased in the contact region, the shapeof the approach curve may provide directinformation on the elasticity of the sample.Upon retracting the probe from the surface,the curve often shows a hysteresis referredto as the adhesion “pull-off” force, whichcan be used to estimate the surface energyof solids or the binding forces between

complementary biomolecules. In the pres-ence of long, flexible molecules, an attrac-tive force, referred to as an elongation force,may develop nonlinearly due to macromolec-ular stretching. In this case, it is common topresent the force curve as the positive pullingforce vs extension.

There are several limitations to keep inmind when dealing with AFM force–distancecurves. First, as opposed to the surface-forcesapparatus, AFM does not provide an inde-pendent measure of the probe–sample sepa-ration distance. As a result, in the presenceof long-range surface forces and high sam-ple elasticity, defining the point of contact(i.e., the zero separation) is difficult. Second,for soft samples such as many microbialsurfaces, separating the relative contribu-tions of repulsive surfaces forces and sampledeformation is a delicate task, and quanti-tative interpretation of the curves—using,for example, the DLVO theory or the Hertzmodel—may be meaningless. Third, arte-facts are frequently encountered (Heinz andHoh, 1999): (a) periodic oscillations in thenon-contact region (optical interference) and(b) offsets between the lines in the contact(nonlinearities of the piezo, friction) and non-contact (viscous drag) regions.

5.3.1.2. Functionalized AFM Probes.Another issue is the control of the probesurface chemistry, which is a prerequisitefor reliable, quantitative force measurements.Most commercial AFM probes have a poorlydefined surface chemistry: The surface of sil-icon oxynitride probes is composed of ioniz-able silanol and silylamine groups that varyin density with pH, which is further compli-cated by the fact that microfabrication resultsin contamination by gold and other mate-rials. To overcome this problem, differentapproaches have been developed to createprobes of well-defined chemistry. Amongthese, functionalizing sharp AFM probeswith self-assembled monolayers (SAM) oforganic alkanethiols combines the advan-tage of strong chemical sensitivity with

Page 87: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

MEASURING PHYSICAL PROPERTIES 81

high spatial resolution (Frisbie et al., 1994;Dufrene, 2000). The procedure involvescoating microfabricated cantilevers with athin adhesive layer (Cr or Ti), followed by a15- to 100-nm-thick Au layer and immersingthe coated cantilevers in dilute (0.1–1 mM)ethanol solutions of the selected alkanethiol.As we shall see, thiols with different terminalfunctionalities (e.g., OH, CH3, COOH, NH2)can be used to map the physicochemicalproperties of microbial surfaces.

Different approaches are also available toattach biomolecules on the AFM probe: non-specific adsorption (Lee et al., 1994b), cova-lent linkage on aminosilane-functionalizedsilica surfaces (Lee et al., 1994a), bindingof proteins onto carboxyl functions using1-ethyl-3-(3-dimethylaminopropyl)carbodii-mide (EDC) and N -hydroxysuccinimide(NHS) (Benoit et al., 2000), and graft-ing of antibodies using polyethylene gly-col derivatives with an amine-reactiveend and a thiol-reactive end (Hinterdorferet al., 1996). Because they allow measur-ing intermolecular forces between individ-ual ligand-receptors, these approaches have atremendous potential for molecular mappingof microbial surfaces.

5.3.2. Physicochemical Properties andMolecular Interactions

Physicochemical properties (surface energy,surface charges) and molecular interactions(solvation forces, van der Waals and electro-static forces, and steric/bridging forces) arehighly relevant to many cellular processes(microbial adhesion and aggregation, molec-ular recognition). Clearly, there is consid-erable challenge in using AFM to probedirectly these properties at high spatialresolution.

5.3.2.1. Adhesion Mapping. Force–vol-ume imaging consists in recording arraysof force curves in the x, y plane (Heinzand Hoh, 1999). Such spatially resolvedforce measurements are very useful tomap physical heterogeneities at cell sur-faces. An example demonstrating the powerof this approach in microbiology is givenin Fig. 5.6. It can be seen that aftergermination, the spores from the fungusPhanerochaete chrysosporium showed a het-erogeneous surface morphology made ofsmooth zones partially covered with roughgranular structures (Fig. 5.6A). This wasdirectly correlated with differences in the

A B

500 nm

100 nm

0 nm

10 nN

0 nN

Figure 5.6. Spatially resolved force measurements. AFM height image (A) and adhesion map (B) acquired with asilicon nitride probe on the same area of a germinating spore of Phanerochaete chrysosporium. The adhesion mapwas obtained by recording 64 × 64 force–distance curves, calculating the adhesion force for each force curve anddisplaying adhesion force values as gray levels. The smoother area in the center of the image showed strong adhesionforces, thought to be responsible for spore aggregation.

Page 88: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

82 PROPERTIES OF MICROBIAL CELL SURFACES EXAMINED BY ATOMIC FORCE MICROSCOPY

adhesion map (Fig. 5.6B): While no adhesionforces were sensed between the siliconnitride probe and the granular material,strong adhesion forces of 9 ± 2 nN mag-nitude were measured on the smooth zone(Dufrene, 2001). The measured adhesionforces may be of great biological significancein that they may be responsible for cellaggregation observed during germination.However, the origin of the forces is uncleardue to the poorly defined probe chemistry.As discussed below, probes functionalizedwith well-defined chemical groups can pro-vide more detailed information about cellsurface properties and molecular interactions.

5.3.2.2. Surface Energy and SolvationInteractions. During the past decades,microbial cell surface hydrophobicity (surfaceenergy) has attracted considerable attentionin view of its role in microbial adhesionprocesses (Doyle and Rosenberg, 1990). Avariety of experimental methods (van derMei et al., 1991) have been developed to

assess microbial hydrophobicity, includingwater contact-angle measurements on celllawns, adhesion to hydrocarbons, partitioningin aqueous two-phase systems, and hydropho-bic interaction chromatography.

AFM with chemically functionalizedprobes can yield high-resolution informationon surface hydrophobicity (surface energy)directly under physiological conditions,which makes it a complementary tool tothese approaches (Dufrene, 2000). UsingOH- and CH3-terminated probes, patterns ofrodlets, ∼10 nm in diameter, were visual-ized, under physiological conditions, at thesurface of dormant spores of P. chrysospo-rium, demonstrating that high resolutioncan be obtained with the modified probes(Fig. 5.7A). Multiple (1024) force–distancecurves recorded over 500-nm × 500-nmareas at the spore surface, either indeionized water or in 0.1 M NaCl solu-tions, always showed no adhesion for bothOH- and CH3-terminated probes (Fig. 5.7B).

A B

100 nm

CH3

CH3CH3 CH3

CH3

CH3

CH31

0

1000

500

0

−1

−20

0 5 10

50 100

Surface separation (nm)

Adhesion force (nN)

For

ce (

nN)

# ev

ents

150 200

Figure 5.7. Mapping cell surface hydrophobicity (surface energy) using chemically functionalized probes. AFMdeflection image (A) and force–distance curve and adhesion force histogram (B) obtained, under aqueous solution,for the surface of P. chrysosporium spores with a hydrophobic, CH3-terminated probe. The adhesion force histogramwas generated by recording 1024 force–distance curves over 500-nm × 500-nm areas; the narrow distribution showsthat the surface was chemically homogeneous. Similar results were obtained with both CH3- and OH-terminatedprobes, indicating that the spore surface was hydrophilic.

Page 89: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

MEASURING PHYSICAL PROPERTIES 83

Comparison with adhesion data obtained onfunctionalized model substrates prior to andafter the cell measurements led to the con-clusion that the spore surface is uniformlyhydrophilic. The highly hydrophilic, nonad-hesive character of the fungal spore surfacemay play an important role in determiningthe spore biological functions, namely, pro-tection and dispersion.

5.3.2.3. Surface Charges and Electro-static Interactions. Force–distance curvescan also be used to probe surface chargesand electrostatic forces at microbial cellsurfaces. A number of studies have con-centrated on microbial layers. In an earlywork, electrostatic forces were measured atdifferent pH values between standard sili-con nitride probes and the surface of pur-ple membranes (Butt, 1992). By comparingthe approach force curves measured on bothpurple membranes and bare alumina, thesurface charge density of the former wasestimated to be −0.05 C/m2. The interac-tion forces between a silica sphere and two-dimensional bacteriorhodopsin crystals werealso measured at different pH values andat two electrolyte concentrations (Hartleyet al., 1998). The surface potential of thecrystal surface could be determined, andthe isoelectric point was found to be about4. Force–distance curves were recorded, atdifferent electrolyte concentrations, betweensilicon nitride probes and the surface ofpurple membranes, OmpF porins, and HPIlayers (Muller et al., 1999b). In the pres-ence of 20 mM KCl, long-range repulsionforces were measured for the surface of pur-ple membranes and OmpF porins, reflect-ing the electrostatic double-layer repulsion.These forces could be adjusted by chang-ing the pH or electrolyte concentration tooptimize the image resolution. No repulsionforce was found for the HPI layers, indi-cating that the surface charge of the lat-ter was either too small to be detected orpositive.

For living microbial cells, the pres-ence of ionizable surface groups such as

amino, carboxyl, and phosphate is classi-cally established by microelectrophoresis,titration, ion-exchange chromatography, andsurface conductivity (James, 1991). With itshigh spatial resolution, AFM with chem-ically functionalized probes appears as apowerful, complementary approach to theseglobal methods. Figure 5.8 illustrates howthe presence of ionizable groups and thelocal isoelectric point of microbial cell sur-faces can be determined using this approach.The force curves recorded between anAFM probe functionalized with COOHgroups and the surface of S. cerevisiae cellsshowed tremendous changes according to pH(Fig. 5.8A): While no adhesion was observedat pH 10, multiple adhesion forces wereobserved at pH 4. Force mapping revealedsimilar results at different locations of thecell surface, indicating that the latter wasfairly homogeneous as far as electrical prop-erties are concerned. High-resolution imag-ing was used to confirm that the surfacenanostructure was not affected by pH varia-tions, with smooth surfaces being observed atboth pH values (Fig. 5.5A). Adhesion forcemeasurements as a function of pH, referredto as “nanotitration” curves, exhibited a sharpincrease in the adhesion force at pH valuesaround 4, with the transition occurring within2 pH units (Fig. 5.8B).

The adhesion observed at low pH canbe attributed to hydrogen bonding betweenthe uncharged COOH probe and neutral cellsurface macromolecules, while the lack ofadhesion at pH values >6 would essen-tially reflect electrostatic repulsion betweenthe negatively charged surfaces. The positionof the maximum of the nanotitration curveat pH ∼ 4 was very close to the isoelec-tric point of the cell surface measured bymicroelectrophoresis measurements. Indeed,the mobility–pH curve obtained previouslyfor this strain was consistent with the pres-ence of an amino-carboxyl surface (Dengiset al., 1995): At pH values smaller than4, the isoelectric point, the surface waspositively charged due to the presence of

Page 90: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

84 PROPERTIES OF MICROBIAL CELL SURFACES EXAMINED BY ATOMIC FORCE MICROSCOPY

A

B

pH 10

pH 41 nN

For

ceA

dhes

ion

forc

e (n

N)

0

2

1.5

0.5

00 2 4 6 8 10 12

1

100 200 300 400

Surface separation (nm)

pH

COO−

COO−

COO−

COOHCOOH

COOHCOOH

COOHCOOHCOOH

−− − − − − − −

COO−COO−COO−COO−

Figure 5.8. Nanotitration of cell surface ionizable functional groups using chemically functionalized probes.Force–distance curves (A) and adhesion force–pH curve (B) obtained for the surface of S. cerevisiae cells withCOOH-terminated probes (1 mM KNO3 solutions; pH adjusted with KOH and HNO3). Multiple force measurementsperformed at different locations of the cell surface showed only small variations, indicating that the surface propertieswere fairly homogeneous. The adhesion maximum at pH 4 was correlated with the isoelectric point of the cell surface.Note that following treatment of the cell surface with Cu2+ ions, significant adhesion forces were measured at neutralpH values, supporting the notion that surface charges were actually probed.

NH3+ groups; above pH 4, an increase

in the negative value of the mobility wasnoted due to dissociation of the weakCOOH groups, until a plateau value wasreached at about pH 6. The nanotitrationcurve was fully consistent with these micro-scopic data, indicating that functionalized

probes represent a powerful approach to maplocal ionizable functional groups and isoelec-tric points.

The question why a significant repulsionwas observed upon approach even at the cellsurface isoelectric point may be raised. Pre-sumably, this reflected the contribution of

Page 91: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

MEASURING PHYSICAL PROPERTIES 85

nonelectrostatic interactions—that is, stericrepulsion associated with hydrated cell surfacepolymers (glucan and mannoproteins for yeastcells). The relative contributions of elec-trostatic and steric interactions associatedwith negatively charged bacterial strains wererecently investigated (Camesano and Logan,2000). The forces measured with siliconnitride probes as a function of pH andionic strength were represented well by anelectrosteric repulsion model but were muchlarger in magnitude and extended over longerdistances than predicted by DLVO theory.Partially removing polysaccharides from thebacterial surfaces resulted in lower repulsiveforces that decayed much more rapidly.

5.3.2.4. The ‘‘Cell Probe’’ Approach. Analternative approach to probe molecular inter-actions associated with microbial cells is toimmobilize them on the AFM probe and mea-sure the forces between the modified probesand solid substrates. In this way, the forcesbetween E. coli-coated probes and solids ofdifferent surface hydrophobicity were mea-sured (Ong et al., 1999). Both attractive forcesand cell adhesion behavior were promotedby substrate surface hydrophobicity, pointingto the role of hydrophobic interactions. Cell-coated probes were used to measure the forcesbetween E. coli strains and polymer bioma-terials (Razatos et al., 1998) and substratescoated with block copolymers (Razatos et al.,2001). Polymeric brush layers appeared to notonly block the long-range attractive forcesof interaction between bacteria and substratesbut also introduce repulsive steric effects.The adhesion of metabolically active Sac-charomyces cerevisiae cells was quantified ata hydrophilic mica surface, a mica surfacewith a hydrophobic coating, and a protein-coated mica surface in an aqueous envi-ronment (Bowen et al., 2001). Greatest celladhesion was measured at the hydrophobicsurface. Prior adsorption of a bovine serumalbumin protein layer at the hydrophilic sur-face did not significantly affect cell adhesion.Changes in yeast surface hydrophobicity and

zeta potential with yeast cell age were corre-lated with differences in adhesion.

5.3.3. Cell Surface Elasticity

How stiff are cell wall components andsurface appendages? This highly relevantquestion can, to a certain extent, be addressedon a local scale with AFM. Note, however,that in view of the complex nature ofmicrobial cell surfaces and of the limitationsof force curves, distinguishing true sampledeformation from repulsive surface forcescan be a delicate task, especially whenworking in aqueous solution.

The mechanical properties of gram-negative magnetic bacteria of the speciesMagnetospirillum gryphiswaldense were in-vestigated by AFM in buffer solution (Arnoldiet al., 1998). Multiple force–distance curvesobtained for the substrate and for the bacte-ria made it possible to determine the effectivecompressibility of the cell wall, which wasabout 42 mN/m. For the same species, a the-oretical expression was derived for the forceexerted by the wall on the cantilever as a func-tion of the depths of indentation generated bythe AFM tip (Arnoldi et al., 2000). Evidencewas provided that this reaction force is a mea-sure for the turgor pressure of the bacterium.Making use of experimental and of theoreticalresults, the turgor pressure was determined tobe in the range of 85–150 kPa.

The elastic properties of isolated bacte-rial cell wall components can also be probedusing a so-called “depression technique” (Xuet al., 1996; Yao et al., 1999). A solutioncontaining the cell wall components (e.g.,archeal sheaths, peptidoglycan sacculi) isplaced on a hard substrate and allowed todry. The substrate contains grooves that arenarrow compared to the width or the lengthof the material to be investigated so that sin-gle wall components bridging one or moregrooves can be obtained. The force measure-ment consists in placing the probe at themidpoint of the unsuspended wall componentregion and increasing/decreasing the force as

Page 92: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

86 PROPERTIES OF MICROBIAL CELL SURFACES EXAMINED BY ATOMIC FORCE MICROSCOPY

0 25 50 75

Separation distance (nm)

For

ce (

nN)

100 125 150

1.5

1

0.5

0 HBC12

HB

HB

HBC12

Figure 5.9. Probing the surface softness of bacterial cells. Approach force–distance curves recorded under waterbetween a silicon nitride probe and Streptococcus salivarius HB or S. salivarius HBC12. The long-range repulsionon the fibrillated strain was attributed to the compression of a soft layer of appendages (fibrils) present at the cellsurface. In view of the complexity of the surfaces and difficulty to define the point of contact, no attempt was madeto fit the curves with models from contact mechanics.

a linear function of time. Comparison of thecantilever deflections between specimen andhard substrate allows an accurate determi-nation of the specimen elasticity. Applyingthis approach to the proteinaceous sheath ofthe archeon Methanospirillum hungatei GP1yielded an elastic modulus of 2–4 × 1010

N/m2, indicating the sheath could withstandan internal pressure of 400 atm, well beyondthat needed for an eubacterial envelope towithstand turgor pressure. For murein sac-culi of gram-negative bacteria in the hydratedstate, elastic moduli of 2.5 × 107 N/m2 weremeasured, in excellent agreement with theo-retical calculation of the elasticity of the pep-tidoglycan network (Boulbitch et al., 2000).

Surface appendages may cause a signif-icant increase of cell surface softness, asillustrated in Fig. 5.9, which presents theapproach curves recorded on the fibrillatedS. salivarius HB bacterial strain and on thebald S. salivarius HBC12 strain (van der Meiet al., 2000). Upon approach of S. salivar-ius HB, a long-range (∼100 nm) repulsionforce decaying exponentially with distancewas detected. By contrast, the repulsion forcefound for S. salivarius HBC12 was of muchshorter range (∼20 nm). In agreement withthe topographic images, the occurrence of along-range repulsion on the fibrillated strain

was attributed to the compression of the softlayer of fibrils present at the cell surface. Infuture experiments, it would be interestingto establish the role that cell surface softnessmay play in controlling cellular events suchas cell growth and cell adhesion.

5.3.4. Elasticity of SingleMacromolecules

When pulling an AFM probe away from amacromolecular system, the retraction curveoften shows attractive elongation forcesdeveloping nonlinearly, which reflect thestretching of the macromolecules. The highforce resolution and positional precision ofAFM allows measurements at the single-molecule level. In recent years, these “single-molecule force spectroscopy” experimentshave tremendously improved our currentunderstanding of the nanomechanical proper-ties of single biomolecules, including DNA,proteins and polysaccharides (for reviewssee Clausen-Schaumann et al., 2000; Jan-shoff et al., 2000).

5.3.4.1. Theoretical Models. When stret-ching long, flexible polymers, two restoringforces may occur: At small displacements,reduction in the number of conformations

Page 93: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

MEASURING PHYSICAL PROPERTIES 87

gives rise to entropic elasticity forces,while at large extensions, tension in themolecular backbone may lead to enthalpicelasticity effects (bond deformation, rup-ture of intramolecular hydrogen bonds, andeven conformational changes of the entiremolecule).

Two models from statistical mechanics areoften used to analyze quantitatively elon-gation forces: the worm-like chain (WLC)and the freely jointed chain (FJC) mod-els (Janshoff et al., 2000). The WLC modeldescribes the polymer as an irregular curvedfilament, which is linear on the scale of thepersistence length, a parameter that repre-sents the stiffness of the molecule. Hence,molecules with low persistence length havea tendency to form coils. Entropic andenthalpic contributions are combined, butextension is limited by the contour lengthof the molecule—that is, the length of thelinearly extended molecule without stretch-ing the molecular backbone (for relevantequations see Janshoff et al., 2000). TheWLC model has been successfully appliedto describe the stretching of single DNA andprotein molecules (Oesterhelt et al., 2000).

In the FJC model, the polymer hasa purely entropic elastic response and isconsidered as consisting of a series ofrigid, orientationally independent statistical(Kuhn) segments, connected through flexiblejoints. The segment length, referred to asthe Kuhn length, is a direct measure of thechain stiffness. An extended FJC model hasbeen developed in which Kuhn segmentscan stretch and align under force. Thepolymer consists of a series of elasticsprings characterized by a given segmentelasticity. The extended FJC model has beensuccessfully applied to describe the elasticbehavior of polysaccharides, amylose anddextran, as probed by single-molecule forcespectroscopy (Rief et al., 1997; Marszaleket al., 1998).

5.3.4.2. Application to Cell SurfaceLayers. Using combined AFM imaging and

force spectroscopy, bacterial proteins fromthe HPI layer of D. radiodurans wereunzipped (Muller et al., 1999a). Force–extension curves recorded for the innersurface of the HPI layer showed saw-toothpatterns with six force peaks of about 300 pN(Fig. 5.10). This behavior, well-fitted with aWLC model, was attributed to the sequentialpulling out of the protomers of the hexam-eric HPI protein complex. After recording theforce curve, a molecular defect the size of ahexameric complex was clearly visualized bymeans of high-resolution imaging (Fig. 5.2).Hence, AFM allows to correlate force mea-surements with resulting structural changes.

In another study combining AFM imagingand force spectroscopy, the unfoldingpathways of individual bacteriorhodopsinswere unraveled (Oesterhelt et al., 2000).Molecules were individually extracted fromthe membrane, with anchoring forces of100–200 pN being found for the differenthelices. Upon retraction, the helices werefound to unfold and the force spectrarevealed the individuality of the unfoldingpathways.

5.3.4.3. Application to Cells. Stretchingsingle molecules directly on living cells is verychallenging due to the complex and dynamicnature of the surface macromolecules. How-ever, recent data indicate that progresses havebeen made in this direction.

Mannan polymers were stretched at thesurface of S. cerevisiae cells using probesthat were functionalized with the lectinconcanavalin A (Gad et al., 1997). Rup-ture lengths up to several hundred nanome-ters were recorded, and the distribution ofthe polymers was mapped using force vol-ume imaging. No attempt was made tofit the curves with a statistical mechanicsmodel. The forces between living Shewanellaoneidensis bacteria and goethite, both com-monly found in Earth near-surface environ-ments, were measured quantitatively (Loweret al., 2001). Energy values derived fromthese measurements showed that the affinity

Page 94: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

88 PROPERTIES OF MICROBIAL CELL SURFACES EXAMINED BY ATOMIC FORCE MICROSCOPY

0 20 40 60 80

For

ce

Extension [nm]

300

pN

Figure 5.10. Using force spectroscopy to unzip single bacterial surface proteins (Muller et al., 1999a).Force–extension curve recorded for the inner surface region of the HPI layer showing a saw-tooth pattern withsix force peaks of about 300 pN. The curves were well-fitted with a WLC model and compared favorably to those oftitin molecules. As can be seen in Fig. 5.2B, the inner surface area imaged after recording the force curve showed amolecular defect the size of a hexameric HPI protein complex. (Used with permission of Prof. A. Engel, UniversitatBasel, Switzerland; copyright 1999, National Academy of Sciences, USA.)

between S. oneidensis and goethite rapidlyincreases by two to five times under anaer-obic conditions in which electron transferfrom bacterium to mineral is expected. Spe-cific signatures in the force–extension curveswere well-fitted with a WLC model andsuggested to reflect the unfolding of a 150-kDa putative iron reductase.

AFM imaging and force spectroscopywere applied to A oryzae spores with theaim to gain insight into cellular struc-ture–function relationships (van der Aaet al., 2001). Germination of A. oryzaespores in liquid medium leads to sporeaggregation (Fig. 5.4A and 5.4B), a pro-cess which is still poorly understood atthe molecular level. Topographic imagingrevealed that, upon germination, the sporesurface changed into a layer of soft granularmaterial attributed to cell surface polysac-charides (Fig. 5.4C and 5.4D). As can beseen in Fig. 5.11, force–extension curvesrecorded on this surface showed attractiveforces of 400 ± 100 pN magnitude, alongwith characteristic elongation forces and rup-ture lengths ranging from 20 to 500 nm.Elongation forces were well-fitted with an

extended FJC model, using fitting parame-ters that were consistent with the stretch-ing of individual dextran molecules. Thissupports the idea that polysaccharides wereactually stretched. Whether single moleculeswere addressed remains unclear. However,given the size of the AFM probe and thewide range of observed contour lengths,it seems unlikely that a large number ofpolysaccharide molecules would adsorb atthe same time on the probe with thesame contour length so that they wouldgive a single, discrete elongation eventupon extension. The macromolecular adhe-sion and elasticity measured for the ger-minating spores may play a key role inthe aggregation process; the sticky and flex-ible nature of long macromolecules, pre-sumably polysaccharides, may indeed pro-mote bridging interactions between spores.In future cell studies, developments willclearly be needed to establish unambiguouslythe exact biochemical nature of the macro-molecules that are being stretched (specificbinding using functionalized AFM probes,mutants lacking specific cell surface macro-molecules).

Page 95: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

CONCLUSIONS AND FUTURE PROSPECTS 89

200 300 400

Extension (nm)

For

ce

500 600

100 pN

Figure 5.11. Stretching cell surface macromolecules. Typical force–extension curve recorded on a germinating sporeof A. oryzae. The elongation force was well-fitted with an extended FJC model (continuous line), with parameterssimilar to values reported for the elastic deformation of single dextran and amylose polysaccharides.

5.4. CONCLUSIONS AND FUTUREPROSPECTS

The data reviewed in this chapter demonstratethat AFM is becoming a key technique inmicrobiology, biophysics, and biomedicine toprobe the structural and physical properties ofmicrobial surfaces. The structure of microbialcell surface layers made of 2-D proteincrystals, including Bacillus S-layers, HPIlayers, purple membranes, and porin OmpFand aquaporin crystals, can be routinelyimaged at a lateral resolution of 0.5–1 nm.Remarkably, conformational changes inducedby a physiological signal can be monitoredin single-membrane proteins providing novelinsight into structure–function relationships.Although high-resolution imaging of livingcells remains challenging, a wealth of novelstructural information can be obtained usingAFM: observing hydrated microbial biofilmsin relation with natural or industrial issues,visualizing surface ultrastructure (rodlets,appendages), following physiological changes(germination, growth), and monitoring theeffect of external agents (antibiotics, met-als). These studies open the door to newpractical applications in biotechnology andbiomedicine, such as the rapid detection

of microorganisms and the rationale designof drugs.

Force–distance curves can be combinedwith high-resolution imaging to map thephysical properties of microbial surfacesunder native conditions, thereby helping toelucidate molecular and cellular functions.Provided that the experiments are carefullyconducted (appropriate sample preparationprocedure, probes of well-defined surfacechemistry, control experiments, validationby independent techniques), force–distancecurves allow one to map molecular interac-tions (solvation forces, electrostatic interac-tions) and physicochemical properties(surface energy, surface charges), to measurethe stiffness of cell wall components and sur-face appendages and to probe the elasticityof single macromolecules.

There are many open challenges for futureresearch. Progress in sample preparationtechniques, instrumentation, and recordingconditions for living cells is still neededto obtain surface topographs of living cellswith subnanometer resolution and to monitorsurface molecular conformational changes.One may anticipate that the development ofnew instruments, such as the cryogenic AFMoperating in liquid nitrogen vapor, will reveal

Page 96: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

90 PROPERTIES OF MICROBIAL CELL SURFACES EXAMINED BY ATOMIC FORCE MICROSCOPY

new structural details on soft microbialspecimens with unprecedented resolution.Another issue is the mapping of specificinteraction forces associated with antibod-ies, lectins, and adhesins, which are involvedin many important functions. Mechanicalproperty measurements on microbial cellwalls open the exciting prospect of measur-ing the turgor pressure in living cells, whichwill provide a better understanding of micro-bial growth and division processes. Single-molecule force spectroscopy on living cellswill make it possible to probe conformationaltransitions as well as the forces required tounfold proteins and to dissociate supramolec-ular assemblages.

REFERENCES

Arnoldi, M., Kacher, C. M., Bauerlein, E., Rad-macher, M., and Fritz, M. (1998). Elastic prop-erties of the cell wall of Magnetospirillumgryphiswaldense investigated by atomic forcemicroscopy. Appl. Phys. A 66:S613–S617.

Arnoldi, M., Fritz, M., Bauerlein, E., Radmacher,M., Sackmann, E., and Boulbitch, A. (2000).

Bacterial turgor pressure can be measuredby atomic force microscopy. Phys. Rev. E62:1034–1044.

Auerbach, I. D., Sorensen, C., Hansma, H. G.,and Holden, P. A. (2000). Physical morphol-ogy and surface properties of unsaturatedPseudomonas putida biofilms. J. Bacteriol.182:3809–3815.

Beech, I. B., Cheung, C. W. S., Johnson, D. B.,and Smith, J. R. (1996). Comparative studies ofbacterial biofilms on steel surfaces using atomicforce microscopy and environmental scanningelectron microscopy. Biofouling 10:65–77.

Benoit, M., Gabriel, D., Gerisch, G., and Gaub,H. E. (2000). Discrete interactions in cell

adhesion measured by single-molecule forcespectroscopy. Nature Cell Biol. 2:313–317.

Beveridge, T. J., and Graham, L. L. (1991).Surface layers of bacteria. Microbiol Rev55:684–705.

Binnig, G., Quate, C. F., and Gerber, C. (1986).Atomic force microscope. Phys. Rev. Lett.56:930–933.

Boonaert, C. J. P., Toniazzo, V., Mustin, C., Duf-rene, Y. F., and Rouxhet, P. G. (2002). Defor-mation of Lactococcus lactis surface in atomicforce microscopy study. Colloids Surfaces B:Biointerfaces 23:201–211.

Boulbitch, A., Quinn, B., and Pink, D. (2000).Elasticity of the rod-shaped gram-negativeeubacteria. Phys. Rev. Lett. 85:5246–5249.

Bowen, W. R., Lovitt, R. W., and Wright, C. J.(2001). Atomic force microscopy study ofthe adhesion of Saccharomyces cerevisiae.J. Colloid Interface Sci. 237:54–61.

Boyle-Vavra, S., Hahm, J., Sibener, S. J., andDaum, R. S. (2000). Structural and topologicaldifferences between a glycopeptide-intermedi-ate clinical strain and glycopeptide-susceptiblestrains of Staphylococcus aureus revealed byatomic force microscopy. Antimicrob. AgentsChemother. 44:3456–3460.

Braga, P. C., and Ricci, D. (1998). Atomic forcemicroscopy: Application to investigation ofEscherichia coli morphology before and afterexposure to cefodizime. Antimicrob. AgentsChemother. 42:18–22.

Braga, P. C., and Ricci, D. (2000). Detectionof rokitamycin-induced morphostructural alter-ations in Helicobacter pylori by atomic forcemicroscopy. Chemotherapy 46:15–22.

Bremer, P. J., Geesey, G. G., and Drake, B.(1992). Atomic force microscopy examinationof the topography of a hydrated bacterialbiofilm on a copper surface. Curr. Microbiol.24:223–230.

Butt, H. J., Downing, K. H., and Hansma, P. K.(1990). Imaging the membrane protein bacteri-orhodopsin with the atomic force microscope.Biophys. J. 58:1473–1480.

Butt, H. J. (1992). Measuring local surfacecharge densities in electrolyte solutions witha scanning force microscope. Biophys. J.63:578–582.

Butt, H. J., Jaschke, M., and Ducker, W. (1995).Measuring surface forces in aqueous electrolytesolution with the atomic force microscope.Bioelectrochem. Bioenerg. 38:191–201.

Camesano, T. A., and Logan, B. E. (2000).Probing bacterial electrosteric interactions us-ing atomic force microscopy. Environ. Sci.Technol. 34:3354–3362.

Camesano, T. A., Natan, M. J., and Logan, B. E.(2000). Observation of changes in bacterial cell

Page 97: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 91

morphology using tapping mode atomic forcemicroscopy. Langmuir 16:4563–4572.

Clausen-Schaumann, H., Seitz, M., Krautbauer,R., and Gaub, H. E. (2000). Force spec-

troscopy with single bio-molecules. Curr. Opin.Chem. Biol. 4:524–530.

Colton, R. J., Engel, A., Frommer, J. E., Gaub,H. E., Gewirth, A. A., Guckenberger, R., Rabe,J., Heckel, W. M., and Parkinson, B. (eds.)

(1998). Procedures in Scanning Probe Micro-scopies, John Wiley & Sons, Chichester.

Dengis, P. B., Nelissen, L. R., and Rouxhet, P.G. (1995). Mechanisms of yeast flocculation:Comparison of top- and bottom-fermentingstrains. Appl. Environ. Microbiol. 61:718–728.

Doyle, R. J., and Rosenberg, M. (eds.) (1990).Microbial Cell Surface Hydrophobicity, Ameri-can Society for Microbiology, Washington, D.C.

Dufrene, Y. F. (2000). Direct characterization ofthe physicochemical properties of fungal sporesusing functionalized AFM probes. Biophys. J.78:3286–3291.

Dufrene, Y. F. (2001). Application of atomicforce microscopy to microbial surfaces: Fromreconstituted cell surface layers to living cells.Micron 32:153–165.

Dufrene, Y. F., Boonaert, C. J. P., Gerin, P. A.,Asther, M., and Rouxhet, P. G. (1999). Directprobing of the surface ultrastructure and molec-ular interactions of dormant and germinat-ing spores of Phanerochaete chrysosporium . J.Bacteriol. 181:5350–5354.

Dufrene, Y. F., Boonaert, C. J. P., van der Mei,H. C., Busscher, H. J., and Rouxhet, P. G.(2001). Probing molecular interactions andmechanical properties of microbial cell surfacesby atomic force microscopy. Ultramicroscopy86:113–120.

Forsythe, J. H., Maurice, P. A., and Hersman, L.E. (1998). Attachment of a Pseudomonas sp. toFe(III)-(hydr)oxide surfaces. Geomicrobiology15:293–308.

Frisbie, C. D., Rozsnyai, L. F., Noy, A., Wrigh-ton, M. S., and Lieber, C. M. (1994). Func-tional group imaging by chemical forcemicroscopy. Science 265:2071–2074.

Gad, M., and Ikai, A. (1995). Method forimmobilizing microbial cells on gel surfacefor dynamic AFM studies. Biophys. J.69:2226–2233.

Gad, M., Itoh, A., and Ikai, A. (1997). Mappingcell wall polysaccharides of living microbialcells using atomic force microscopy. Cell Biol.Int. 21:697–706.

Grantham, M. C., and Dove, P. M. (1996). Inves-tigation of bacterial-mineral interactions usingfluid tapping mode atomic force microscopy.Geochim. Cosmochim. Acta 60:2473–2480.

Gunning, P. A., Kirby, A. R., Parker, M. L.,Gunning, A. P., and Morris, V. J. (1996). Com-parative imaging of Pseudomonas putida bacte-rial biofilms by scanning electron microscopyand both DC contact and AC non-contactatomic force microscopy. J. Appl. Bacteriol.81:276–282.

Haberle, W., Horber, J. K. H., and Binnig, G.(1991). Force microscopy on living cells.J. Vac. Sci. Technol. B 9:1210–1213.

Hancock, I. C. (1991). Microbial cell surfacearchitecture. In: Microbial Cell Surface Anal-ysis: Structural and Physicochemical Methods,N. Mozes, P. S. Handley, H. J. Busscher, andP. G. Rouxhet, (eds.), pp. 21–59, VCH Pub-lishers, New York.

Hartley, P., Matsumoto, M., and Mulvaney, P.(1998). Determination of the surface poten-tial of two-dimensional crystals of bacteri-orhodopsin by AFM. Langmuir 14:5203–5209.

Heinz, W. F., and Hoh, J. H. (1999). Spatiallyresolved force spectroscopy of biological sur-faces using the atomic force microscope.Tibtech 17:143–150.

Hinterdorfer, P., Baumgartner, W., Gruber, H. J.,Schilcher, K., and Schindler, H. (1996). Detec-tion and localization of individual anti-body–antigen recognition events by atomicforce microscopy. Proc. Natl. Acad. Sci USA93:3477–3481.

James, A. M. (1991). Charge properties of micro-bial cell surfaces. In: N. Mozes, P. S. Handley,H. J. Busscher, and P. G. Rouxhet, (eds.), Mic-robial Cell Surface Analysis: Structural andPhysicochemical Methods, pp. 221–262, VCHPublishers, New York.

Janshoff, A., Neitzert, M., Oberdorfer, Y., andFuchs, H. (2000). Force spectroscopy of molec-ular systems—single molecule spectroscopy ofpolymers and biomolecules. Angew. Chem. Int.Edit. 39:3213–3237.

Kapoor, A., and Viraraghavan, T. (1995). Fungalbiosorption—an alternative treatment option

Page 98: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

92 PROPERTIES OF MICROBIAL CELL SURFACES EXAMINED BY ATOMIC FORCE MICROSCOPY

for heavy metal bearing wastewaters: A review.Bioresource Technol. 53:195–206.

Karrasch, S., Hegerl, R., Hoh, J., Baumeister, W.,and Engel, A. (1994). Atomic force microscopyproduces faithful high-resolution images of pro-tein surfaces in an aqueous environment. Proc.Natl. Acad. Sci. USA 91:836–838.

Kasas, S., Fellay, B., and Cargnello, R. (1994).Observation of the action of penicillin on Bacil-lus subtilis using atomic force microscopy:Technique for the preparation of bacteria. Surf.Interface Anal. 21:400–401.

Kasas, S., and Ikai, A. (1995). A method foranchoring round shaped cells for atomic forcemicroscope imaging. Biophys. J. 68:1678–1680.

Lee, G. U., Chrisey, L. A., and Colton, R. J.(1994a). Direct measurement of the forcesbetween complementary strands of DNA.Science 266:771–773.

Lee, G. U., Kidwell, D. A., and Colton, R. J.(1994b). Sensing discrete streptavidin–biotininteractions with atomic force microscopy.Langmuir 10:354–357.

Lower, S. K., Hochella, M. F., and Beveridge,T. J. (2001). Bacterial recognition of mineralsurfaces: Nanoscale interactions between She-wanella and α-FeOOH. Science 292:1360–1363.

Marszalek, P. E., Oberhauser, A. F., Pang, Y. P.,and Fernandez, J. M. (1998). Polysaccharideelasticity governed by chair–boat transitions ofthe glucopyranose ring. Nature 396:661–664.

Moller, C., Allen, M., Elings, V., Engel, A., andMuller, D. J. (1999). Tapping-mode atomicforce microscopy produces faithful high-resolu-tion images of protein surfaces. Biophys J77:1150–1158.

Moller, C., Buldt, G., Dencher, N. A., Engel, A.,and Muller, D. J. (2000). Reversible loss ofcrystallinity on photobleaching purple mem-brane in the presence of hydroxylamine. J. Mol.Biol. 301:869–879.

Mozes, N., Handley, P. S., Busscher, H. J., andRouxhet, P. G. (eds.) (1991). Microbial CellSurface Analysis: Structural and Physicochem-ical Methods. VCH Publishers, New York.

Muller, D. J., and Engel, A. (1999). Voltage andpH-induced channel closure of porin OmpFvisualized by atomic force microscopy. J. Mol.Biol. 285:1347–1351.

Muller, D. J., Buldt, G., and Engel, A. (1995a).Force-induced conformational change of bacte-riorhodopsin. J. Mol. Biol. 249:239–243.

Muller, D. J., Schabert, F. A., Buldt, G., and En-gel, A. (1995b). Imaging purple membranes inaqueous solutions at subnanometer resolutionby atomic force microscopy. Biophys. J.68:1681–1686.

Muller, D. J., Baumeister, W., and Engel, A.(1996). Conformational change of the hexag-onally packed intermediate layer of Deinococ-cus radiodurans monitored by atomic forcemicroscopy. J. Bacteriol. 178:3025–3030.

Muller, D. J., Engel, A., and Amrein, M. (1997a).Preparation techniques for the observation ofnative biological systems with the atomic focremicroscope. Biosens. Bioelectron. 12:867–877.

Muller, D. J., Schoenenberger, C. A., Schabert,F., and Engel, A. (1997b). Structural changesin native membrane proteins monitored atsubnanometer resolution with the atomic forcemicroscope: A review. J. Struct. Biol. 119:149–157.

Muller, D. J., Fotiadis, D., and Engel, A. (1998).Mapping flexible protein domains at sub-nanometer resolution with the atomic forcemicroscope. FEBS Lett. 430:105–111.

Muller, D. J., Baumeister, W., and Engel, A.(1999a). Controlled unzipping of a bacterialsurface layer with atomic force microscopy.Proc. Natl. Acad. Sci. USA 96:13170–13174.

Muller, D. J., Fotiadis, D., Scheuring, S., Muller,S. A., and Engel, A. (1999b). Electrostaticallybalanced subnanometer imaging of biologicalspecimens by atomic force microscope. Bio-phys. J. 76:1101–1111.

Oesterhelt, F., Oesterhelt, D., Pfeiffer, M., Engel,A., Gaub, H. E., and Muller, D. J. (2000).Unfolding pathways of individual bacteri-orhodopsin. Science 288:143–146.

Ohnesorge, F., Heckl, W. M., Haberle, W., Pum,D., Sara, M., Schindler, H., Schilcher, K., Kie-ner, A., Smith, D. P. E., Sleytr, U. B., and Bin-nig, G. (1992). Scanning force microscopystudies of the S-layers from Bacillus coagulansE38-66, Bacillus sphaericus CCM2177 and ofan antibody binding process. Ultramicroscopy42–44:1236–1242.

Ong, Y. L., Razatos, A., Georgiou, G., and Shar-ma, M. M. (1999). Adhesion forces between

Page 99: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 93

E. coli bacteria and biomaterial surfaces.Langmuir 15:2719–2725.

Persike, N., Pfeiffer, M., Guckenberger, R., Rad-macher, M., and Fritz, M. (2001). Direct obser-vation of different surface structures onhigh-resolution images of native halorhodopsin.J. Mol. Biol. 310:773–780.

Pum, D., Weinhandl, M., Hodl, C., and Sleytr, U.B. (1993). Large-scale recrystallization of

the S-layer of Bacillus coagulans E38-66 atthe air/water interface and on lipid films.J. Bacteriol. 175:2762–2766.

Pum, D., and Sleytr, U. B. (1995). Monomolec-ular reassembly of a crystalline bacterialcell surface layer (S-layer) on untreated andmodified silicon surfaces. Supramolecular Sci.2:193–197.

Pum, D., and Sleytr, U. B. (1996). Molecularnanotechnology and biomimetics with S-layers.In: Crystalline Bacterial Cell Surface Pro-teins (U. B. Sleytr, P. Messner, D. Pum, andM. Sara, eds.), pp. 175–209, R. G. Landes,Austin, TX.

Razatos, A., Ong, Y. L., Sharma, M. M., andGeorgiou, G. (1998). Molecular determinantsof bacterial adhesion monitored by atomicforce microscopy. Proc. Natl. Acad. Sci. USA95:11059–11064.

Razatos, A., Ong, Y. L., Boulay, F., Elbert, D.L., Hubbell, J. A., Sharma, M. M., and Geor-giou, G. (2001). Force measurements betweenbacteria and poly(ethylene glycol)-coated sur-faces. Langmuir 16:9155–9158.

Rief, M., Oesterhelt, F., Heymann, B., and Gaub,H. E. (1997). Single molecule force spec-troscopy on polysaccharides by atomic forcemicroscopy. Science 275:1295–1297.

Savage, D. C., and Fletcher, M. (eds.) (1985).Bacterial Adhesion, Plenum Press, New York.

Schabert, F. A., Henn, C., and Engel, A. (1995).Native Escherichia coli OmpF porin surfacesprobed by atomic force microscopy. Science268:92–94.

Scheuring, S., Ringler, P., Borgnia, M., Stahl-berg, H., Muller, D. J., Agre, P., and Engel, A.(1999). High resolution AFM topographs of theEscherichia coli water channel aquaporin Z.EMBO J. 18:4981–4987.

Sokolov, I. Y., Firtel, M., and Henderson, G. S.(1996). In situ high-resolution atomic forcemicroscope imaging of biological surfaces. J.Vac. Sci. Technol. A 14:674–678.

Steele, A., Goddard, D. T., and Beech, I. B.(1994). An atomic force microscopy studyof the biodeterioration of stainless steel inthe presence of bacterial biofilms. Int. Biodet.Biodeg. 34:35–46.

Telegdi, J., Keresztes, Z., Palinkas, G., Kalman,E., and Sand, W. (1998). Microbially influ-enced corrosion visualized by atomic forcemicroscopy. Appl. Phys. A 66:S639–S642.

van der Aa, B. C., Michel, R. M., Asther, M.,Zamora, M. T., Rouxhet, P. G., and Dufrene,Y. F. (2001). Stretching cell surface macro-molecules by atomic force microscopy. Lang-muir 17:3116–3119.

van der Aa, B. C., and Dufrene, Y. F. (2002).In situ characterization of bacterial extracellu-lar polymeric substances by AFM. ColloidsSurf. B: Biointerfaces 23:173–182.

van der Mei, H. C., Rosenberg, M., and Buss-cher, H. J. (1991). Assessment of microbialcell surface hydrophobicity. In: MicrobialCell Surface Analysis: Structural and Physic-ochemical Methods (N. Mozes, P. S. Handley,H. J. Busscher, P. G. Rouxhet, eds.), pp. 263–287, VCH Publishers, New York.

van der Mei, H. C., Busscher, H. J., Bos, R., deVries, J., Boonaert, C. J. P., and Dufrene, Y. F.(2000). Direct probing by atomic forcemicroscopy of the cell surface softness of afibrillated and non-fibrillated oral streptococcalstrain. Biophys. J. 78:2668–2674.

Xu, W., Mulhern, P. J., Blackford, B. L., Jeri-cho, M. H., Firtel, M., and Beveridge, T. J.(1996). Modeling and measuring the elasticproperties of an archaeal surface, the sheathof Methanospirillum hungatei, and the impli-cation for methane production. J. Bacteriol.178:3106–3112.

Yao, X., Jericho, M., Pink, D., and Beveridge, T.(1999). Thickness and elasticity of gram-negative murein sacculi measured by atomicforce microscopy. J. Bacteriol. 181:6865–6875.

Page 100: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

CHAPTER 6

SCANNING PROBE MICROSCOPY OF PLANTCELL WALL AND ITS CONSTITUENTSKSENIJA RADOTICCenter for Multidisciplinary Studies, University of Belgrade, Kneza Viseslava 1, 11000 Belgrade,Serbia and Montenegro

MIODRAG MICICMP Biomedicals, Inc., 15 Morgan, Irvine, California, 92618-2005

MILORAD JEREMICFaculty of Physical Chemistry, University of Belgrade, Studentski Trg 16, 11000 Belgrade, Serbiaand Montenegro

6.1. INTRODUCTION

In the majority of plant cells, the cell wallis a compartment lying outside the plas-malemma. It is involved in the determina-tion of both the morphology and functionof the plant cell. Since the cell wall isthe outermost barrier of the plant cell, itsmajor biological function is mechanical sup-port and stress protection. Walls or selectedwall components have chemical roles too,such as ion exchangers, bacterial agglutinins,and sources of messages (the oligosaccha-rines) to instruct the protoplast. Cell wall isbuilt from lignin, polysaccharides, and pro-teins. The general architectural plan of thecell wall involves a fibrous material embed-ded in an amorphous matrix. The chemicaland physical structure of the wall dependson the plant group and cell type, but a gen-eral characteristic of all plant cell walls isthat they are not chemically and physically

homogeneous (Goodwin and Mercer, 1985;Varner and Lin, 1989).

Polysaccharides in the cell wall canbe subdivided in two categories: thosethat exist within the wall in crystallineform and those that form rather amorphousphase. The former polysaccharides arevery long, unbranched molecules aggregatedtogether in bundles called microfibrils. Themicrofibrils consists of individual cellulosemolecules (a β-4-linked D-glucan) associatedinto crystalline or near-crystalline arraysthat are nearly free of water (Roelofsen,1965). Xylans (β-4-linked xylosyl residues)are also present in form of microfibrilsin the cell walls. The microfibrils areembedded in a matrix composed of thenoncrystalline polysaccharides. These arehighly branched macromolecules containingseveral different species of monosaccharideresidues. They are covalently or hydrogenbonded to the other polysaccharide and

Force Microscopy: Applications in Biology and Medicine, edited by Bhanu P. Jena and J.K. Heinrich Horber.Copyright 2006 John Wiley & Sons, Inc.

95

Page 101: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

96 SCANNING PROBE MICROSCOPY OF PLANT CELL WALL AND ITS CONSTITUENTS

nonpolysaccharide components of the cellwall. Matrix polysaccharides are comprisedof hemicelluloses (xylans, xyloglucans,mannans, galactanes) and pectins (mostlybuilt from polyuronic acid). Lignin, abranched polymer of phenolic alcohols, hasan important role in mechanical support andstress protection in the cell. It strengthensthe wall by forming a ramified networkthroughout the matrix, thus anchoring thecellulose microfibrils more firmly. Certainproteins in the cell walls have structural role(most abundant is extensin), while some ofthem are enzymes. Water is an extremelyimportant constituent of the cell walls. Itpermeates the matrix, but not the crystallineregions of the microfibrils. The amount ofwater in the wall depends on the natureof the matrix polysaccharides, with whichit forms close intermolecular associationsand gel-like structures, and on the degreeof lignification (the more lignin, the lesswater is present). Lignin and cellulose areconsidered as molecules important in watertransport through the xylem of the higherplants (Laschimke, 1989).

The processes of synthesis and secre-tion of wall components are mostly known,but assembly of these components intoa growing wall is poorly understood. Itis conceivable that the addition of non-cellulosic components to an existing wallis entirely by self-assembly, with eachcomponent designed to fit appropriatelywith others. Cellulose microfibrils are laiddown by apposition on the inner surfaceof the existing wall. The matrix com-ponents—hemicelluloses, pectins, and pro-teins—are added by intussusceptions, andthey can occur at any place in the existingwall (Goodwin and Mercer, 1985).

6.2. AFM EXAMINATIONOF CELL WALLS

Atomic force microscopy (AFM) may con-tribute in revealing the general architecture

of the cell walls, as well as in the way partic-ular components in the whole wall structureare assembled. Using AFM, it has been pos-sible to image the architecture of isolatedcell wall fragments, as in case of the Chi-nese water chestnut cell walls (Morris et al.,1997). The cell wall is revealed as a lam-inated fibrous structure. Molecular structurewas clearly represented throughout the topo-graphical image. Using different image pro-cessing methods, it was possible to obtainthe wall thickness. Error signal mode imag-ing effectively flattens the rough surface,thereby generating a better visual impressionof the structure within the surface. How-ever, it is not possible to obtain measure-ments of layer thickness or fiber diameterfrom the error signal mode images. Molec-ular structure is clearly represented through-out the topographical image, but the numberof the gray levels required to represent thisimage is too large to be perceived by the eye.Thus regions of the image appear devoid ofthe structure. It is necessary to isolate thehigh-frequency structural information fromthe low-frequency curvature of the cell wallsurface. One approach is to use a high-passfrequency filter. This improves the image,but there is some leeway in the choice ofthe cutoff frequency. An alternative is tolocally average or smooth the surface. Thiscan be subtracted from the topographicalimage yielding a “flattened” projection ofthe surface. The measured fiber thickness issmaller for fibers packed into arrays than forisolated fibers due to a reduction in probebroadening. Cellulose microfibrils were dis-tinguished within the whole wall fragments,but hemicelluloses or pectin networks couldnot be distinguished. Disruption of the wallfragments may produce some artifacts, butreveals laminated nature of the cell wall.Using AFM, the laminated structure of thecell walls isolated from maize roots has beenobserved (Radotic, 1994). The fibrous struc-tures in cell wall fragments were ascribed tocellulose, according to their dimensions inthe Radotic study.

Page 102: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

AFM EXAMINATION OF PECTINS 97

In a paper by Round et al. (1996), the gen-eral conditions and difficulties in AFM con-tact and error mode imaging of the plant cellwalls has been addressed, the most impor-tant being sample hydrations. When the sam-ple is imaged in the hydrated state, it doesnot stick to the substrate, and the force ofthe AFM tip is enough to peel it off, mak-ing imaging very difficult, if not impossi-ble. On the other hand, dehydrated sampleimaging in air is very difficult and intro-duces artifacts, as the dry samples becomeextremely rough, and additionally, any mois-ture that is present on the surface of the sam-ple could induce artifacts by both pulling thetip down and by playing with refraction ofthe laser beam. Later, studies determined thatbutanol is the best solvent for imaging plantcell wall samples. The observed structure ofthe primary cell wall associated with plas-malemma expressing polysaccharides fibrilstructure packed into the closed arrays isvery similar to the one previously observedby TEM (Blankey et al., 1983; McCannet al., 1990) and suggested by Roelofsenet al. (1965).

Duchesne and Daniel (1999) have re-viewed various microscopic methods, appliedin the study of wood fiber ultrastructure,including using AFM. The high degree ofsimilarities between FE-SEM and other elec-tron microscopy methods and observed fib-rilar features on the pulp fiber surfaces byAFM is extraordinary. However, the appli-cability of the AFM in studying in situpulp is questionable due to the fact thatits ability to obtain good quality imagesis compromised by the condition of harshphysical–chemical environment, capable ofaltering the surface properties of both tipand substrate. Another paper by Niemiet al. (2002) specifically reviewed the appli-cations of scanning probe microscopy inwood, paper, and fiber research. Both dry-air imaging and liquid imaging have beenpresented, and special attention has beengiven to the modification occurring at the

nanoscale at the fiber surface in differentstage of pulping.

6.3. AFM EXAMINATION OF PECTINS

Pectins are one of the main constituents ofthe plant cell walls, being highly branchedpolysaccharides whose primary, secondary,and tertiary structures are still little under-stood (Ridley et al., 2001). Moreover, thefunction of the pectins in the physiology ofthe plant cell, as well as the process of itsbiosynthesis, is only partially known (Ridleyet al., 2001). One of the most important ques-tions in plant physiology today is understand-ing the properties, functions, and biosynthe-sis of this class of polymers. The practicalaspect of knowing more about the pectinstructures comes from their role in food; thatis, they are the main components of manyplant-based foods for human consumption.

Because branched pectins are highly irreg-ular structures, and are also intertwined withother plant cell wall constituents, it is vir-tually impossible to make them into crys-talline form after their extraction, whichmakes them unsuitable for x-ray diffrac-tion or neutron scattering studies. A classicalmethod of chemical analysis introduces dif-ferent artifacts, mainly in the forms of side-chain cleavage. Atomic force microscopy(AFM) allows direct observation, with sub-nanometer spatial resolution of extractedpolysaccharides (Round et al., 1997, 2001),as well as of fragmented cell walls (Morriset al., 1997; Round et al., 1996).

In a study by Round et al. (1997), branch-ing in isolated pectin polysaccharides wasobserved for the first time. One of therepresentative images of such a pectin-branched macromolecule is presented inFig. 6.1. The large linear branch between30 and 170 nm has been observed in thatstudy. The pectin macromolecules wereisolated from the cell walls of unripe toma-toes for the study. Extracted pectin solu-tion (1–3 µg/ml) was applied to the surface

Page 103: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

98 SCANNING PROBE MICROSCOPY OF PLANT CELL WALL AND ITS CONSTITUENTS

Figure 6.1. AFM image of pectins. Image size250 × 250 nm. (Courtesy of Dr. A. N. Round, Instituteof Food Research, Norwich, UK.)

of cleaved mica, and it was imaged byAFM in the contact mode in butanol. AFMmicrographs of the extracts show aggregatesand single molecules. To distinguish singlepectin macromolecules from aggregates, theheight measurement was used. According toRound et al. (1997), the observed lengthsof the individual pectin chains are in therange between 30 and 390 nm, with log-normal distribution of chain lenght. Interest-ingly, the average molecular weight, calcu-lated from the length of the macromolecularcontours, are in the range of 50,000 Da,which is at least 200 times smaller thanthe estimated molecular weight of pectins.About 20% of the macromolecules shownin the AFM micrographs branched into sidechains, and 30% of the branched macro-molecules are multiply branched, forming aso-called “macromolecular brush” structure.The differentiation between covalent molec-ular branching and statistical overlapping oftwo independent chains has been performed,based on the height distribution. Becausethe average diameter of the polymer back-bone is in the range from 0.5 to 0.8 nm,the height at the branching point should bein the same range. On the other hand, if

the “node point” is higher, then it is to beassumed that the AFM image shows just thetwo overlapping polymer chains. Lengths ofthe side chains have been estimated to be inthe range from 30 to 170 nm. One explana-tion is that the pectin branches are made ofpoly-galactouronic acids; while the alterna-tive is that the branches are made of neutralsugar monomers, attached to the backboneof polymer via branched rhamnose. The lateris less likely, since there is no confirmationof the existence of long neutral sugar chains,by methylation chemical analysis. Studies bythe Round et al. (2001) shows that the dis-tributions of the total amount of polymericbranches do not match the distribution of theneutral sugars in pectins. The authors in thestudy further suggest that the long chains aremost likely polygalacturonic acid attached tothe pectin macromolecular backbone, withoccasional long neutral sugar branches. Themajority of the neutral sugars are attachedto the pectin backbone in the form of veryshort branched chains, a model which is inagreement with the results of sequencinganalysis of pectins. The chemical analysisof pectins is performed by Semana hydrol-ysis followed by identification of monomersusing gas chromatography (Blankey et al.,1983). It shows that there is no statisticallysignificant relationship between the neutralsugars (arabinose and galactose) content andthe branch lengths.

In a separate study by Morris et al. (1997),AFM was used to image fragments of the cellwall, pectin, and carrageenan. The polysac-charides were imaged in situ (i.e., embeddedin their natural environment in the cell wallfragments) as well as extracted, from tomatoand from the Chinese water chestnut. Bothcell fragments and CDTA extracted polysac-charides were imaged on cleaved mica sur-face, under butanol, applying the differ-ential contact mode (“error mode”) AFM.The polysaccharide microfibers are easilyobserved as packed into the arrays, in laminarstructure of the plant cell wall. However, itwas impossible to distinguish between pectin

Page 104: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

USE OF AFM IN THE STUDY OF CELLULOSE 99

and hemicelluloses fibers in the AFM micro-graphs generated.

6.4. USE OF AFM IN THE STUDYOF CELLULOSE

Native cellulose has been the most stud-ied subject in polymer science. Cellulosemicrofibrils in a typical primary cell wallare elliptical in cross section with axes of5–30 nm. They constitute 20–30% of thedry weight of the wall. Despite many reports,summarized in review articles (Bayer et al.,1998; O’Sullivan, 1997) and books (Kres-sig, 1993), the ultrastructure of cellulose hasnot been completely resolved. It is not yetclear how new cellulose chains are assem-bled into microfibrils at the cell surface,and our knowledge of the structure of thebasic crystalline units is incomplete (Haet al., 1997). Although the enzymatic appa-ratus for cellulose synthesis is poorly under-stood, it is apparently contained in particlesobserved in electron micrographs as orderedgroups of rosettes in the plasmalemma (Hertand Hausser, 1984). The cellulose moleculesextruded by a single rosette aggregate withthose from adjacent rosettes to form themicrofibrils, where cellulose molecules arepacked in a parallel fashion (Varner and Lin,1989). The microfibrils are then assembledinto superstructures, such as the cell wall andfibers.

AFM is idealy suited to study cellulosesurfaces at near-angstrom resolution. AFMimages of Valonia cellulose microcrystalsunder propanol and water demonstrates a res-olution approximating that of the best TEMmicrographs (Baker et al., 1997). The AFMimages revealed clear structural details con-sistent with the 0.54-nm repeat unit (glucose)along the cellulose chains. An intermolecu-lar spacing of about 0.6 nm is also observed.AFM has also been used to evaluate thephase dimensions of cellulosic blends andcomposites. Although most of these stud-ies employed primarily topographical imag-ing, there have also been studies involving

phase imaging. For example, the distributionof lignin on the surface of a wood pulp fibercould be visualized on the phase image whileit did not appear in the height images (Borasand Gatenholm, 1999). This is because theproperties of lignin and its functions, suchas adhesion, friction, and viscoelasticity, aredifferent from those of cellulose and hemi-celluloses. These studies suggest that AFMphase imaging can distinguish between theunmodified cellulose regions and the cellu-lose ester regions in the modified pulp fibers.

Digestion of cellulose by cellulolyticmicroorganisms has been the subject ofintensive research efforts for decades, yetthe exact course of the initial binding anddisruption of the cellulose fibers remainsunclear. Cellulolytic microorganisms, suchas the fungus Trichoderma reesei, degradecrystalline cellulose by secreting mixtures ofcellulases that have different but comple-mentary modes of action on the substrate.Cellulases from both bacterial and fungalsources are responsible for the hydrolysis ofthe β-1,4-glucosidic bonds in cellulose. Sincecellulose is resistant to microbial degrada-tion, multiple enzyme systems are requiredto efficiently degrade its crystalline structure.Such systems comprise a collection of freecellulases and/or multicomponent complexescalled cellulosomes (Bayer et al., 1998). Thetrend in recent years has been to deter-mine the atomic structure and then to iden-tify the catalytic residues of members ofa given cellulase family. There have alsobeen efforts to reveal the interaction of cel-lulase with cellulose and the mechanismof cellulase-catalyzed hydrolysis of cellulose(Chanzy and Henrissat, 1985; Hayashi et al.,1998). Studies on two cellulases from Tri-choderma reesei on the cotton fibers pro-vide the first physical evidence of (a) cel-lulase action at the surface of cotton fibersand (b) its movement along the fibers, (Leeet al., 2000). For over four decades therehas been debate on whether a physical factornecessary for degrading crystalline celluloseis present in cellulases. AFM study of the

Page 105: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

100 SCANNING PROBE MICROSCOPY OF PLANT CELL WALL AND ITS CONSTITUENTS

interaction between catalytically active andinactive Trichoderma reesei cellulase com-ponents and cotton fibers have shown thatinactivated major enzyme component (CBHI) still had the ability to bind to the cot-ton fiber. AFM also provided evidence thatbinding alone is not sufficient to disrupt themicrofibril surface, but catalytic activity ofCBH I is required for fiber disruption (Leeet al., 1996).

6.5. SCANNING PROBE MICROSCOPYON LIGNIN

Although lignin has been extensively stud-ied in different scientific disciplines for morethan 100 years, much remains unknownabout its formation, structure and bondingenvironment in the cell wall. Since lignin ishighly interlinked with the other constituentsof the cell wall, its isolation in unalteredform is impossible. There is opinion thatlignin does not exist outside the cell wall.Therefore, knowledge on lignin structure hasaccumulated from the studies of lignin likepolymers and isolated fragments of naturalmacromolecule. Experimental evidence hasshown that enzymatic synthesis of lignin pro-ceeds through free radical polymerization ofphenolic alcohols (coniferyl, synapyl, and p-coumaryl alcohol—called monolignols), cat-alyzed by different peroxidases in the cellwalls (Lewis and Yamamoto, 1990). Phenolicalcohols are synthesized in the cell startingfrom phenylalanine and tyrosine (Erdtman,1957; Freudenberg and Niedercorn, 1958;Higuchi and Brown, 1963; Schubert andNord, 1957). They are subsequently exportedto the cell wall enclosed in vesicles, inthe form of glucosides (Bardinskaya andSafonov, 1959; Schmid and Grisebach, 1982;Schmid et al., 1982). After fusion of the vesi-cles with the plasma membrane, glucosidesare released by exocytosis (Pickett-Heaps,1968) into the cell wall space. β-Glucosidase,localized in cell walls, hydrolyzes the glu-cosides (Marcinowski and Grisebach, 1977;

Schmid et al., 1982), releasing phenolic alco-hols, which serve as the substrate for theperoxidases in lignin synthesis. Hydrogenperoxide, necessary for the peroxidase reac-tion, is produced in the cell walls, but themechanism responsible for its formation isstill obscure. There is a lack of appropri-ate methodology that would allow us todirectly monitor the processes of lignin syn-thesis and the activity of different peroxi-dases present in the cell wall (Lewis andYamamoto, 1990). Most in vivo experimentsare based on the histochemical methods oftissue dyeing in the course of lignification(Freudenberg et al., 1958; Harkin and Obst,1973; Catesson et al., 1986; Chibbar and vanHuystee, 1986), the use of the immunologi-cal probes (Bernardini et al., 1986; Conroy,1986), or the electrophoretic identification ofthe isoenzymes (Gaspar et al., 1985; Imbertyet al., 1985; Catesson et al., 1986). Mono-lignols have been rarely used as substratesin these in vivo experiments, due to thelack of suitable method for direct monitor-ing of their oxidation (Lewis and Yamamoto,1990). The majority of experiments of suchkind deal with the conversion of radioac-tively labeled lignin precursors, added to themedium where the plants were grown, intothe lignin in the cell walls (Stone, 1953;Stewart et al., 1953; Marcinowski and Grise-bach, 1977; Shann and Blum, 1987). In vitroexperiments of phenolic alcohols polymer-ization in the presence of peroxidase havecontributed to an understanding of the pro-cess of enzymic polymerization of phenolicalcohols in cell walls (Freudenberg et al.,1958; Nozu, 1967; Freudenberg and Toribio,1969; Hwang, 1982).

As a response to external stress condi-tions, the amount of phenolic compoundsincreases in the cell walls, as well as pro-duction of hydrogen peroxide (Mehlhorn andKunert, 1986). One of the first responsesto the stress is the increase in activity ofextracellular peroxidases, which use phenoliccompounds and hydrogen peroxide as sub-strates. A parallel increase in lignin quantity

Page 106: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

SCANNING PROBE MICROSCOPY ON LIGNIN 101

occurs, as an indication of the stress intensity(Castillo, 1986; Pandolfini et al., 1992; Polleand Chakrabarti, 1994).

The structure and function of lignin havebeen extensively studied by various methods.The structural changes of lignin at themolecular level are in close connection withits protective function in the plant cell.Therefore, a study of the effect of externalperturbations on the bonding pattern andmolecular organization of lignin is importantfor understanding the relationship betweenmechanism of lignin formation and structureon one hand, and plant response to stress onthe other.

Due to its complex structure, which is stillpuzzling even today, lignin is an interestingsubject for ultramicroscopic studies. Eluci-dating the secondary and tertiary structureof lignin can be considered a holy grail inplant physiology. Besides the importance inunderstanding the role of lignin in higherplant cells, there is also a very practicalaspect of this research. There has been a longhistory of microscopic investigation of theplant cells, starting with Antonvan Leeuwen-hoek’s first description of the cells in a cork-wood. The modern lignin and plant cell wallmicroscopy can be traced back to the late1970 with several investigations based onthe transmission electron microscopy (Fen-gel et al., 1981; Falkehag, 1975) and scan-ning electron microscopy studies (Kosikovaet al., 1978; Zakutna et al., 1983; Karmanovet al., 1993) of plant cell wall and extractedlignins. Since then, the microscopic studiesof lignin could be divided into the two majorlines: one directed toward the fundamentalinvestigation, and the other aimed at gainingknowledge necessary for improving practicalapplications such as pulp krafting and foodprocessing. With regard to the fundamentalaspect of lignin ultramicroscopy research, itis directed toward imaging either (a) primaryand secondary cell walls or (b) lignin andlignin-model compounds.

Since isolation of lignin by various chemi-cal procedures produces material of different

complexity, there was a dilemma whetherlignin formation in the cell wall is an uncon-trolled process, the result of which wouldbe a random polymer structure; or it is aprocess finely regulated in time and space(Lewis and Yamamoto, 1990). Scanning tun-neling microscopic (STM) study of in vitroenzymic polymerization of coniferyl alco-hol in time course studies (Fig. 6.2) showsome of the intermediate products and thefinal polymer structure (Radotic et al., 1994).The STM images with molecular resolution(Figs. 6.2A and 6.2B) reveal individual aro-matic monomers and many dimers and smalloligomers. The generally accepted pathwayof lignin synthesis starts from the coniferylalcohol monomer from which a free radical isproduced by hydrogen abstraction. The freeradical, which has several resonance forms(Elder and Worley, 1984; Hwang, 1985),attacks at the β position of the propenylgroup of a coniferyl alcohol monomer toform different dimer radical combinations.Subsequently, it undergoes various dehydro-genation, oxidation, hydration, and conden-sation reactions in which a variety of func-tional groups are formed on the aromatic ringand in the side chain. The structural patternshown in the inset of Fig. 6.2B and proposedby Hwang (1985) on the basis of LCAO-MOcalculations as one of the possible interme-diates of lignin formation is compatible withthe STM image in Fig. 6.2B. The imagesof final polymer, obtained 2 days after thestart of the polymerization (Fig. 6.2C), showthat the enzymatic polymer has a highlyordered structure even in in vitro conditions(Radotic et al., 1994). This study has alsoshown the modular organization of ligninpolymer. An approximate molecular weightof 85,000 ± 25,000 for the observed struc-tural unit of 5 ± 0.5 nm in diameter wasobtained. This would mean that one struc-tural unit, which we called supermodule,is composed of 500 monomers of coniferyl

Page 107: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

102 SCANNING PROBE MICROSCOPY OF PLANT CELL WALL AND ITS CONSTITUENTS

Figure 6.2. STM images of temporal formation and organization of DHP deposited onto HOPG substrate. (A) Twohours after mixing the components, ordered structure of DHP is not yet visible. CA molecules (arrow ) are observable.U = 199.9 mV, IT = 0.50 nA. (B) Detail from part A. CA molecules, 1.5 nm in diameter, are indicated byarrows. U = 199 mV, IT = 0.41 nA. Inset : One of possible intermediates of lignin formation, proposed by Hwang,compatible with encircled pattern on the image. (C) DHP 2 days after mixing components, with the sphericalregions. U = 20.1 mV, IT = 0.50 nA. (D) The same as part C, seen at another place of the surface. Supermodules,5 nm in diameter, are indicated by arrows. U = 20 mV, IT = 0.50 nA. (E) Three-dimensional image of DHPpresenting the layer of DHP with spherical regions. U = 20.1 mV, IT = 0.50 nA. (F) DHP 2 days after mixing thecomponents, without spherical regions. U = 180.1 mV, IT = 0.46 nA. (G) The same as part F, showing anotherregion of the sample. Supermodules, 5 nm in diameter, are indicated by arrows. U = 180.1 mV, IT = 0.60 nA.(H) Three-dimensional image of DHP layer without spherical regions. U = 180.1 mV, IT = 0.46 nA.

Page 108: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

SCANNING PROBE MICROSCOPY ON LIGNIN 103

Figure 6.3. STM images of DHP deposited onto thegold substrate 2 days after mixing the components.(A) U = 166.6 mV, IT = 0.83 nA. (B) Lattice-likestructure of DHP. U = 200 mV, IT = 1.2 nA.

alcohol. The substructure of supermodules isresolved in Fig. 6.3A. From the size of thesesubunits, the calculated approximate molecu-lar weight is 6000 ± 4000. This would meanthat a subunit, which is called a module, ismade up of 20 monomers.

The calculated molecular weights are ingood agreement with those obtained experi-mentally (Wayman and Obiaga, 1974). Theseresults demonstrate that modules consist of20 monomers and are formed first. Theysubsequently polymerize to form supermod-ules, which in turn aggregate to yield glob-ules of different size, as schematically rep-resented in Fig. 6.7. The bonds betweenthe modules in a supermodule are weakerthan the bonds between monomers within

the modules. However, they are of cova-lent type, since both modules and super-modules exist as distinct molecular speciesin solution and are quite stable. On theother hand, the supermodules are intercon-nected by intermolecular forces within glob-ules. These globules exist only within solidlignin and cannot be detected in solution asindependent molecular species. Such a prin-ciple of polymer organization is summarizedin Fig. 6.7, which highlights the major stepsof polymerization and organization of enzy-matic polymer. More detailed investigationof self-organization of lignin aggregates intohigher structural levels, and influence of sub-strate has been examined by ESEM of syn-thetic lignin (Micic et al., 2000b).

AFM study of polymer obtained usingenzymic polymerization of coniferyl alco-hol has confirmed the existence of strongintermolecular forces, which are responsi-ble for holding lignin supermodules withinthe globule and also holding the globuletogether. Micic et al. (2001a) have exam-ined enzymatic polymer of coniferyl alco-hol (DHP), using contact mode AFM. Inthe study, mechanical and rheological prop-erties of individual globules of the lignin,as well as the forces of interaction betweenlignin globules, have been examined. Theobjective of this research was to answer ifassembly of lignin semi-ordered structures,as observed by various imaging methods, is aresult of random collision and packing of thelignin globules or is a consequence of inter-molecular forces, such as hydrogen bondingand London force. Force spectroscopy stud-ies have been performed using Si3N can-tilever tips, incubated with a lignin suspen-sion for 12 h at 37◦C. The ESEM imageof functionalized tip is shown in Fig. 6.4.Examination of mechanical and rheologicalproperties of lignin globule was performedusing a clean cantilever tip. Results showelastic behavior of the enzymatic DHP poly-mer. In force scans with clean tip therewere no observed events which could bedescribed as structure alteration such as

Page 109: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

104 SCANNING PROBE MICROSCOPY OF PLANT CELL WALL AND ITS CONSTITUENTS

Figure 6.4. AFM tip functionalized with the lignin model compound (SEM micrography).

unwinding of lignin globules or filamentpulling. Slight repulsive double-layer elec-trostatic force combined with a repulsivehydration force has also been observed. Mea-sured adhesion forces were found to be in therange of 5–7 nN. Retraction curve showsadhesion due to van der Waals forces andindentation of the sample by the tip (Moyet al., 1994).

Much more interesting results are obtainedwith the use of a lignin functionalizedtip. Some of the characteristic force scansare presented in Fig. 6.5. During the can-tilever approach, which corresponds to com-pression, complex interactions are observed,resulting in nonlinear shape of the approach-ing curve. This shape indicates some kind ofhydrophobic interaction, as there is a grad-ual pull-off. Also, some secondary struc-ture unfolding and reorganization may con-tribute to the shape of the curve. The non-linear event observed during the approachand retraction of the functionalized cantileverin the proximity of the substrate can bedescribed as a fusion of two globules orglobule secondary structure. However, theseforce curves clearly reveal the existence ofthree cohesion peaks during the cantileverretraction, within the distance correspondingto the diameter of individual lignin globules.Peaks with a force intensity range of 1–8 nN

are reproducible, but their exact positionand intensity vary. As lignin is the mostentropic macromolecule in terms of the ran-domness of its structure as found in nature,it is believed that the discrepancy in posi-tion and intensity of peaks could be asso-ciated by the different ways that two ligninmacromolecules or macromolecular assem-blies can interact. The shape of the retractionpeaks, which indicates a jump-off, expressesa strong hydrophobic van der Waals forceand hydrogen bonding interactions.

Results of the AFM force scans, combinedwith the previous observation with ESEM,lead to the conclusion that the lignin glob-ule consists of at least two-layered spherical,“onion-like” structure, which is composed ofindividual macromolecular subunits. An inte-rior space between these shells may be filledwith water or with water and air in order toaccommodate hydrophobic and hydrophilicregions of lignin macromolecules. Layeredstructure of lignin has been finally con-firmed by NSOM (Near Field Scanning Opti-cal Microscopy) (Micic et al., 2004). NSOMprovides possibility to observe objects belowdiffraction limit. Schematic view of a ligninglobule layered structure is presented onFig. 7. Such organization of lignin globulesmay have physiological relevance, becauseit reduces the mass and amount of materials

Page 110: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

SCANNING PROBE MICROSCOPY ON LIGNIN 105

1

0

−1

−2

1.0

0.5

0.0

−0.5

−1.0

−1.5

−2.0

1.0

0.5

0.0

−0.5

−1.0

−1.5

−2.0

4.4 4.6 4.8 5.0 5.2 5.4

4.4 4.6 4.8 5.0 5.2 5.4

4.64.4 4.8 5.0 5.2 5.4

µm

µm

µm

vv

v

3 nN

200 nm

Figure 6.5. Atomic force scans obtained using AFM tip functionalized with lignin.

necessary to (a) create an individual ligninglobule, (b) and make the globule act asa “shock absorber” in response to outsidemechanical stress. Such structure also sup-ports the concept of lignin-mediated watertransport in plants (Laschimke, 1989). Thestructure is also in excellent agreement with

electron spin resonance observations of theexistence of areas of different mobility inlignin. The nonlinear shape of the peaks indi-cates complex interactions between ligninmacromolecules in globular assemblies. Theexistence of intermolecular attractive forcesbetween two lignin globules gives a clue

Page 111: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

106 SCANNING PROBE MICROSCOPY OF PLANT CELL WALL AND ITS CONSTITUENTS

that semi-ordered higher structures, such aschannel and pore-like hexagonal structures,are not generated by the random collisionof individual globules as earlier suggestedby Falkehag (1975) and Jurasek (1995), butmay be orchestrated by the actions of bothhydrophilic and hydrophobic intermolecularattractive forces, such as π–π interactions,hydrogen bonding, and van der Waals forces.The observed elastic property of lignin, alongwith its ability to reshape supramolecularstructure under mutual interaction of glob-ules, partially explains lignin’s adaptabilityto grow directionally within a geometricallyconstrained cell matrix.

Light is one of the external factors affect-ing plant metabolism. There is a trend towarda permanent increase in low-wavelengthUV radiation reaching the earth’s surface(Bojkov, 1994), affecting plants as a formof stress. It has been found that short-wavelength UV light penetrates through theepidermal cell layer of the conifer needlesinto the mesophyll tissue (Delucia et al.,1992). Phenolic alcohols can be polymerizednot only enzymatically, but also by pho-toirradiation to produce lignin-like polyer.However, their photochemical transforma-tions have been less studied in the past.The cell wall is the first target of lightaction in the cell. The study of the mech-anism of photochemical polymerization ofphenolic alcohols to lignin-like polymer andthe determination of structural differences ofthis polymer from the enzymatic polymeris a contribution to understanding how UVradiation affects plant tissue. Recent stud-ies have shown that UV radiation inducedpolymerization of coniferyl alcohol proceedsthrough an ionic mechanism. Intermediateproducts of polymerization have been identi-fied and characterized (Radotic et al., 1997).The structural differences between enzy-matic and photochemical lignin can be well-discerned using STM (Fig. 6.2, 6.3, and6.6). It appears that photochemical poly-mer contains the building units or modules,for larger assemblies such as supermodules.

Figure 6.6. STM images of the photochemical poly-mer of coniferyl alcohol deposited onto a goldplate. (A) U = 300 mV, IT = 1 nA, (B) U = 300 mV,IT = 1 nA.

However, within the photochemical polymerthere is a lower degree of order when com-pared with the enzymatic polymer (Radoticet al., 1998). The STM data are in agreementwith the data of molecular mass distribu-tion of the two polymers. The photochem-ical polymer contains, besides the two mainfractions corresponding to the two fractionsof the enzymatic polymer, two additionalfractions that are probably the main rea-son for the observed structural differences.The overall features of the UV absorptionspectrum, as well as 1H NMR and Ramanspectra of both polymers (Radotic, 1996;Radotic et al., 1998), show a general simi-larity, but also fine differences at the bondlevel.

Page 112: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

SCANNING PROBE MICROSCOPY ON LIGNIN 107

Figure 6.7. Schematic representation of lignin globulestructure. The scheme is step-wise blown-up fromglobule to the assumed molecular structure of thefirst stage of polymerization (module). Thick and thinlines represent hydrophilic and hydrophobic regions,respectively.

AFM study of intermolecular forces inphotochemical polymer confirms a higherrandomness to the structure. Contrary tothe enzymic polymer, photochemical poly-mers are nonfunctional (Micic et al., 2001b).Results from this experiment provide evi-dence for the previously stated hypothe-sis that photochemical lignin polymerizationmay be one of a degrading effect of UV radi-ation on plant cells.

A comprehensive STM and AFM studyof lignin model compounds and lignin hasbeen done by Shevchenko et al. (1996). Inthe paper, the milled wood lignins from birchand spruce, dioxane extracted lignin fromeucalyptus, DHP coniferyl alcohol-basedpolymeric model compound, and dimericlignin model compound 1-(4-hydroxy-phenyl)-2-(2-methodyphenoxy) ethanol wereimaged. In the same paper, examples of

humic acids were also examined, morespecifically soil fulvic acid and river fulvicacid. All samples were deposited from thesuspension as single droplets onto cleavedmice for AFM imaging, and STM imag-ing onto cleaved HOPG. The observationswere justified by applying molecular model-ing (SYBYL) to simulate observed motifs.Irregular aggregates of solid lignin com-pounds were observed using the AFM. Thesubstrate conductivity and hydrophobicitysignificantly influence the lignin morphol-ogy at the single molecular level, in bothlignin model compounds and in the extractedlignins. Molecular modeling of lignin form-ing the extended and flexible helices werealso performed. The observation of suchstructures has been later confirmed by Micicet al. (2000a,b) at the supramolecular levelusing the ESEM, at the 100 times higherorders of magnitude.

In order to avoid artifacts, AFM, in both thecontact and tapping mode, has been employedby Simola-Gustafsson et al. (Simola et al.,2000; Simola-Gustafsson et al., 2001). Sur-face of the suspended colloids from pulp wereanalyzed in different stages of pulping pro-cess—that is, with and without the oxygenand alkali delignification. In these studies,there is a granular surface observed, like onesseen in studies of lignin model compounds(Micic et al., 2000a,b). The smallest globu-lar structures, measuring 20 nm in diameter,correspond to the expected molecular weightof a single lignin macromolecule—that is, asupermodule, which is 55,000 Da.

Since lignin is a constituent of theinterfacial structure in a plant cell wall,there is the need to study lignins topo-graphical features in a simulated environ-ment. One way to achieve this is to mak-ing use of Langmuir–Blodgett films (Con-stantino et al., 2000; Pasquini et al., 2002).For example, Constantino et al. (2000) haveexamined Langmuir–Blodgett mono- andmultiple-layer films of either (a) pure ligninextracted from sugar cane bagasse or (b)composite of lignin and amphiphillic salt

Page 113: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

108 SCANNING PROBE MICROSCOPY OF PLANT CELL WALL AND ITS CONSTITUENTS

(cadmium-stearate). Even given the poor sta-bility of lignin monolayer, successful trans-fer to cleaved mica substrates have beenpossible. Langmuir–Blodgett film samples,ranging from monolayer to 21-fold lay-ers, have been investigated using the AFMimaging in both contact and tapping mode.The Langmuir–Blodgett film of pure ligninexpress a smooth flat topography. In com-posite Langmuir–Blodgett films, comprisedof cadmium-stearate amphiphile and lignin,there is phase separation, showing the clearseparate domains of lignin and amphiphile.Lignin domains have more or less globu-lar shape, with size similar to that of bulklignin.

Pasquini et al. (2002) have also performedstudies using Langmuir–Blodgett films oftwo different lignins, one cleaved of sugarsand the other saccharified. The clean ligninhas a flat surface, on L-B film morphology,while the saccharified forms irregular, roughsurfaces. Based on the combined AFM andFTIR data, it is concluded that pure ligninarranges parallel to the substrate, formingcompact film, while the saccharified ligninorients perpendicularly. Both lignins haverandom molecular organizations.

6.6. CONCLUSIONS

Scanning probe microscopies, especiallyAFM, is a powerful tool with unprecedentedspatial resolutions and is finding its wayinto the different aspects of plant sciences,ranging from fundamental molecular biol-ogy and biophysics, to the applied fields ofwood pulp, paper, and the food industry. Theunique ability of the AFM to unveil detailsat the molecular or at least supramolecu-lar level, without alternation of the sample,is providing a new opportunity for studiesof fragile biological structures, even in vivo,which was not possible with previous high-resolution structural methods such as theelectron microscopy. In this study, we havefocused only on the plant cell wall struc-tures, but nevertheless, AFM and other SPM

techniques have been used and will con-tinue to be used in much greater extent instudying other interesting aspects of plants,such as the photosynthetic center, structureof vacuoles, and the cell nuclei. In funda-mental studies of the plant cell wall struc-tures, we will witness in the near future sig-nificant advancements in our understandingof the complex structure of the plant cellwall. With further advent in design of themore user-friendly SPM setups, it is to beexpected that in the near future SPM tools,especially tapping-mode AFM, will becomea more common tool for the plant anatomist.Even more promising is the advancement ofAFM techniques, which are still in their earlystages of development but will enable simul-taneous topographic and chemically specificimaging of structures at the nanoscale level.This will be mainly achieved by some vari-ation of the near-field optical microscopy(NSOM), combined with the AFM, of whichmost promising candidates are a combina-tion of AFM and surface-enhanced Ramanspectroscopy (SERS) and fluorescence life-time imaging (FLIM) (Micic et al., 2002;Suh et al., 2002). It is to be envisioned thatthe applications of chemically specific imag-ing at the ultrastructural level in vivo willlead to a complete new understanding of the“metabolomics” of the plant cell. With thedevelopment of the high-throughput AFMmicroscope setups, it is expected that at somepoint AFM will probably be used as an ana-lytical tool in the everyday production andquality control in the pulp and paper indus-try, as well as in food-related industries.

REFERENCES

Baker, A., Helbert, W., Sugiyama, J., and Miles,M. (1997). High-resolution atomic forcemicroscopy of native Valonia cellulose Imicrocrystals. J. Struct. Biol. 119:129–138.

Bardinskaya, M. S., and Safonov, V. I. (1959).Izuchenie biosinteza lignina v izolirivannihtkanjah morkovi. Dokl. Akad. Nauk SSSR129:205–208.

Page 114: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 109

Bayer, E. A., Chanzi, H., Yamed, R., and Sho-ham, Y. (1998). Cellulose, cellulases and cellu-losomes. Curr. Opin. Struct. Biol. 8:548–557.

Bernardini, N., Penel, C., and Grepin, H. (1986).Preliminary immunological studies on horsera-dish and spinach peroxidase isoenzymes.In: Molecular and Physiological Aspects ofPlant Peroxidases (H. Greppin, C. Penel, andT. Gaspar, eds.), pp. 97–104, University ofGeneva.

Blankey, A. B., Harris, P. J., Henry, R. J., andStone, B. A. (1983). A simple and rapid prepa-ration of alditol acetates for monosaccharideanalysis. Carbohydrate Res. 113:291–299.

Bojkov, R. D. (1994). The ozone layer—recentdevelopments. World Meteorol. Org. Bull.43:113–116.

Boras, L., and Gatenholm, P. (1999). Surfaceproperties of mechanical pulps prepared undervarious sulfonation conditions and preheatingtime. Holzforschung 53:429–434.

Castillo, F. J. (1986). Extracellular peroxidasesas markers of stress? In: Molecular andPhysiological Aspects of Plant Peroxidases(H. Greppin, C. Penel, and T. Gaspar, eds.),pp. 419–426, University of Geneva.

Catesson, A. M., Imberty, A., Goldberg, R., andCzaninski, Y. (1986). Nature, localizationand specificity of peroxidases involved inlignification processes. In: Molecular andPhysiological Aspects of Plant Peroxidases(H. Greppin, C. Penel, and T. Gaspar, eds.),pp. 189–198, University of Geneva.

Chanzy, H., and Henrissat, B. (1985). Unidirec-tional degradation of Valonia cellulose micro-crystals subjected to cellulase action. FEBSLett. 184:285–288.

Chibbar, R. N., and van Huystee, R. B. (1986).Immunochemical localization of peroxidasein cultured peanut cells. J. Plant Physiol.123:477–486.

Conroy, J. M. (1986). Immunological probes ofrelationships among plant peroxidases: poly-clonal and monoclonal antibodies. In: Molec-ular and Physiological Aspects of Plant Per-oxidases (H. Greppin, C. Penel, and T. Gaspar,eds.), pp. 85–96, University of Geneva.

Constantino, C. J. L., Dhanabalan, A., Cotta,M. A., Pereira-daSilva, M. A., Curvelo,A. A. S., and Oliveira, O. N., Jr. (2000).Atomic force microscopy (AFM) investigation

of Langmuir–Blodgett (LB) films of sugar canebagasse lignin. Holzforschung 54:55.

Delucia, E. H., Day, T. A., and Vogelman, T. C.(1992). Ultraviolet-B and visible-light penetra-tion into needles of two species of sub-alpineconifers during foliar development. Plant CellEnviron. 15:921–929.

Duchesne, I., and Daniel, G. (1999). The ultra-structure of wood fibre surfaces as shown bya variety of microscopical methods—a review.Nordic Pulp Paper Res. J. 14:129.

Elder, T. J., and Worley, S. D. (1984). Theapplication of molecular orbital calculationsto wood chemistry. Wood Sci. Technol.18:307–315.

Erdtman, H. (1957). Outstanding problems inlignin chemistry. Ind. Eng. Chem. 49:1385–1386.

Falkehag, S. I. (1975). Lignin in materials. Appl.Polym. Symp. 28:247–257.

Fengel, V. D., Wegener, G., and Feckl, J. (1981).Beitrag zur Charakterisierung analytischerund technischer Lignine. Holzforschung 35:111–118.

Freudenberg, K., and Niedercorn, F. (1958).Umwandlung des phenylalanins in coniferinund fichtenlignin. Chem. Ber. 91:591–597.

Freudenberg, K., and Toribio, P. (1969). Derabbau des dehydrierungsproduktes aus coni-ferylalcohol-(14CH2OH) und des in gegenwartphenylalanin-(14CO2H) gewarhesenen lignins.Chem. Ber. 102:1312–1315.

Freudenberg, K., Harkin, J. M., Reichert, M., andFukuzumi, T. (1958). Die an der verholzungbeteiligten enzyme. Die dehydrierung dessinapinalkohola. Chem. Ber. 91:581–590.

Gaspar, T., Penel, C., Castillo, F., and Grep-pin, H. (1985). A two-step control of basicand acidic peroxidases and its significancefor growth and development. Physiol. Plant.64:418–423.

Goodwin, T. W., and Mercer, E. I. (1985). Intro-duction to Plant Biochemistry, Pergamon Press,Oxford.

Ha, M. A., Apperley, D. C., and Jarvis, M. C.(1997). Molecular rigidity in dry and hydratedonion cell walls. Plant Physiol. 115:593–598.

Harkin, J. M., and Obst, J. R. (1973). Lignifica-tion in trees: Indication of exclusive peroxidaseparticipation. Science 180:296–297.

Page 115: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

110 SCANNING PROBE MICROSCOPY OF PLANT CELL WALL AND ITS CONSTITUENTS

Hayashi, N., Sugiyama, J., Okano, T., and Ishi-hara, M. (1998). The enzymatic susceptibilityof cellulose microfibrils of the algal-bacterialtype and the cotton-ramie type. CarbohydrateRes. 305:261–269.

Hert, W., and Hausser, I. (1984). Chitin and cel-lulose fibrillogenesis in vivo and their exper-imental alteration. In: (W. M. Dugger andS. Bartnicki-Garcia, eds.), Structure, Func-tion and Biosynthesis of Plant Cell Walls,pp. 89–119, American Society of Plant Physi-ologists, Rockville, MD.

Higuchi, T., and Brown, S. A. (1963). Studiesof lignin biosynthesis using isotopic carbonXI. Reactions relating to lignification in youngwheat plants. Canad. J. Biochem. Physiol.41:65–76.

Hwang, R. H. (1982). An approach to lignifica-tion in plants. Biochem. Biophys. Res. Commun.105:509–514.

Hwang, R. H. (1985). A lignification mechanism.J. Theor. Biol. 116:21–44.

Imberty, A., Goldberg, R., and Catesson, A.(1985). Isolation and characterization ofPopulus isoperoxidases involved in the last stepof lignin formation. Planta 164:221–226.

Jurasek, L. J. (1995). Toward a 3-dimensionalmodel of lignin. J. Pulp Paper Sci. 21:J274.

Karmanov, A. P., Fillipov, V. N., and Mosk-vicheva, T. V. (1993). Issledovanie poverhnos-tnoi morfologiceskoi strukturi lignina. Khim.Drev. 1–3:123–127.

Kosikova, B., Zakutna, L., and Joniak, D. (1978).Investigation of the lignin–saccharidic com-plex by electron microscopy. Holzforschung32:15–18.

Kressig, H. A. (ed.) (1993). Cellulose. Structure,Accessibility and Reactivity. Gordon andBreach Science, Yverdon.

Laschimke, R. (1989). Investigation of the wetingbehaviour of natural lignin—a contributionto the cohesion theory of water transport.Thermochim. Acta 151:35–36.

Lee, I., Evans, B. R., Lane, L. M., and Wood-ward, J. (1996). Substrate–enzyme interac-tions in cellulase systems. Biosci. Technol.58:163–169.

Lee, I., Evans, B. R., and Woodward, J. (2000).The mechanism of cellulase action on cottonfibers: evidence from atomic force microscopy.Ultramicroscopy 82:213–221.

Lewis, N. G., and Yamamoto, E. (1990). Lignin:Occurrence, biogenesis and biodegradation.Annu. Rev. Plant Physiol. Plant Mol. Biol.41:455–496.

Marcinowski, S., and Grisebach, H. (1977).Turnover of coniferin in pine seedlings. Phy-tochemistry 16:1665–1667.

McCann, M. C., Wells, B., and Roberts, K.(1990). Direct visualization of cross-links inthe primary plant cell wall. J. Cell Sci.96:323–334.

Mehlhorn, H., and Kunert, K. J. (1986). Ascorbicacid, phenolic compounds and plant peroxi-dases: A natural defense system against per-oxidative stress in higher plants? In: Molecularand Physiological Aspects of Plant Peroxidases(H. Greppin, C. Penel, and T. Gaspar, eds.),pp. 437–449, University of Geneva.

Micic, M., Jeremic, M., Radotic, K., andLeblanc, R. M. (2000a). A comparative studyof enzymatically and photochemically poly-merized artificial lignin supramolecular struc-tures using environmental scanning elec-tron microscopy. J. Coll. Interface Sci.231:190–194.

Micic, M., Radotic, K., Jeremic, M., Mavers, M.,and Leblanc, R. M. (2000b). Visualizationof artificial lignin supramolecular structures.Scanning 22:288–294.

Micic, M., Benitez, I., Ruano, M., Mavers, M.,Jeremic, M., Radotic, K., Moy, V., and Le-blanc, R. M. (2001a). Probing the ligninnanomechanical properties and lignin–lignininteractions using the atomic force microscopy.Chem. Phys. Lett. 347:41–45.

Micic, M., Radotic, K., Benitez, I., Ruano, M.,Jeremic, M., Moy, V., and Leblanc, R. M.(2001b). Topographical characterization andsurface force spectroscopy of the photochem-ical lignin model compound. Biophys. Chem.94:257.

Micic, M., Klymyshyn, N., Suh, Y. D., and Lu,H. P. (2003). Finite element methods simula-tion of the field distribution for the AFM metal-lic tip enhanced SERS microscopy, J. Phys.Chem. B. 107:1574–1584..

Micic, M., Radotic, K., Jeremic, M., Djikanovic,D, Kammer, S. (2004). Study of the ligninmodel compound supramolecular structure bycombination of near-field scanning optical

Page 116: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 111

microscopy and atomic force microscopy. Coll.Surf. B: Biointerfaces 34:33.

Morris, V. J., Gunning, A. P., Kirby, A. R.,Round, A., Waldron, K., and Ng, A. (1997).Atomic force microscopy of plant cell walls,plant cell wall polysaccharides and gels. Int. J.Biol. Macromol. 21:61–66.

Moy, V. T., Florin, E. L., and Gaub, H. E.(1994). Intermolecular forces and energiesbetween ligands and receptors. Science266:257.

Niemi, H., Paulapuro, H., and Mahlberg, R.(2002). Application of scanning probe micros-copy to wood, fiber and paper research. Paperi,Ja. Puu-Paper and Timber 84:389–406.

Nozu, Y. (1967). Studies on the biosynthesis oflignin. J. Biochem. 62:519–530.

O’Sullivan, A. C. (1997). Cellulose: The struc-ture slowly unravels. Cellulose 4:173–207.

Pandolfini, T., Gabrielli, R., and Comparini, C.(1992). Nickel toxicity and peroxidase activityin seedlings of Triticum aestivum L. Plant CellEnviron. 15:719–725

Pasquini, D., Balogh, D. T., Antunes, P. A., Con-stantino, C. J. L., Curvelo, A. A. S., Aroca,R. F., and Oliviera, O. N. (2002). Surfacemorphology and molecular organization oflignins in Langmuir–Blodgett films. Langmuir18:6593.

Pickett-Heaps, J. D. (1968). Xylem wall depo-sition. Radioautographic investigations usinglignin precursors. Protoplasma 3:55–64.

Polle, A., and Chakrabarti, K. (1994). Effects ofmanganese deficiency on soluble apoplasticperoxidase activities and lignin content inneedles of Norway spruce (Picea abies). TreePhysiol. 14:1191–1200.

Radotic, K. (1994). Biophysical and biochemicalcharacterization of the structure and functionof the cell walls from maize (Zea mays L.).Doctorial thesis. University of Belgrade (inSerbian).

Radotic, K. (1996). Enzymatic and photochemicalpolymerization of coniferyl alcohol: A kineticand structural study. Polyphenol Commun.96:497–498.

Radotic, K., Simi-Krsti, J., Jeremi, M., and Tri-funovi, M. (1994). A study of lignin formationat the molecular level by scanning tunnelingmicroscopy. Biophys. J. 66:1763–1767.

Radotic, K., Zakrzewska, J., Sladi, D., andJeremi, M. (1997). Study of photochemicalreactions of coniferyl alcohol. I. Mechanismand intermediate products of UV radiation-induced polymerization of coniferyl alcohol.Photochem. Photobiol. 65:284–291.

Radotic, K., Budinski-Simendic, J., and Jeremic,M. (1998). A comparative structural study ofenzymic and photochemical lignin by scanningtunneling microscopy and molecular massdistribution. Chem. Ind. 52:347–350.

Ridley, B. L., O’Neill, M. A., and Mohnen,D. A. (2001). Pectins: Structure, biosynthesis,and oligogalacturonide-related signaling. Phy-tochemistry 57:929–967.

Roelofsen, P. A. (1965). Ultrastructure of the wallin growing cells and its relation to the directionof the growth. Adv. Bot. Res. 2:69–149.

Round, A. N., Kirby, A. R., and Morris, V. J.(1996). Collection and processing of AFMimages of plant cell walls. Microscopy andAnalysis, Sept:33–35.

Round, A. N., McDougall, A. J., Ring, S. G.,and Morris, V. J. (1997). Unexpected branch-ing in pectin observed by atomic forcemicroscopy. Carbohydrate Res. 303:251–253.

Round, A. N., Rigby, N. M., MacDougall, A. J.,Ring, S. G., and Morris, V. J. (2001). Inves-tigating the nature of branching in pectinby atomic force microscopy and carbohydrateanalysis. Carbohydrate Res. 331:337–342.

Schmid, G., and Grisebach, H. (1982). Enzymicsynthesis of lignin precursors. Purificationand properties of UDPglucose: Coniferylalcohol glucosyltransferase from cambial sapof spruce (Picea abies L.). Eur. J. Biochem.123:363–370.

Schmid, G., Hammer, D. K., Ritterbusch, A., andGrisebach, H. (1982). Appearance and immu-nohistochemical localization of UDP-glucose:coniferyl alcohol glucosyltranferase in spruce(Picea abies (L.) Karst) seedlings. Planta156:207–212.

Schubert, W. J., and Nord, F. F. (1957). Mech-anism of lignification. Ind. Eng. Chem.49:1387–1390.

Shann, J. R., and Blum, U. (1987). The utilisationof exogenously supplied ferulic acid in ligninbiosynthesis. Phytochemistry 26:2977–2982.

Shevchenko, S. M., and Bailey, G. W. (1996).Nanoscale morphology of lignins and their

Page 117: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

112 SCANNING PROBE MICROSCOPY OF PLANT CELL WALL AND ITS CONSTITUENTS

chemical transformation products. Tappi 79:227.

Simola, J., Malkavaara, P., Alen, R., and Ple-tonen, J. (2000). Scanning probe microscopyof pine and birch kraft pulp fibers. Polymer41:21.

Simola-Gustafsson, J., Hortling, B., and Pleto-nen, J. (2001). Scanning probe microscopy andenhanced data analysis on lignin and elementalchlorine-free or oxygen delignified pine kraftpulp. Coll. Polym. Sci. 279:221.

Stewart, A., Brown, K., Tanner, G., and Stone,J. E. (1953). Studies of lignin biosynthe-sis using isotopic carbon II. Short-termexperiments with C14O2. Canad. J. Chem.31:755–760.

Stone, J. E. (1953). Studies of lignin biosynthesisusing isotopic carbon I. Long-term experimentwith C14O2. Canad. J. Chem. 31:207–213.

Stout, G. H., and Jensen, L. H. (1968). X-RayStructure Determination, Macmillan, NewYork.

Suh, Y. D., Schenter, G. K., Zhu, L., and Lu,H. P (2003). Probing nano-surface enhancedRaman scattering fluctuation dynamics usingcorrelated AFM and confocal microscopy.Ultramicroscopy 97:89–94.

Varner, J. E., and Lin, L.-S. (1989). Plant cellwall architecture. Cell 56:231–239.

Wayman, M., and Obiaga, T. I. (1974). Themodular structure of lignin. Can. J. Chem.52:2102–2111.

Zakutna, L., Kosikova, B., and Eberingerova A.(1983). Partial alkaline delignification of spurcewood. IV Electron microscopy of the ligninand hemicellulose fractions isolated from pulp.Dervarsky Vyskum 28:15.

Page 118: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

CHAPTER 7

CELLULAR INTERACTIONS OF NANO DRUGDELIVERY SYSTEMSRANGARAMANUJAM M. KANNANDepartment of Chemical Engineering and Material Science, and Biomedical Engineering, WayneState University, Detroit, Michigan

OMATHANU PILLAI PERUMALDepartment of Pharmaceutical Sciences, South Dakota State University College of Pharmacy,Brookings, South Dakota

SUJATHA KANNANDepartment of Critical Care Medicine, Children’s Hospital of Michigan, Detroit, Michigan

7.1. INTRODUCTION

The deciphering of the human genome hasopened up multiple avenues for identifyingnew targets and developing novel therapies(Drews, 2000). With the availability of aplethora of genomic data and high-throughputscreens, there is a compelling need to translatethis “new knowledge” into successful thera-pies. Intelligent design of drug delivery sys-tems (DDS) to meet this need requires a thor-ough understanding of the factors influenc-ing the disease, drug, destination and deliv-ery (Fig. 7.1). Drug delivery scientists havebeen successful in achieving spatial and tem-poral control of drug from DDS, but thepath for transport of drug molecules fromthe delivery system to the site of actionremains largely uncontrolled. Targeting andreleasing the drug as close as possible tothe site of action can fulfill this objective.The efficient functioning and regulation ofthe biological system are strictly controlled

by structural hierarchy varying from centime-ter to nanometer scales (Esfand and Toma-lia, 2001). Many of the bioassemblies andendogenous molecules (such as enzymes, hor-mones and nucleic acids) exist and functionat the nanoscale level. Majority of the ther-apeutic targets exist intracellularly or at thesurface of the cells. Hence the goal of thedrug delivery system is to deliver drugs tothe intended target with minimal interferencewith normal cellular functioning and with theleast systemic side effects.

With the development and use of forcemicroscopy and advances in nanotechnologywe have an opportunity to design andassemble DDS at nanoscale dimension todeliver the payload at the required targetsite at the appropriate time. By engineeringnanoscale drug delivery vehicles to mimicthe biological system, it is possible to achievedifferent degrees of targeting from organ totissue and eventually at the cellular level.

Force Microscopy: Applications in Biology and Medicine, edited by Bhanu P. Jena and J.K. Heinrich Horber.Copyright 2006 John Wiley & Sons, Inc.

113

Page 119: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

114 CELLULAR INTERACTIONS OF NANO DRUG DELIVERY SYSTEMS

PathophysiologyAcute/Chronic

Molecularmechanism

Identify targets

Molecular wtDose

ChargeLipophilicity

Systemic/localTargeting ligands

Route ofadministration

Release kineticsPharmacokinetics

EfficacyBiocompatibility

DELIVERY

DISEASE DRUG DESTINATION

Figure 7.1. Four-dimensional approach to design of drug delivery. The clinical need based on pathophysiology ofthe disease is the major driving force for developing new drug delivery systems. “Smart” drug delivery systems canmaximize the therapeutic efficacy of the drug by targeting it to the desired destination.

A better appreciation of the natural celltrafficking mechanisms in cells has helpedin the design of nanoscale DDS targetedto specific transport pathways. Based onthe location and function of cells, severalspecific transport mechanisms have beendeveloped to meet the essential nutrientrequirements of the intracellular organellesand also “kick out” unwanted foreign bod-ies and toxins to the extracellular medium(Fig. 7.2). Apart from simple diffusion ofvery small molecules, cells have evolvedvarious mechanisms to internalize smallmolecules, macromolecules, and particlesand target them to specific organelles withinthe cytoplasm. Collectively, these processesare referred to as “endocytosis” (De Duve,1963), which includes (i) phagocytosis or celleating, (ii) pinocytosis or cell drinking, (iii)

clathrin-dependent receptor-mediated endo-cytosis, and (iv) clathrin-independent recep-tor-mediated endocytosis.

Phagocytosis, which involves the uptakeof large particles (>300 nm) takes placein many cells and most importantly inspecialized cells such as macrophages, whereit plays an important role in cellular immu-nity. Macropinocytosis is a bulk internal-ization process and is seen in a num-ber of phagocytic and nonphagocytic cells(Johannes and Lamaze, 2002; Amyere et al.,2002). Receptor-mediated endocytosis takesplace in all nucleated vertebrate cells andplays an important role in many of thephysiological processes. Clathrin-coated pitsare ∼150-nm invaginated structures of theplasma membrane varying from 0.4% ofmembrane surface area in adipocytes to 3.8%

Page 120: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

INTRODUCTION 115

LE

RE

G

EE

C

LY

N

L

L

N P C

Figure 7.2. Various cell trafficking pathways. C, clathrin-coated pits; NC, non-clathrin pits; P, phagocyto-sis/macropinocytosis; CE, caveolae; EE, early endosome; LE, late endosome; LY, lysosome; L, ligand; N, nucleus;G, Golgi apparatus; RE, recycling endosomes. The pH in the endosomes is 5.9–6.0, while the lysosomal pH is5.9–5.0. The broken arrows represent the escape of the ligand from the endososmes or from the lysosomes.

in some human fibroblasts (Goldberg et al.,1987; Nilsson et al., 1983). Some receptorsare constitutively concentrated in the coatedpits (low-density lipoprotein receptor), butothers become concentrated upon ligand bind-ing (epidermal growth factor). By electronmicroscopy, caveolae appear as ∼70-nm-diameter flask-shaped plasmalemmal invagi-nations formed through the oligomerization ofstructural coat protein caveolin (Peters et al.,1985). They are present on many cell typesincluding fibroblasts, cardiomyocytes, andadipocytes mediating endocytosis to delivera variety of molecules to intracellular com-partments including endoplasmic reticulum,golgi apparatus, endosomes, and lysosomes(Schnitzer, 2001).

After internalization, the molecules aredegraded and recycled back to the cell sur-face. Endoctyosis presents the ligand to theearly endosomes and then to late endo-somes, where the pH drops to 5.9–6.0. Thenthe molecules progress to lysosomes, wherethe pH further reduces from 6.0 to 5.0. Inaddition, the lysosomes contain enzymes to

breakdown the cargo. Alternative fates suchas trafficking to organelles like golgi appara-tus or translocation into cytosol also occur.The receptors are recycled back to the cellsurface by the recycling endosomes. Cellulartrafficking is a complex process (Mukher-jee et al., 1997) that depends on physico-chemical properties of internalized molecules(ligands, pH optima of ligand-carrier dis-sociation) and the biological characteristicsof the cell organelles that carry them (size,shape, luminal pH, lipids, and composition).Based on the intracellular targets and theunique functions of the target cells, nanoscaledrug delivery devices that would exploit var-ious cellular trafficking mechanisms can bedesigned such that the drug is delivered tothe specific intracellular site.

Pathophysiological conditions such ascancer present certain unique characteristicsthat are not observed in normal tissue, includ-ing (i) extensive angiogenesis, (ii) defective(leaky) vascular architecture, (iii) impairedlymphatic drainage, and (iv) increased pro-duction/expression of permeability mediators

Page 121: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

116 CELLULAR INTERACTIONS OF NANO DRUG DELIVERY SYSTEMS

on the cell surface (Brigger et al., 2002).These characteristics may also help in con-centrating the nanoscale drug delivery sys-tems at the site of the tumor.

By tagging suitable ligands to the nano-vehicles, the drug cargo can be selectivelytargeted to the diseased cell. On the otherhand, by using organelle-specific molecularprobes and imaging techniques, the kineticsand location of nanovehicle can be followedto understand and validate the nanodrugdelivery systems for intracellular drug deliv-ery. This chapter discusses how the variousnanovehicles can be engineered by manipu-lating the chemistry and the physicochemicalproperties of the vehicle to selectively andspecifically interact with the biological sys-tem (Fig. 7.3), for achieving improved ther-apeutic efficacy and clinical outcome. Thefocus of this chapter is restricted to mainlylipid-based and polymer-based nanovehicles.To highlight the importance of size withinthe nanometer scale, nanovehicles varyingin size from 1 nm (dendrimers) to 1000 nm(liposomes) have been discussed. However,the major emphasis is given on dendrimer-based drug delivery systems because theseare the smallest of the nanoscale deliverysystems and have been used as a case study

with two model drugs (ibuprofen and methyl-prednisolone) to demonstrate the chemistry,cell entry dynamics, and biological activityof the nanovehicle (dendrimer) per se and inpresence of the drug as a conjugate.

7.2. LIPID-BASED SYSTEMS

7.2.1. Conventional Liposomes

Liposomes are probably one of the earli-est known nanosized drug delivery vehi-cles and are made of biodegradable lipidvesicle (phospholipids) with aqueous phasesurrounded by a lipid bilayer. Liposomeshave been used to entrap a wide varietyof drug substances. Water-soluble drugs areentrapped in the aqueous compartment, whilethe lipophilic substances get incorporated inthe lipid bilayer. Depending on the lipids,preparation method, and number and posi-tion of lipid lamellae, liposomes can varywidely in size (10–1000 nm) (Barenholz andCrommelin, 1994). They are rapidly takenup by the reticuloendothelial system fromthe circulation and hence can be used forpassive targeting to the lymphatic system.The size of the liposomes is the main factorcontrolling the lymphatic uptake, and small

Dendrimers (5−15 nm)

Micelles (50−80 nm)

Liposomes (10−1000 nm)

Pharmacokinetics

MEDICINE

Trafficking

Size Ligand Physiology Target Efficacy Toxicity

BIOLOGY

Hydrophobicity

CHEMISTRY

Nanoparticles (100−400 nm)

Figure 7.3. The interaction of chemistry and biological principles in the design of nanodrug delivery systems formedicine.

Page 122: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

LIPID-BASED SYSTEMS 117

liposomes seem to achieve high lymphaticuptake (Oussoren and Storm, 1997; Ous-soren et al., 1997). Similarly, surface chargeand the route of administration also governthe lymphatic delivery of drugs (Kim andHan, 1995). Phagocytosis by macrophagesplays an important role in the cellular uptakeof liposomes. Alternate pathways of cellularentry of liposomes include fusion with thecell membrane, charge interaction of cationicliposomes with the membrane, endosomaluptake, and release of water-soluble contentsinto the cytoplasm (Felgner and Ringold,1989).

Liposomes have been widely studied asa vehicle for targeting antitumor agents.The amount of liposomes accumulating inthe tumor is dictated by (i) residencetime in blood, (ii) transfer from blood totumor interstitial spaces, (iii) local reten-tion of liposomes in tumor tissue, and(iv) efflux from the tumor tissue to blood(Nagayasu et al., 1999). In tumor tissue,

the permeability of capillary endotheliumis enhanced, while the lymphatic system ispoorly developed in comparison to normaltissues, and this phenomenon is termed asenhanced permeability and retention (EPR)effect (Fig. 7.4). This altered physiologyhas been exploited for passively targetingliposome-encapsulated drugs to the tumor(Jain, 1987). Smaller, long-circulating lipo-somes (∼100 nm) have higher frequency ofextravasation into tumor tissue through theleaky vasculature and are retained in thetumor longer due to the compromised lym-phatic drainage in the rapidly dividing cells.The size of the liposomes also plays a role indictating its blood residence time, as largerparticles are rapidly cleared by macrophagesby opsonization of plasma proteins. It hasbeen reported that 100 nm represents an opti-mal size for blood to tumor transfer andfor longer retention and biodistribution intumor (Nagayasu et al., 1996). At the sametime, the lipid composition is also critical for

Healthy Tissue

Tumor

Lymphatic Drainage

Lymphatic DrainageX

Figure 7.4. Schematic representation of enhanced permeability effect (EPR) in the tumor tissue in comparison tothe healthy tissue.

Page 123: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

118 CELLULAR INTERACTIONS OF NANO DRUG DELIVERY SYSTEMS

controlling drug release in addition to pro-longing the circulation half-life of liposomes.

7.2.2. Modified Liposomes

Drug delivery by conventional liposomesis severely limited by their rapid detectionand blood clearance by the macrophages.This, in addition to reducing the efficacyof the drugs, also poses potential toxic-ity to the macrophages and causes bonemarrow suppression (Senior, 1987; Daemanet al., 1997). Surface modification of lipo-somes with polyethylene glycol (PEG) orother selective hydrophilic polymers offers anumber of advantages in this regard (Woodle,1998); (i) Prolonged circulation is indepen-dent of liposomal bilayer, hence drug load-ing and release can be adjusted independentfrom blood circulation; (ii) pharmacokinet-ics and tissue distribution now becomes lipidindependent, and therefore drug loading canbe increased; (iii) ligands with other func-tionality can be attached to the polymer foractive targeting. The modified liposomes arereferred to as “stealth” or “sterically sta-bilized” liposomes, which reduces proteinadsorption and consequently cellular inter-actions (Liu, 1997). This results in reducedblood clearance of liposomes and concomi-tant changes in tissue distribution. When con-ventional liposomes are used for intracellulardelivery of macromolecules such as proteinsand nucleic acids, the cargo is inactivatedby the acidic environment and enzymes inlysosomes before reaching the site of actionin the cytosol. Incorporation of lipids thatundergo phase transformation in responseto a pH stimuli have been used to deliverthe contents intact to the cytosol (Hafezand Cullis, 2001). Dioleoyl phosphatidylethanolamine (DOPE) is a nonbilayer lipidthat is used in the preparation of so-called“fusogenic liposomes.” It derives its namedue to its ability to undergo lamellar tohexagonal phase transformation in responseto acidic pH stimulus (Felgner et al., 1987).Following endocytosis, pH-sensitive lipo-somes undergo destabilization and fuse with

the endosomal membrane upon encounteringacidic pH (5–6.5) in the endosomes. Themembrane fusion helps in releasing the lipo-somal contents into the cytosol, therebyavoiding the degradation in the lysosomes(Fig. 7.5). Cationic lipid–DNA lipoplexesare commercially available as lipofectin,which contains a mixture of two cationicliposomes namely 2,3-bis(oleoyl)oxipropyltrimethyl ammonium chloride (DOTMA)and DOPE. This showed improved DNAtransfection in several cells (de Lima et al.,2001). However, the cationic lipids causeformation of aggregates by interacting withthe negatively charged serum proteins, henceaffecting the biodistribution of liposomes intissues (Zhu et al., 1993).

A novel fusogenic liposome known as“virosome” (Kaneda et al., 1999) was con-structed using a fusogenic envelope derivedfrom UV-inactivated heamaglutinating virusof Japan (HVJ; Sendai virus). As viro-somes, liposomes acquire viral functionsand can fuse with almost all cells exceptperipheral lymphocytes. Fusion is completewithin 10–30 min at 37◦C as opposedto 5–20 h with cationic liposomes. Theunique characteristics of virosomes havebeen studied using confocal microscopy.Cytoplasm was found to be stained withFITC-dextran, when delivered by fusogenicliposomes, while aggregated fluorescenceindicating endosomal presence in cells cul-tured with FITC-dextran-encapsulated con-ventional liposomes (Kunisawa et al., 2001).The endocytosis inhibitor (cytochalesin B)did not affect the delivery of virosomes,indicating that virosomes delivered theircontents into the cytosol via an endocytosis-independent pathway by fusing directly withthe cell membrane. Similarly, the deliveryof nucleic acid by virosomes to the nucleushas been demonstrated using FITC-labeledoligonucleotide, where within 5 min fluores-cence was observed in the nucleus and wasstable for 72 h (Morishita et al., 1994).

Liposomes that respond to thermal stim-ulus can be designed by suitable choice of

Page 124: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

LIPID-BASED SYSTEMS 119

Endocytosis

Endosome

Lysosome

Nucleus

Figure 7.5. Schematic representation of the intracellular fate of conventional (left-hand panel; green) and fusogenicliposomes (right-hand panel; red). The fusogenic liposomes release the contents, when there is a changes in the lipidphase in the acidic pH environment of the endosome.

lipid that undergoes gel-to-sol transition inresponse to temperature. Alternatively, poly-(N -isopropyl acrylamide) [poly(NIPAM)],which has a lower consolute solution temper-ature, can be attached on the liposome sur-face. Such liposomes can be used to selectivelyrelease the drug contents in the tumor in con-junction with hyperthermia (Kono, 2001).

To achieve specific cellular targeting, var-ious ligands have been attached to liposomes,including peptide-based ligands, sugar-basedligands, or glycoproteins. These ligand-decorated liposomes utilize the receptor-mediated endocytosis to get internalizedinto the cell. Galactosylated liposomes aretaken up selectively by asialoglycoproteinsreceptor-positive liver parenchymal cells,while mannose residues resulted in specificdelivery of genes to nonparenchymal livercells (Hashida et al., 2001).

A very attractive tumor-selective ligandis folate receptor, which is absent in mostnormal tissues but elevated in over 90%of ovarian carcinomas and expressed at ahigh frequency in other human malignancies.As a low-molecular-weight (MW 441) lig-and, folate offers numerous advantages overpolypeptide based targeting ligands: (i) lackof immunogenicity, (ii) unlimited availabil-ity, (iii) functional stability, (iv) high affin-ity for the folate receptor (Kd ∼ 10−10 M),(v) defined conjugation chemistry, and (vi)favorable nondestructive cellular internal-ization pathway (Brzezinska et al., 2000;Michael and Curiel, 1994). It has beenshown that folate-receptor-targeted liposomaloligonucleotide exhibited greater efficacy insuppressing cellular growth and inhibition ofepidermal growth factor receptor expression

Page 125: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

120 CELLULAR INTERACTIONS OF NANO DRUG DELIVERY SYSTEMS

compared to free oligonucleotide and nontar-geted liposomal oligonucleotide (Wang andLow, 1998).

7.3. POLYMER-BASED SYSTEMS

7.3.1. Nanoparticles

Nanoparticles are submicron-sized (<1000nm) systems, where the drug is dissolved,entrapped, or encapsulated and/or to whichthe drug is adsorbed or attached. Accord-ing to the method of preparation, eithernanospheres or nanocapsules can be ob-tained. Nanocapsules are vesicular systems inwhich the drug is confined to an oily or aque-ous core surrounded by a unique polymericmembrane (Fig. 7.6). On the other hand,nanospheres are matrix systems in whichthe drug is uniformly dispersed (Fig. 7.6).Nanoparticles offer advantages over lipo-somes, because they are more stable andare more efficient drug carriers than lipo-somes (Fattal et al., 1991). Conventionally,nanoparticles are prepared mainly by twomethods: (i) dispersion of preformed poly-mers and (ii) polymerization of monomers.Recently, supercritical fluids have becomeattractive alternatives to conventional meth-ods, because the particles can be preparedin high purity without any trace organicsolvents (Randolph et al., 1993). A vari-ety of natural and synthetic polymers havebeen used for preparing nanoparticles. Syn-thetic polymers have the advantages ofsustaining the release of encapsulated agent

Nanocapsule Nanosphere

Figure 7.6. A comparative representation of nanocap-sule and nanosphere. The blue layer in the nanocapsulerepresents encapsulated drug while drug is interdis-persed with the polymer matrix in nanosphere.

over a period of days to several weekscompared to natural polymers, which havea relatively short duration of drug release(Soppimath et al., 2001). Nanoparticles canbe made from synthetic polymers such aspolylactide–polyglycolide copolymers, poly-acrylates, and polycaprolactones or naturalpolymers such as albumin, gelatin, alginate,collagen, and chitosan. Of these polymers,polylactides (PLA) and poly(DL-lactide-co-glycolide) (PLGA) have been the most exten-sively investigated for drug delivery. Thispolyester polymer undergoes hydrolysis inthe body forming biologically compatibleand metabolizable moieties (glycolic acid,lactic acid), which are eventually eliminatedfrom the body by citric acid cycle.

Drug loading in the nanoparticles can beachieved by either incorporating drug at thetime of nanoparticle production or by adsorb-ing drug to the preformed nanoparticles byincubating them in drug solution (Alensoet al., 1991; Ueda et al., 1998). The for-mer method results in greater drug loadingthan the latter. Adsorption capacity is relatedto the hydrophobicity of the polymer andthe specific surface area of the nanoparti-cles. In the case of the drug entrapmentmethod, the monomer amount needs to beoptimized for achieving high drug load (Rad-wan, 1995).

Surface active agents and stabilizers addedduring polymerization have an effect ondrug loading apart from influencing parti-cle size and molecular mass of nanoparticles(Egea et al., 1994). Yoo et al. (1999) havereported that the encapsulation efficiency canbe improved by chemically conjugating tonanoparticles. For doxorubicin, an encapsu-lation efficiency of 97% was observed asopposed to 6.7% for unconjugated doxoru-bicin in PLGA nanoparticles.

Release rate of drugs from nanoparticlesdepends upon a number of factors includ-ing (i) desorption of surface bound/adsorbeddrug; (ii) diffusion through the nanoparticlematrix; (iii) diffusion, in the case of ananocapsule through the polymer wall;

Page 126: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

POLYMER-BASED SYSTEMS 121

(iv) nanoparticle matrix erosion; and (v)combined erosion/diffusion process. Thusdiffusion and biodegradation govern the drugrelease from nanoparticles (Soppimath et al.,2001). The degradation of the polymer can bevaried by altering the block copolymer com-position and molecular weight, to tune therelease of the therapeutic agent from days tomonths (Lin et al., 2000). Nanoparticles areefficiently internalized through an endocyticprocess, and the uptake is both concentration-and time-dependent (Panyam et al., 2002;Panyam and Labhasetwar, 2003a; Davdaand Labhasetwar, 2002). In vascular smoothmuscles, PLGA nanoparticle internalizationhas been shown in part to take placethrough fluid-phase pinocytosis and in partthrough clathrin-coated pits (Foster et al.,2001; Suh et al., 1998). Following theirinternalization into the cells, nanoparticleswere observed to be transported to primaryendosomes and then probably to sortingendosomes. The remaining fraction is trans-ported to secondary endosomes, which thenfuse with the lysosomes (Panyam and Lab-hasetwar, 2003b). Time-dependent studieshave shown that nanoparticles escaped theendolysosomes within 10 min of incubationand entered the cytosol. From transmissionelectron microscopy, Panyam and Labhaset-war (2003a) found that PLGA nanoparticlesundergo charge reversal (to cations) in theacid environment of the endosomes leadingto localized destabilization of the membraneand escape of nanoparticles into the cytosol.This was further substantiated from stud-ies with polystyrene, where no endolyoso-mal escape was observed, because thesenanoparticles do not undergo charge reversalwith change in pH. Furthermore, the authorsobserved exocytosis of PLGA nanoparticles,when the external concentration gradient ofnanoparticles was removed. Probably, thealbumin adsorbed onto nanoparticles inter-acted with the exocytic pathway leading to anincrease in exocytosis (Tomoda et al., 1989).However, a fraction of the dexamethasone

nanoparticle retained in the cell demon-strated sustained anti-proliferative action for2 weeks as opposed to 3 days for drug insolution (Panyam and Labhasetwar, 2003a).Since PLGA nanoparticle can bypass thelysosomal compartment, it could serve as anattractive carrier for sustained gene expres-sion (Labhasetwar et al., 1999). By vary-ing the surface charge of nanoparticle, it ispossible to target nanoparticles to specificintracellular cell organelles. Surface mod-ification of biodegradable and long-actingpolymeric nanoparticles can be achieved by(i) surface coating with hydrophilic poly-mers/surfactants and (ii) development ofbiodegradable copolymers with hydropho-bic segments. Some of the surface coat-ing materials include polyethylene glycol,polyethylene oxide, poloxamer, poloxam-ine, and polysorbate 80. Surface coatinghas been found to reduce opsonizationby plasma proteins and improve the cir-culation time of nanoparticles (Soppimathet al., 2001). Polysorbate-coated poly(butylcyanoacrylate) nanoparticle showed en-hanced drug transport of anti-inflammatorydrug dalagrin (Alyautdin et al., 1995) intothe brain by a number of mechanisms:(i) binding of nanoparticles to the innerendothelial lining of the brain capillariesand subsequently particles deliver drugs tothe brain by providing a large concentra-tion gradient, thus enhancing passive dif-fusion; and (ii) brain endothelial uptakeby phagocytosis. Physicochemical proper-ties of nanoparticles—namely, size, surfacecharge, hydrophobicity and release charac-teristics—can be varied by altering the com-position or the method of preparation ofnanoparticles to achieve targeted and sus-tained drug delivery to various tissues andcells.

7.3.2. Polymeric Micelles

Block copolymers with amphiphilic charac-ters exhibiting a large solubility differencebetween hydrophilic and hydrophobic seg-ments self-assemble in an aqueous milieu

Page 127: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

122 CELLULAR INTERACTIONS OF NANO DRUG DELIVERY SYSTEMS

into polymeric micelles with nanoscopicdimensions (Tuzar and Kratochvil, 1976).These micelles have a fairly narrow sizedistribution and are characterized by theirunique shell-core architecture, where hydro-phobic segments are segregated from theaqueous exterior to form an inner core sur-rounded by a palisade of hydrophilic seg-ments. Polymeric micelles show distinctadvantages over surfactant-based micellessuch as greater solubilization capacity, betterkinetic stability, and lesser toxicity (Lavasan-ifar et al., 2002).

Polymeric micelles mimic aspects of bio-logical transport systems (lipoproteins andviruses) in terms of structure and function.The hydrophilic shell helps them remainunrecognized by the blood circulation (Dunnet al., 1994). At the same time, the viral-like size (10–50 nm) prevents their uptakeby reticulo-endothelial system (RES) andfacilitates their extravasation at leaky cap-illaries for passive targeting to tumors.Poly(ethylene oxide) (PEO) is usually thehydrophilic shell, and others include poly(N -isopropyl acrylamide). A hydrophobic blockincludes poly(α-hydroxy acids) and poly(L-amino acids). Unlike the shell-forming block,the choice for the core-forming block is rel-atively diverse. Therefore, by suitable choiceof the block copolymers the micelles can beconstructed to respond to thermal or pH stim-uli (Jeong et al., 1997; Tailefer et al., 2000).Targeting ligands may dot the surface ofblock copolymers for site-specific delivery.

Micellar cores serve as a nanoreservoir forloading and controlled release of hydropho-bic molecules that are conjugated or com-plexed with the polymeric backbone or phys-ically encapsulated in the core (Kwon, 1998;Allen et al., 1999; Kataoka et al., 2001).For drugs physically encapsulated in thecore of the micelle, release is controlledby the rate of drug diffusion in the micel-lar core or breakup of the micelles. Inthe case of drug conjugation, the covalentbond between the drug and the polymerhas to be cleaved for drug release. Water

penetration and hydrolysis of the labile bondsin the micelles core (bulk erosion) followedby drug diffusion may occur in relativelyhydrophobic liquid-like core structures. Inthe case of hydrophobic rigid cores, releaseis dependent on rate of micellar dissoci-ation. Slow dissociation of micellar struc-ture and further hydrolysis of labile bondsmay result in a sustained drug release (Liand Kwan, 2000). The multifunctional natureof polymeric micelles makes it a versatiledrug carrier capable of achieving selectiveand sustained drug delivery at different lev-els. Micelles based on PEO-b-poly(L-aminoacids) (PLAA) block copolymers are uniqueamong drug carriers owing to a tailor-madenonpolar core of PLAA, which can takeup and protect water insoluble drug. Thesingle greatest advantage of PEO-b-PLAAover other micelle forming block copoly-mers is its potential for attaching drugs andgenes through the functional groups of theamino acid side chain (amine or carboxylicacid). Three types of nanodrug delivery sys-tems based on these micelles have beeninvestigated including (i) micelle formingblock copolymer–drug conjugate, (ii) micel-lar nanocontainer, and (iii) polyion complexmicelles. These strategies have been used todeliver antitumor agents and nucleic acids,where the block copolymer micelle protectsthe drug in the circulation and delivers it tothe intracellular targets in a sustained manner(Kakizawa and Kataoka, 2002).

7.3.3. Dendrimers

7.3.3.1. Introduction. Dendrimers arehighly branched, well-defined, polymericnanostructures that have recently been ex-plored as potential drug carriers (Liu andFrechet, 1999; Tomalia et al., 1985, 1986;Hawker and Frechet, 1990; Stiriba et al.,2002; Gebhart and Kabanov, 2001; Cloninger, 2002; Liu et al., 2000; Kolhe et al.,2003; Kono et al., 1999; Zhuo et al., 1999;Padilla De Jesus et al., 2002; Ihre et al.,2002; Malik et al., 2000; Wiwaattanapatapee

Page 128: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

POLYMER-BASED SYSTEMS 123

et al., 2000; Kukowska-Latallo et al., 1996;Tajarobi et al., 2001; El-Sayed et al., 2002;Kannan et al., 2004; Kohle et al., 2004a,b;Khandare et al., 2004). The initial pioneer-ing work by Tomalia and Newkome in the1980s has paved the way for a numberof research groups to contribute substan-tially in different methodologies for synthesisand specific applications of these dendrimers(Tomalia et al., 1985, 1986; Hawker et al.,1990). Due to their unique structure andproperties, they have been evaluated formany applications, such as nanoscale cata-lysts, micelle mimics, encapsulation of drugmolecules, biological recognition, chemicalsensors, immunosensors in medical imag-ing, and drug/gene delivery (Fig. 7.7). Den-drimers are unimolecular micelles, whichare much smaller (∼10 nm) and morestable than liposomes. Dendritic polymers(size ∼10 nm) are emerging as promis-ing candidates for “smart” delivery nanode-vices, because they present a large den-sity of tailorable functional groups at theperiphery. The nanoscale branching archi-tecture provides them with several advan-tages over linear polymers, nanoparticles,and liposomes, such as rapid cellular entry,reduced macrophage uptake, and targetabil-ity. A range of polyamidoamine (PAMAM)dendrimers including partially hydrolyzedPAMAM dendrimers have been shown tobe effective vectors for gene transfection(Stiriba et al., 2002; Gebhart and Kabanov,2001; Cloninger, 2002).

One of the most exciting applications ofdendrimers in the medical field has beenthe potential use of dendrimers for targeteddrug delivery. These polymers have multiplefunctional end groups in their peripheriesthat can be easily modified to potentiallyattach a large number of drug molecules,ligands, or antibodies, making them idealfor use in drug targeting. Drugs can beeither encapsulated within the dendrimerby electrostatic or hydrophobic interactionsor complexed/conjugated to the end groups(Liu et al., 2000; Kohle et al., 2003).

Solubilizing group

Drug

Branching point

Targeting moiety

Figure 7.7. The large number of surface functionalgroups within ∼10 nm in diameter enables surfaceattachment of active molecules, ligands, imaging agents,and targeting moieties, leading to a multifunctionalnanodevice that is much smaller than typical liposomesand nanoparticles.

Chemotherapeutic agents like methotrexate,folic acid, doxorubicin, and 5-fluorouracilhave been conjugated to dendrimers andshown to successfully release the drugin vitro (Kono et al., 1999; Zhuo et al., 1999;Padilla De Jesus et al., 2002; Ihre et al.,2002). Dendritic unimolecular micelles havebeen synthesized with a hydrophobic coreand a hydrophilic shell and have been used asa model for dendrimer-based drug delivery.

To take advantage of dendrimers forsuperior cellular delivery of drugs, itis essential to study and understandthe transport of dendrimers into cells.Duncan and co-workers found that somegenerations of PAMAM dendrimers causedhaemolysis and were cytotoxic even at lowerconcentrations. However, anionic dendrimersdid not result in haemolysis and cytotoxicity(Malik et al., 2000). They also found thatin an everted rat intestinal sac system,125I-labeled anionic dendrimers exhibit rapidserosal transfer rates and low tissuedeposition. In contrast, cationic PAMAMdendrimers showed lower transport rates

Page 129: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

124 CELLULAR INTERACTIONS OF NANO DRUG DELIVERY SYSTEMS

TABLE 7.1. Properties of Dendrimers and Dendrimer–Drug Conjugates a .

DendrimersMolecular

Weight (Da)Generation

NumberNumber of

Surface Groups

DrugMolecules per

Dendrimer(Payload)c

PAMAM-G4-NH2 (G4) 14,215b 4 64 —PAMAM-G4-OH (G4-OH) 14,279b 4 64 —G4-OH-Ibuprofen 24,785c 4 64 51 (41%)G4-OH-GA-Methyl prednisoloned 19,154c 3 64 13 (26%)

a Where imaging was used, these materials were FITC-labeled with 3–8 molecules of FITC per dendrimerb Reported by Tomalia et al. (1990).cEstimated from 1H NMR.d GA, glutaric acid spacer. OH represents hydroxyl terminated dendrimer

(Wiwaattanapatapee et al., 2000). Baker andco-workers studied the transfection abilityof PAMAM dendrimers for DNA, acrossseveral cell lines and reported that PAMAMdendrimers can be used as an efficienttransfection vehicle for DNA/dendrimercomplexes (Kukowska-Latallo et al., 1996).This suggests that dendrimers can entera variety of cells. Recently, Ghandehariand co-workers addressed the permeabilityof dendrimers across Madin–Darby caninekidney (MDCK) cells and caco-2 cellmonolayers (Tajarobi et al., 2001; El-Sayedet al., 2002). They showed that permeabilityof the dendrimers across MDCK cell lineswas in the order G4 � G1 ≈ G0 > G3 >

G2, where G represents the generationnumber of the dendrimer. Their resultssuggested that the permeability of thedendrimers appeared to be related to thegeneration number, concentration, incubationtime, and probable modulation of the cellmembrane by cationic dendrimers.

In recent studies by Kannan and co-workers, the intracellular trafficking of den-drimers, dendrimer–drug conjugates, andtheir cellular activities were investigatedusing a combination of flow cytometry,fluorescence and confocal microscopy, andUV/Vis spectroscopy (Kannan et al., 2004;Kohle et al., 2004a,b; Khandare et al., 2004).A key aspect of these studies was theestablishment of time scales for entries

and the “quantification” of the amount ofdendrimer transported into cells. A vari-ety of dendrimers and drug conjugates wereinvestigated. Chemistry and cellular interac-tions of drug–dendrimer conjugates of anti-inflammatory drugs—namely, ibuprofen andmethyl prednisolone—are highlighted.

7.3.3.2. Chemistry. Ibuprofen is a rela-tively small drug (molecular weight 206Da), while methyl prednisolone (molecularweight 376 Da) is a relatively large steroidaldrug. The drugs were conjugated to a neutralPAMAM-G4 hydroxyl-terminated dendrimerusing N ,N ′-dicyclohexyl-carbodiimide (DC-C) as a coupling agent and was followed byextensive purification before isolating theconjugate. For the larger methyl predniso-lone, a highly reactive glutaric acid spacermolecule was used to overcome the sterichindrance of the aromatic ring in the drug.For the cell trafficking studies, these conju-gates were further labeled with a fluorescentprobe, fluorescein isothiocyanate (FITC).This resulted in dendrimer–drug–FITC con-jugates. Care was taken to remove any traceamount of free FITC in the FITC-labeledconjugates. More details can be found else-where (Kannan et al., 2004; Kohle et al.,2004a,b; Khandare et al., 2004).

7.3.3.3. Cellular Interactions of FreeDendrimer. Rapid rate of cellular entry of

Page 130: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

POLYMER-BASED SYSTEMS 125

the tagged G4-NH2 is evident from the highdegree of correlation in the absorbance lossin the supernatant (from UV analysis) andthe simultaneous absorbance increase in thecell lysate (Fig. 7.8A) This correlates wellwith the flow cytometry data (Fig. 7.8B),which shows a significant (exponential) pick-up in the FITC intensity, which is associatedwith the dendrimer uptake by the cells.These results were qualitatively consistentwith fluorescence microscopy images onthe FITC-labeled pure dendrimers (notshown). Approximately 90% of PAMAM

dendrimers (amine-terminated generation 3and 4) enter lung epithelial cells within1 h. Surprisingly, a neutral PAMAM-G4-OH showed a comparable cellular entryprofile to that of the cationic G4-NH2.Epithelial cells are known to possess anoverall anionic charge. Therefore the entryof cationic PAMAM dendrimers may bebecause of the electrostatic interaction withnegatively charged epithelial cells probablyvia the fluid-phase pinocytic route. Since G4-OH does not have cationic amine surfacegroups, the cellular entry may be because

100

0

20

40

60

80

0 1 2 3 4 5

Time (hours)

(A)

% C

hang

e in

abs

orba

nce

6 7

PAMAM G4 NH2 - cell Iystate

PAMAM G4 NH2 - supernatant

200

Cou

nts

0

100 101 102

FL1-H

103 104

40

80

5 min2 min

60 min

120 min

30 min160

120

(B)

Figure 7.8. (A) The normalized absorbance evolution versus time. For the supernatant [A(t) − A(final)]/[A(initial)−A(final)] is used. For the cell lysate, [A(t)/A(final)] is used. It can be seen that the increase in the normalizedabsorbance (A) of the FITC labeled PAMAM G4-NH2 dendrimer with respect to time (t) in the cell lysate correlatesvery well with the decrease in absorbance in the cell supernatant. (B) Flow cytometry of the cell entry dynamicsof pure PAMAM-G4-NH2 in A549 lung epithelial cell line. The log of FITC absorption intensity (FL1-H x axis) isplotted against the number of cells (counts). The exponential increase in the cellular uptake of polymers within fewminutes is evident. Based on this and the UV/Vis data (Part A), it is evident that dendrimers enter cells rapidly.

Page 131: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

126 CELLULAR INTERACTIONS OF NANO DRUG DELIVERY SYSTEMS

of the nonspecific adsorptive route, withsome specific interactions due to the interioramine groups that would be cationic. Anotherimportant aspect in the rapid cellular entry inthe A549 cells could be due to the higherpermeation of cancer cells. Preliminarymeasurements on normal smooth musclecells show that the entry is somewhat slowerin normal cells (Kannan et al., 2004).

7.3.3.4. Cellular Interactions of Drug-Conjugated Dendrimer. The transportof the drug carrying dendritic nanode-vices into cells is vital to the ultimatedelivery and the therapeutic effect. Den-drimer–drug conjugates with high drug pay-loads have been investigated. The neutraldendrimer PAMAM-G4-OH was conjugatedwith ibuprofen and methyl prednisolone.These studies are among the first to look

at the transport and therapeutic efficiency ofdendrimer-conjugated noncancerous drugs.

7.3.3.4.1. Dendrimer–Ibuprofen Nano-devices. Using flow cytometry, it was foundthat the dendrimer–drug conjugate enteredcells rapidly, with significant fluorescenceintensity increase within 15 min, correspond-ing to a conjugate uptake of >30%. Morethan 50% of the G4-OH–ibuprofen conju-gate entered the cells within 60 min. Thecellular entry was also visualized by usingphase contrast and fluorescence microscopy(Fig. 7.9A–D). It is evident that both thefree and the dendrimer-conjugated ibupro-fen entered the cells. As indicated by themicroscopy images, the conjugates appearto be present mostly in the cytosol. Fordrugs such as ibuprofen and methyl pred-nisolone, the mechanism of action requires

A. Control

C. FITC labeled ibuprofen, D. FITC labeled ibuprofen-dendrimer

B. FITC-labeled Dendrimer

Figure 7.9. Confocal fluorescence images of A549 lung epithelial cells after treatment for 1 hour with (A) control,(B) FITC-labeled G4OH dendrimer, (C) FITC-labeled ibuprofen, and (D) FITC-labeled ibuprofen–dendrimerconjugate after 4 h.

Page 132: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

POLYMER-BASED SYSTEMS 127

them to be delivered to the cytoplasm.Therefore, the delivery of the drug to thecytoplasm is sufficient. On the other hand,for DNA delivery, the DNA complexed withthe cationic dendrimer must be transportedto the nucleus for it to be effective.

The mode of action of ibuprofen involvesthe acetylation of cyclooxygenase-2 (COX-2) which blocks access and egress to/fromthe active site, inhibiting the production ofprostaglandin. The efficiency of the den-drimer–ibuprofen conjugates to suppressCOX-2 was assessed by measuring theprostaglandin (PGE2) in the cell super-natant. Free ibuprofen did not inhibit theprostaglandin release from A549 cells after30 min of incubation, while the dendrimer–ibuprofen conjugate showed a significantinhibition within the same time period (p <

0.05). However, there was no significantdifference (p > 0.05) in inhibition of theprostaglandin (PGE2) release between freeand conjugated ibuprofen at 60 and 360 min(Fig. 7.10). Neither blank dendrimer nor thesolvent showed any suppression of PGE2

synthesis. The above results imply that con-jugates rapidly enter the cell and producethe desired pharmacological action at the tar-get site in the cytosol. Although a moderate

fraction of free ibuprofen enters the cellswithin 30 min, it may not achieve a suf-ficient concentration to elicit a rapid phar-macological response. On the other hand,the dendrimer–ibuprofen conjugate achievesa high local concentration in the cell dueto its high drug payload. At this point, itis unclear whether the ibuprofen is releasedfrom the nanodevice inside the cell or ifthe drug is effective even in the conjugatedform. However, once the device enters insidethe cell, it is conceivable that the acidic pHand the enzymes in the endosomes wouldhydrolyze the ester bond in the conjugate,thereby releasing the free drug in the cytosolto suppress prostaglandin synthesis.

7.3.3.4.2. Dendrimer–Methyl Predniso-lone Nanodevices. Methyl prednisolone isa relatively hydrophobic steroid used totreat lung inflammation in asthma. Pre-liminary studies have indicated that den-drimers could increase the lung residencetime in mice models for lung inflamma-tion. To assess the cellular transport andactivity of the steroid containing dendrimerconjugates, microscopy and prostaglandinassay were used. The A549 lung epithelial

120

0

20

40

60

100

80

30 36060

Per

cent

inhi

bitio

n of

PG

E2

Time in minutes

Ibuprofen

G4-Ibuprofen conjugate

Figure 7.10. Percent inhibition of prostaglandin (PGE2) for free ibuprofen and PAMAM-G4-OH–ibuprofenconjugate as a function of treatment time at 30, 60, and 360 min (average of four measurements with error bars) inA549 lung epithelial cell line. Blank dendrimer and solvent did not show any inhibition of prostaglandin release.

Page 133: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

128 CELLULAR INTERACTIONS OF NANO DRUG DELIVERY SYSTEMS

(A)

(C)

(B)

(D)

Figure 7.11. (A–C) Fluorescence microscopic images (magnification 40×) of A549 cells 4 h of treatment with (A)control (no treatment), (B) FITC labeled methylprednisolone, and (C) FITC-labeled dendrimer–methylprednisoloneconjugate, (D) Confocal microscopy images of the FITC-labeled dendrimer–methylprednisolone conjugate inside thecell. The presence of the nanodevices in the cytoplasm is shown using both fluorescence and confocal microscopy.

cancer cells were treated with the G4-OH-methyl prednisolone conjugates. At theend of four hours, the cells were observedunder fluorescence microscopy to identifythe cellular localization of the free anddrug conjugated dendrimer (Fig. 7.11A–C).Fluorescence microscopy images of treatedA549 lung epithelial cells provide insightsinto the location and distribution of thedrug and conjugates inside the cells. Thecells without FITC show no fluorescence asexpected (Fig. 7.11A). Both free and drug-conjugated dendrimers were mainly foundto be in the cell cytoplasm. Furthermore,this was substantiated from confocal micro-scopic images in Figs. 7.11B and 7.11C,where the fluorescence is seen mostly inthe cytoplasm. This suggests that the freemethyl prednisolone and dendrimer–methylprednisolone conjugate are mainly localizedin the cytosol in the cell. Free methyl pred-nisolone entered the cells faster than the

methyl prednisolone–dendrimer conjugate(data not shown). The efficiency of theendocytosis mechanism is influenced bythe molecular size, and further studies arerequired to understand the cellular uptakemechanism. For methyl prednisolone, cyto-plasmic delivery is sufficient, since it exertspharmacological action by binding to theglucocorticoid receptors in the cytoplasmiccompartment.

Dendrimer–methyl prednisolone conju-gate inhibited prostaglandin synthesis to thesame extent (more than 95% inhibition) asfree methyl prednisolone (Fig. 7.12) after 4 hof treatment. The results indicate that theconjugate was able to enter the cell and pro-duce the desired pharmacological action. Itis interesting to note that even though only40% of the conjugate was inside the cellas opposed to >50% of the free MP, itproduced a comparable (p > 0.05) pharma-cological action to free methyl prednisolone.

Page 134: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

POLYMER-BASED SYSTEMS 129

50

40

30

20

Normal Control MP MPG

Con

c of

PG

E2

(pg/

ml)

10

0

−10

Figure 7.12. Box plot showing prostaglandin (PGE2) concentration in the cell supernatant for different treatmentperiods with A549 lung epithelial cell lines. Normal indicates the cells with no induction of prostaglandin synthesis.Control represents cells with prostaglandin induction but with no treatment. MP and MPG indicate the cells, whichwere induced to secrete prostaglandin and was treated with free methyl prednisolone and dendrimer conjugatedmethyl prednisolone respectively. The cells treated with MP/MP–dendrimer conjugate (4 h) suppressed prostaglandinsecretion, and the PGE2 levels were comparable to normal cells. Except normal and control treatment (n = 2).

From these results, it appears that high drugpayload in the dendrimer conjugate producesa high local drug concentration inside the cellto elicit a significant therapeutic response. Atthis point, it is unclear whether the MP isreleased from the dendrimer conjugate insidethe cell or if the drug is effective even in theconjugated form. However, once the deviceenters inside the cell, it is conceivable thatthe acidic pH and the enzymes in the endo-somes would eventually hydrolyze the esterbond in the conjugate to release the freedrug in the cytosol (McGraw et al., 1991).Based on previous results on dextran–methylprednisolone conjugates, the hydrolysis pro-cess is likely to take much longer than thetime frame used in current studies. There-fore, we may tentatively conclude that theconjugate may not have released the drugappreciably in 4 h. Nevertheless, the highpayload drug conjugate shows significanttherapeutic activity. It may be possible thatthe conjugate may slowly release the drugover a sustained period, thereby providingtherapeutic effect over longer times. Further

studies are required to investigate the stabil-ity of dendrimer–drug conjugate at variouspHs and in the presence of enzymes to under-stand the intracellular hydrolysis of the con-jugate.

7.3.3.5. Summary (Dendritic Nano-devices). Dendritic polymers are a rela-tively new class of tree-like polymers thatare typically much smaller and more sta-ble than liposomes and nanoparticles. Theyhave shown reduced macrophage uptakeand decreased inflammatory response com-pared to liposomes. As more drug deliv-ery approaches are developed using dendriticpolymers (including hyperbranched poly-mers), understanding their cellular uptake isbecoming critical. Fluorescence and confocalmicroscopy play a key rule in understand-ing the biological properties of these nan-odevices. Most of the delivery efforts havefocused on cancer drugs. Non-cancer drugsrepresent an interesting opportunity to studythe cellular dynamics of these nanodevices.Based on the current understanding, they

Page 135: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

130 CELLULAR INTERACTIONS OF NANO DRUG DELIVERY SYSTEMS

appear to enter cells very rapidly and trans-port the conjugated or complexed drugseffectively. A predominant quantity of thenanodevice appears to localize in the cytosol.The conjugated drugs are therapeuticallyeffective, even if the drug is not released. Asthe drugs get released through the action ofthe enzymes on the conjugates to release thedrug, these nanodevices may show sustaineddelivery also. These are subjects of currentinvestigation.

7.4. CLINICAL ASPECTS AND FUTUREPERSPECTIVES

From a clinical viewpoint, an ideal drugdelivery system is expected to (i) enhancethe therapeutic efficacy of the drug and (ii)reduce the adverse effects of the drug onother organs. Cancer therapy for instance isseverely limited by the toxicity caused bythe antitumor agents on normal cells. Dox-orubicin treatment is associated with severecardiotoxicity and hepatotoxicity, but whenit was administered using nanoparticles, nei-ther of theses side effects were observed dueto the localization of the nanoparticle formu-lation to the tumor (Kattan et al., 1992). Itis critical that the drug delivery vehicle doesnot upset the normal physiology of the bio-logical system, while exploiting the uniquepathophysiological characteristics of dis-eased tissue or cell. Therefore, it is not suffi-cient to just selectively target the therapeuticagent to the cell, but is also essential to targetto the specific compartment in the cell. Clin-ical relevance of specific drug targeting canbe exemplified comparing the case of vac-cine development for bacteria and viruses.Exogenous antigens (like bacteria) are usu-ally taken up into cells by phagocytosis orendocytosis, which after degradation in thelysosomes are presented in a major histocom-patibility complex (MHC)-class II restrictedmanner. Such antigen presentation inducesantigen-specific antibody production but notcytotoxic T-lymphocyte response (CTL). On

the contrary, endogenous antigens (cytoplas-mic antigens such as viruses) are degraded byproteasomes in cytoplasm and presented withMHC class-I molecules, eventually leadingto the induction of CTL response (Kunisawaet al., 2001). This antigenic response plays akey role against viral infectious diseases andcancer. Though conventional liposomes havemet with some success for bacterial vaccines,they have achieved limited success for viraland tumor vaccines due to the lack of induc-tion of CTL response (Sheikh et al., 2000).When diphtheria toxin A is delivered usingconventional liposomes, no CTL responsewas detected because it is taken up by theendosomes and transported to the lysosomes.In sharp contrast, with fusogenic liposomesthe CTL response was observed, because thetoxin was released directly into the cytosolby the fusion of “fusogenic liposome” withthe cell membrane (Nakanishi et al., 2000).Specific and selective targeting of nanovehi-cles can also be accomplished by attachingcell-specific ligands to the nanovehicles orby modifying the composition of the vehi-cle to respond to various chemical, physi-cal, or biological stimuli. Targeting results ina high local drug concentration at the siteof action with minimal systemic exposure,thus ultimately reducing the overall dose ofthe drug and consequently eliminating thedose related toxicity of the therapeutic agent.However, one of the challenges is to achievea high drug payload in the nanovehicle. Fromthis perspective, dendrimers offer tremen-dous potential as a high drug payload vehi-cle due to the presence of numerous func-tional groups on the surface (Kohle et al.,2004b). The nanosystems such as micellesand nanoparticles, in addition to transportingthe therapeutic agent inside the cell, can alsoact as a sustained or controlled drug deliverysystem (Panyam and Labhasetwar, 2003b; Liand Kwon, 2000). Chemistry of the nanove-hicle and the nature of the drug linkage tothe carrier play an important role in mod-ulating the release of the drug at the siteof action (Joseph et al., 2004). By achieving

Page 136: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

CLINICAL ASPECTS AND FUTURE PERSPECTIVES 131

sustained or controlled release of the drugfrom the vehicle, one can reduce the dosingfrequency, which significantly improves thepatient compliance to therapy.

Nanodelivery systems can positively alterthe tissue distribution of the therapeutic agentby its ability to partition into specific tis-sue compartments, leading to increase inthe plasma half-life and volume of distri-bution, while decreasing the plasma clear-ance of the drug. Pluronic block copoly-mers (PEO-b-PPO) or poloxamers have beenshown to inhibit P-glycoprotein efflux whichis partially responsible for multidrug resis-tance in cancer cells and reduced perme-ation across biomembranes (Batrakova et al.,1996, 1998). The polymer can form micelleswhich can carry drug in the nanocontainerof the micelle, thus bypassing the efflux sys-tem. Alternatively, the nanoparticles havebeen coated with poloxamer to improve theefficacy of the therapeutic agent (Soppimathet al., 2001).

Biocompatibility and safety of the vehicleis a critical component in clinical applicationof nano drug delivery systems. It hasbeen shown that the biodistribution andbiocompatibility of dendrimers depend on

its surface charge and generation number(Malik et al., 2000). In general, studies haveshown that balancing the surface hydropho-bicity and hydrophilicity improves the bio-compatibility of polymers. Neutral polymersand polyanions show less toxicity than poly-cations, while low-molecular-weight poly-mers have been found to undergo less pro-tein adsorption and platelet adhesion (Wanget al., 2004). Therefore, understanding thestructure–property relationships of vehiclesholds the key for the rational design of nan-odrug delivery systems.

A number of products based on nanodrugdelivery systems are in various stages of clin-ical development (Duncan, 2003), and someof them are already available for clinicaluse such as Ambisome (amphotericn B lipo-some) and Daunoxome (Daunorubicin cit-rate liposomal formulation). The formulationissues (scalingup, sterilization, and lyophiliza-tion) and biological issues (biocompatibility,in vitro– in vivo correlations) remain to beresolvedbeforeothernanoscopicdrugdeliverysystems can progress from the lab to the clinic.Nevertheless, the advances in nanotechnologyhas provided a platform for multidisciplinaryapproach to move closer to developing an ideal

Stimuli-responsivecomponent

Nanodeliveryvehicle

Cell-specific ligand

Therapeuticagent

Diagnostic agent

Figure 7.13. Schematic representation of an idealistic “smart NanoDrug delivery system.”

Page 137: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

132 CELLULAR INTERACTIONS OF NANO DRUG DELIVERY SYSTEMS

“smart nanodrug delivery system,” which canbe decorated with a targeting ligand and adiagnostic or imaging ligand apart from thetherapeutic agent of interest (Fig. 7.13). Theavailability of such “smart nanodrug deliverysystems” in the near future would allow us todetect,diagnose, target,modulatedelivery,andtrack the progression of therapy remotely andnoninvasively.

REFERENCES

Alenso, M. J., Losa, C., Calvo, P., and Vila-Jato, J. L. (1991). Approaches to improvethe association of amikacin sulfate to poly(cyanoacrylate) nanoparticles. Int. J. Pharm.68:69–76.

Allen, C., Maysinger, D., and Eisenberg, A.(1999). Nano-engineering block copolymeraggregates for drug delivery. Colloids Surf. B:Biointerfaces 16:3–27.

Alyautdin, R., Gothier, D., Petrov, V., Kharke-vich, D., and Kreuter, J. (1995). Analgesicactivity of the hexapeptide dalargin adsorbedon the surface of polysorbate 80 coatedpoly(butylcyanoacrylate) nanoparticles. Eur. J.Pharm. Biopharm. 41:44–48.

Amyere, M., Mettlen, M., Der Smissen, P. V.,Platek, A., Paurastre, B., Veithan, A., andCourtoy, P. J. (2002). Origin, originality,functions, subversion and molecular signalingof macropinocytosis. Int. J. Med. Microbiol.291:487–494.

Barenholz, Y., and Crommelin, D. J. A. (1994).Liposomes as pharmaceutical dosage forms. In:Encyclopedia of Pharmaceutical Technology (J.Swarbrick, ed.), pp. 1–39, Marcel Dekker, NewYork.

Batrakova, E. V., Dorodnich, T. Y., Klinski,E. Y., Kliushenkova, E. N., Shemchukova,O. B., Goncharova, O. N., Arjakova, V. Y.,Alakhov, V., and Kabanov, A. V. (1996).Anthracycline antibiotics noncovalently incor-porated into the block copolymer micelles:In vivo evaluation of anticancer activity. Br. J.Cancer 74:1545–1552.

Batrakova, E. V., Han, H. Y., Alakhov, V.,Miler, D. W., and Kabanov, A. V. (1998).Effect of pluronic block copolymers on drug

absorption in Caco2 cell monolayers. Pharm.Res. 15:850–855.

Brigger, I., Dubernet, C., and Couveur, P. (2002).Nanoparticles in cancer therapy and diagnosis.Adv. Drug Deliv. Rev. 54:631–651.

Brzezinska, D., Winska, P., and Balinksa, M.,(2000). Cellular aspects of folate and anti-folate membrane transport. Acta Biochim. Pol.47:735–749.

Cloninger, M. J. (2002). Curr. Opin. Chem. Biol.6(6):742.

Daeman, T., Regts, J., Meesters, M., Tenkate,M. T., Baker-Woudenberg, I. A. J. M., andScherphof, G. L. (1997). Toxicity of doxoru-bicin entrapped within long-circulating lipo-somes. J. Controlled Release 44:65–76.

Davda, J., and Labhasetwar, V. (2002). Charac-terization of nanoparticle uptake of endothelialcells. Int. J. Pharm. 233:51–59.

De Duve, C. (1963). Lysosomes. In: CibaFoundation Symposium (A. V. S. De Reuckand M. P. Cameron, eds.), pp. 411–412.Churchill, London.

de Lima, M. C. P., Simoes, S., Pires, P.,Faneca, H., and Duzgunes, N. (2001). Cationiclipid–DNA complexes in gene delivery: frombiophysics to biological applications. Adv.Drug Deliv. Rev. 47:277–294.

Drews, J. (2000). Drug discovery: A historicalperspective. Science 287:1960–1964.

Duncan, R. (2003). The dawning era of polymertherapeutics. Nature Rev. 2:347–360.

Dunn, S. E., Brindley, A., Davis, S. S., Davies,M. C., and Illum, S. (1994). Polystyrene-poly(ethyleneglycol) (PS-PEG-2000) particlesas model systems for site specific drugdelivery. 2. The effect of PEG surface densityon the in vitro cell interaction and in vivobiodistribution. Pharm. Res. 11:1016–1022.

Egea, M. A., Gamisani, F., Valero, J., Garcia,M. E., Garcia, M. L. (1994). Entrapment of cis-platin into biodegradable polyalkylcyanoacry-late nanoparticles. Farmaco 49:211–217.

El-Sayed, M., Ginski, M., Rhodes, C., andGhandehari, H. (2002). J. Controlled Release81(3):355.

Esfand, R., and Tomalia, D. A. (2001). Poly-(amidoamine) (PAMAM) dendrimers: frombiomimicry to drug delivery and biomedicalapplications. Drug Discov. Today 6:427–436.

Page 138: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 133

Fattal, E., Rojas, J., Youssef, M., Couvuer, P., andAndremont, A. (1991). Liposome entrappedampicillin in the treatment of experimentalmurin listeriosis and salmanellosis. Antimicrob.Agents Chemother. 35:770–772.

Felgner, P. L., and Ringold, G. M. (1989).Cationic liposomes mediate transfection. Nature337:387–388.

Felgner, P. L., Gadek, T. R., Holm, M., Roman,R., Chan, H. W., Wenz, M., Northrop, J. P.,Ringold, G. M., and Danielsen, M. (1987).Lipofectin: A highly efficient lipid-mediatedDNA-transfection procedure. Proc. Natl. Acad.Sci. USA. 84:7413–7417.

Foster, K. A., Yazdanian, M., and Audus, K. L.(2001). Microparticulate uptake: Mechanismsof in vitro cell culture models of the respiratoryepithelium. J. Pharm. Pharmacol. 53:57–66.

Gebhart, C. L., and Kabanov, A. V. (2001). J.Controlled Release 73:401.

Goldberg, R. I., Smith, R. M., and Jarett,L. (1987). Insulin and α2-macroglobulin-methylamine undergo endocytosis by differentmechanisms in rat adipocytes. I. Comparisonof cell surface events. J. Cell. Physiol.133:203–212.

Hafez, I. M., Cullis, P. R. (2001). Roles of lipidpolymorphism in intracellular delivery. Adv.Drug Deliv. Rev. 47:139–148.

Hashida, M., Nishikawa, M., Yamashita, F., andTakakura, Y. (2001). Cell specific delivery ofgenes with glycosylated carriers. Adv. DrugDeliv. Rev. 52:187–196.

Hawker, C. J., and Frechet, J. M. J. (1990).Preparation of Polymers with ControlledMolecular Architecture. A New ConvergentApproach to Dendritic Macromolecules. J. Am.Chem. Soc. 112:7638.

Ihre, H. R., Padilla De Jesus, O. L., Szoka, F. C.,Jr., and Frechet, J. M. J. Polyester (2002). Den-dritic Systems for Drug Delivery Applications:Design, Synthesis, and Characterization. Bio-conjugate Chemistry. 13(3):443.

Jain, R. K. (1987). Transport of molecules acrosstumor vasculature. Cancer Metastasis Rev.6:559–593.

Jeong, B., Bae, Y. H., Lee, D. S., and Kim,S. W. (1997). Biodegradable block copolymeras injectable drug delivery systems. Nature388:860–862.

Johannes, L., and Lamaze, C. (2002). Clathrin-dependent or not: Is it still the question? Traffic3:443–451.

Joseph, A., Souza, M. D., and Topp, E. M.(2004). Release from polymeric prodrugs.Linkages and their degradation. J. Pharm. Sci.93:1962–1979.

Kakizawa, Y., and Kataoka, K. (2002). Blockcopolymer micelles for delivery of gene andrelated compounds. Adv. Drug Deliv. Rev.54:203–222.

Kaneda, Y., Saeki, Y., and Morishita, R. (1999).Gene therapy using HVJ-liposomes: The bestof both worlds? Mol. Med. Today 5:298–303.

Kannan, S., Kolhe, P., Kannan, R. M., Lieh-Lai,M., and Glibatec, M. (2004). J. Biomater. Sci.Polymers 15(3):311.

Kataoka, K., Harada, A., and Nagasaki, Y. (2001).Block copolymer micelles for drug delivery:Design, characterization and biological signifi-cance. Adv. Drug Deliv. Rev. 47:113–131.

Kattan, J., Droz, J. P., Couveur, P., Marino,J. P., Boutan-Laroze, A., Rougier, P., Brault,P., Vranckx, H., Grognet, J. M., Morge, X.,and Sancho-Garrier, H. (1992). Phase I clinicaltrial and pharmacokinetics evaluation of dox-orubicin carried by poly isohexylcyanoacrylatenanoparticles. Invest. New Drugs 10:191–199.

Kim, C. K., and Han, J. H. (1995). Lymphaticdelivery and pharmacokinetics of methotrexateafter intramuscular injection of differentlycharged liposomes-entrapped methotrexate torats. J. Microencapsulation 12:437–446.

Kolhe, P., Khandare, J., Pillai, O., Kannan, S.,Lieh-Lai, M., and Kannan, R. M. (2004).Hyperbranched polymer-drug conjugates withhigh drug payload for enhanced cellulardelivery. Pharm. Research. 21(12):2185.

Kolhe, P., Khandare, J., Pillai, O., Kannan, S.,Lieh-Lai, M., and Kannan, R. M. (2005). Syn-thesis, cellular transport, and activity of poly-amidoamine dendrimer–methylprednisoloneconjugates. Bioconjugate Chem. 16(4):1049.

Kolhe, P., Khandare, J., Pillai, O., Kannan, S.,Lieh-Lai, M., and Kannan, R. M. (2006). Pre-paration, cellular transport, and activity of poly-amindoamine-based dendritic nanodevices witha high drug payload. Biomaterials. 27(4):660.

Kolhe, P., Misra, E., Kannan, R. M., Kannan, S.,and Lieh-Lai, M. (2003). Int. J. Pharmaceutics259(1–2):143.

Page 139: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

134 CELLULAR INTERACTIONS OF NANO DRUG DELIVERY SYSTEMS

Kono, K. (2001). Thermosensitive polymermodified liposomes. Adv. Drug Deliv. Rev.53:307–319.

Kono, K., Liu, M., and Frechet, J. M. J. (1999).Bioconjugate Chem. 10(6):1115.

Kukowska-Latallo, J. F., Bielinska, A. U., John-son, J., Spindler, R., Tomalia, D. A., and Baker,J. R., Jr. (1996). Proc. Natl. Acad. Sci. USA, 93,4897.

Kunisawa, J., Nakagawa, S., and Mayumi,T. (2001). Pharmacotherapy by intracellulardelivery of drugs using fusogenic liposomes:Application to vaccine development. Adv. DrugDeliv. Rev. 52:177–186.

Kwon, G. S. (1998). Diblock copolymer nanopar-ticles for drug delivery. Crit. Rev. Ther. DrugCarrier Syst. 15:481–512.

Labhasetwar, V., Bonadio, J., Goldstein, S. A.,and Levy, R. J. (1999). Gene transfection usingbiodegradable nanospheres results in tissueculture and a rat osteotomy model. ColloidsSurf. B: Biointerfaces 16:281–290.

Lavasanifar, A., Samuel, J., and Kwon, G. S.(2002). Poly(ethylene oxide)-block–Poly(L-amino acid) micelles for drug delivery. Adv.Drug Deliv. Rev. 54:169–190.

Lin, S. Y., Chen, K. S., Teng, H. H., andLi, M. J. (2000). in vitro degradation anddissolution behavior of microspheres preparedby low molecular weight polyesters. J.Microencapsulation 17:577–586.

Liu, D. (1997). Biological factors involved inblood clearance of liposomes by liver. Adv.Drug Deliv. Rev. 24:201–213.

Liu, M. J., and Frechet, J. M. J. (1999). Pharma-ceutical Sci. Technol. Today 2(10):393.

Liu, M., Kono, K., and Frechet, J. M. J., (2000).J. Controlled Release 65(1–2):121.

Li, Y., and Kwon, G. S. (2000). Methotrexateesters of poly(ethylene oxide) block-poly(2-hydroxyethyle-L-aspartamide), Part I: Effectsof the level of methotrexate conjugation onthe stability of micelles and on drug release.Pharm. Res. 17:607–611.

Malik, N., Wiwattanakpatapee, R., Klopsch,R., Lorenz, K., Frey, H., Weener, J. W.,Meijer, E. W., Paulus, W., and Duncan, R.(2000). Dendrimers: Relationships betweenstructure and biocompatibility in vitro andpreliminary studies on the biodistribution

of 125I labeled polyamidoamine dendrimersin vivo. J. Controlled Release 65:133–148.

McGraw, T. E., Maxifield, F. R., Juliano, R. L.(1991). Targeted Drug Delivery, pp. 11–41.Springer Verlag, New York.

Michael, S. I., and Curiel, D. T. (1994). Strategiesto achieve targeted gene delivery via thereceptor mediated endocytosis pathway. GeneTherapy 1:223–232.

Morishita, R., Gibbons, G. H., Kaneda, Y., Ogi-hara, T., and Dzau, V. J. (1994). Pharmacoki-netics of antisense oligonucleotides (Cyclin B1and C and C2 kinase) in the vessel wall:Enhanced therapeutic utility for restonsis byHVJ-liposome method. Gene 149:3–9.

Mukherjee, S., Ghosh, R. N., and Maxfield,F. R. (1997). Endocytosis. Physiol. Rev.77:760–803.

Nagayasu, A., Uchiyama, K., and Kiwada, H.(1999). The size of liposomes: a factor whichaffects their targeting efficiency to tumorsand therapeutic activity of liposomal antitumordrugs. Adv. Drug Deliv. Rev. 40:75–87.

Nagayasu, A., Uchiyama, K., Nishida, T.,Yamagiwa, Y., Kawai, Y., and Kiwada, H.(1996). Is control of distribution of liposomesbetween tumors and bone marrow possible?Biochim. Biophys. Acta 1278:29–34.

Nakanishi, T., Hayashi, A., Kunisawa, J.,Tsutsum, Y., Tanka, K., Yashiro-Ohtani, Y.,Nakanishi, M., Fujiwara, H., Hamaoka, T.,and Mayumi, T. (2000). Fusogenic liposomesefficiently deliver exogenous antigen throughthe cytoplasm into the MHC-class I processingpathway. Eur. J. Immunol. 30:1740–1747.

Nilsson, J. J., Thyberg, J., Heldin, C. H.,Westermark, B., and Wasteson, A. (1983).Surface binding and internalization of plateletderived growth factor in human fibroblasts.Proc. Natl. Acad. Sci. USA 80:5592–5596.

Oussoren, C., and Storm, G. (1997). Lymphaticuptake and biodistribution of liposomesafter subcutaneous injection. III. Influenceof surface modification with poly(ethyleneglycol). Pharm. Res. 14:1479–1484.

Oussoren, C., Zuidema, J., Crommelin, D. J. A.,and Storm, G. (1997). Lymphatic uptake andbiodistribution of liposomes after subcutaneousinjection. II. Influence of liposomal size, lipidcomposition and lipid dose. Biochim. Biophys.Acta 1328:261–272.

Page 140: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 135

Padilla De Jesus, O. L., Ihre, H. R., Gagne, L.,Frechet, J. M. J., and Szoka, F. C., Jr. (2002).Bioconjugate Chem. 13(3):453.

Panyam, J., and Labhasetwar, V. (2003b).Biodegradable nanoparticles for drug and genedelivery to cells and tissues. Adv. Drug Deliv.Rev. 55:329–347.

Panyam, J., and Labhasetwar, V. Dynamicsof endocytosis and exocytosis of poly (D,L-lactide-co-glycolide) nanoparticles in vascularsmooth muscle cells. Pharm. Res. 20:110–118.(2003a).

Panyam, J., Zhou, W. Z., Prabha, S., Sahoo,S. K., and Labhasetwar, V. Rapid endo-lysosomal escape of poly (D,L-lactide co-glycolide) nanoparticle. Implication for drugand gene delivery. FASEB J. 16:1217–1226.(2002).

Peters, K. R., Carely, W. W., and Palade, G. E.(1985). Endothelial plasmalemmal vesicleshave a characteristic stripped bipolar surfacestructure. J. Cell. Biol. 101:2233–2338.

Radwan, M. A. (1995). In vitro evaluationof evaluation of poly isobutylcyanoacrylatenanoparticles as a controlled drug carrierfor theophylline. Drug Dev. Ind. Pharm.21:2371–2375.

Randolph, T. W., Randolph, A. D., Meber, M.,and Yeung, S. (1993). Sub-micron sizedbiodegradable particles of poly (L-lactic acid)via the gas antisolvent spray precipitationprocess. Biotechnol. Prog. 9:429–435.

Schnitzer, J. E. (2001). Caveolae: from basictrafficking mechanisms to targeting transcytosisfor tissue specific drug and gene deliveryin vivo. Adv. Drug Deliv. Rev. 49:265–280.

Senior, J. H. (1987). Fate and behavior ofliposomes in vivo: A review of controllingfactors. Crit. Rev. Ther. Drug Carrier Syst.3:123–193.

Sheikh, N. A., al- Shamisi, M., and Morrow,W. J. (2000). Delivery systems for molecularvaccination. Curr. Opin. Mol. Ther. 2:37–54.

Soppimath, K. S., Aminabhavi, T. M., Kulka-rni, A. R., and Rudzinski, W. E. (2001).Biodegradable polymeric nanoparticles asdrug delivery devices. J. Controlled Release70:1–20.

Stiriba, S.-E., Frey, H., and Haag, R. (2002).Angew. Chem. Int. Ed. Engl. 41:1329.

Suh, H., Jeong, B., Ralhi, R., and Kim, S. W.(1998). Regulation of smooth muscle cell pro-liferation using paclitaxel loaded poly(ethyleneoxide)–poly(lactide-glycolide) nanospheres. J.Biomed. Mater. Res. 42:331–338.

Tailefer, J., Jones, M. C., Brasseur, N., Van Liear,J. E., and Leroux, J. C. (2000). Preparation andcharacterization of pH responsive polymericmicelles for the delivery of photosensitizinganticancer drugs. J. Pharm. Sci. 89:52–62.

Tajarobi, F., El-Sayed, M., Rege, B. D., Polli,J. E., and Ghandehari, H. (2001). Int. J. Pharm.215(1–2):263.

Tomalia, D. A., Baker, H., Dewald, J., Hall,M., Kallos, G., Martin, S., Roeck, J.,Ryder, J., and Smith, P. (1985). A NewClass of Polymers: Starburst–DendrimersMacromolecules. Polym. J. 17(1):117.

Tomalia, D. A., Baker, H., Dewald, J., Hall,M., Kallos, G., Martin, S., Roeck, J., Ryder,J., and Smith, P. (1985). Dendritic macro-molecules: Synthesis of starburst dendrimers.Macromolecules. 19:2466.

Tomalia, A., Naylor, A. M., Goddard, W. A.,III., (1990). Starburst dendrimers: control ofsize, shape, surface chemistry, topology andflexibility in the conversion of atoms tomacroscopic materials. Angewandte. Chemie.102(2):119.

Tomoda, H., Kishimoto, Y., and Lee, Y. C.(1989). Temperature effect of endocytosis andexocytosis by rabbit alveolar macrophages.J. Biol. Chem. 264:15445–15450.

Tuzar, Z., and Kratochvil, P. (1976). Block andgraft co-polymer micelles in solution. Adv.Colloid Interface Sci. 6:201–232.

Ueda, M., Iwara, A., and Kreuter, J. (1998).Influence of the preparation methods onthe drug release behavior of loperamideloaded nanoparticles. J. Microencapsulation15:361–372.

Wang, S., and Low, P. S. (1998). Folate mediatedtargeting of anti-neoplastic drugs, imagingagents and nucleic acids to cancer cells. J.Controlled Release 53:39–48.

Wang, Y. Y., Robertson, J. L., Spillman, W. B.,Jr., and Claus, R. O. (2004). Effect of thechemical structure and the surface properties ofpolymeric biomaterials on the biocompatibility.Pharm. Res. 21:1362–1373.

Page 141: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

136 CELLULAR INTERACTIONS OF NANO DRUG DELIVERY SYSTEMS

Wiwaattanapatapee, R., Carreno-Gomez, B.,Malik, N., and Duncan, R. (2000). Pharm. Res.17:991.

Woodle, M. C. (1998). Controlling liposomeblood clearance by surface grafted polymers.Adv. Drug Deliv. Rev. 32:139–152.

Yoo, H. S., Oh, J. E., Lee, K. H., and Park, T. G.(1999). Biodegradable nanoparticles containing

doxorubicin–PLGA conjugate for sustainedrelease. Pharm. Res. 6:1114–1118.

Zhu, N., Liggitt, D., Liu, Y., and Debs,R. (1993). Systemic gene expression afterintravenous DNA delivery into adult mice.Science 261:209–211.

Zhuo, R. X., Du, B., and Lu, Z. R. (1999). J.Controlled Release 57:249.

Page 142: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

CHAPTER 8

ADAPTING AFM TECHNIQUES FOR STUDIESON LIVING CELLSJ. K. HEINRICH HORBERDepartment of Physics, University of Bristol, Bristol, United Kingdom

8.1. COMBINING AN AFM WITHOPTICAL MICROSCOPY ANDELECTROPHYSIOLOGICALTECHNIQUES

The AFM (Binnig et al., 1986) was thoughtfrom the very beginning to be an ideal toolfor biological research. Imaging living cellsunder physiological conditions and study-ing dynamic processes at the plasma mem-brane were envisioned (Drake et al., 1989),although it was clear that such experimentsare quite difficult, as the AFM cantilever isby far more rigid than cellular structures. Inthis context the way cells are supported isquite important. Also a parallel optical obser-vation is necessary to control the cantilevertip approach to distinct cellular features andto link the measurements to well-establishedoptical microscopy techniques.

8.1.1. AFM on an Inverted OpticalMicroscope

In order to address these problems, we startedin 1988 the development of a special AFMbuilt into an inverted optical microscope,which finally provided the first reproducibleimages of the outer membrane of a liv-ing cell held by a pipette in its normal

growth medium (Haberle et al., 1991, 1992;Horber et al., 1992; Ohnesorge et al., 1997).A conventional piezo-tube scanner movedthis pipette. The detection system for thecantilever movement used was in principlethe normal optical detection scheme, but aglass fiber was used as a light source veryclose to the cantilever. This setup allowed avery fast scanning speed of up to a pictureper second for imaging cells in the variabledeflection mode. This is possible because theparts moving in the liquid are very smalland, quite different from the imaging of cellsadsorbed to a microscope slide, no significantexcitation of disturbing waves or convectionin the liquid occurs. It was possible to keepthe cell alive and well for days within theliquid cell built. This made studies of liveactivities and kinematics possible, in additionto the application of other cell physiologicalmeasuring techniques. With this step in thedevelopment of scanning probe instruments,the capability of optical microscopy to inves-tigate the dynamics of biological processesof cell membranes under physiological con-ditions could be extended with the help ofthe AFM into the nanometer range.

At high imaging rates of up to oneframe per second, we finally could observestructures as small as 10 nm. This made it

Force Microscopy: Applications in Biology and Medicine, edited by Bhanu P. Jena and J.K. Heinrich Horber.Copyright 2006 John Wiley & Sons, Inc.

137

Page 143: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

138 ADAPTING AFM TECHNIQUES FOR STUDIES ON LIVING CELLS

possible to study processes such as the bind-ing of labeled antibodies, endo- and exocy-tosis, pore formation, and the dynamics ofsurface and membrane cytoskeletal structuresin general. The problem that still has to besolved, unfortunately for each cell individ-ually, is to control the interaction betweentip and plasma membrane structures, whichare influenced quite strongly by the so-calledextracellular matrix of cells containing abroad variety of sugars and other polymerstructures.

With the integrated tip of the cantileverforces in the range of some 10 pN up to some10 nN are applied to the investigated cell, themechanical properties of cell surface struc-tures therefore dominate the imaging process.On one hand, this fact mixes topographicand elastic properties of the sample in theimages; on the other hand, it provides addi-tional information about cell membranes andtheir dynamics in various situations duringthe life of the cell, if these two aspects canbe separated in the AFM images. To separatethe elastic and topographic properties, furtherinformation is needed, which can be providedeither by additional topographic data fromelectron microscopy or by using AFM modu-lation techniques. The pipette–AFM conceptis very well suited for such modulation mea-surements because, as mentioned, convectionor excitation of waves in the solution due tothe pipette movement is negligible and sothe modulation can be done very fast. Nev-ertheless, for a thorough analysis of a cellwall elasticity map, one would have to recordpixel-by-pixel a complete frequency spec-trum of the cantilever response and deriveimage data in various frequency regimes.This would need too much time for a highlydynamic system like a living cell even withthe speed possible with a cell at the endof a pipette. Experiments we have done onliving cells showed in some cases a certainweak mechanical resonance in the regime ofseveral kilohertz. Such resonance might beused to characterize cells and provide a kind

of spectroscopic fingerprint for cellular pro-cesses or even drug effects, which may leadto new methods of medical diagnosis at thecellular level.

Figure 8.1 shows the schematic arrange-ment of the AFM above the objective ofan inverted optical microscope. The samplearea can be observed from below through aplanar surface defined by a glass plate witha magnification of 600×–1200× and fromabove by a stereo microscope with a mag-nification of 40×–200×. The illumination isfrom the top through the less well-definedsurface of the aqueous solution. In order notto block the illumination, the manipulatorfor the optical fiber and for the micropipettepoint toward the focal plane at an angle of45◦. The lever is mounted in a fixed posi-tion within the liquid slightly above the glassplate tilted by 45◦. The single-mode opticalfiber is used to reduce the necessary opti-cal components by bringing the end as closeas possible to the lever. To do so, the lastseveral millimeters of the fiber’s protectivejacket are removed. The minimum distanceis determined by the diameter of the fibercladding and the geometry of the lever. Wenormally use fibers for 633-nm light whichhave a nominal cladding diameter of 125 µmand levers which are 200 µm long. Holdingthe fiber at an angle of 45◦ with respect to thelever means that we can safely bring the fibercore as close as 150 µm to the lever. The4-µm-diameter core has a numerical apertureof 0.1, and the light emerging from the fibertherefore expands with an apex angle of 6◦.For the geometry given above, the smallestspot size achievable is 50 µm, approximatelythe size of the triangular region at the end ofthe cantilever. Due to the construction of themechanical pieces holding the pipette and thefiber positioner, the closest we can bring theposition-sensitive quadrant photodetector tothe lever is approximately 2 cm. This impliesa minimum spot size of 2.1 mm, withinthe 3-mm × 3-mm borders of the detec-tor. Using a 2-mW HeNe laser under nor-mal operational conditions, the displacement

Page 144: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

COMBINING AN AFM WITH OPTICAL MICROSCOPY AND ELECTROPHYSIOLOGICAL TECHNIQUES 139

Figure 8.1. Schematic drawing of the AFM built onto an inverted optical microscope with a patch-clamp pipette asa sample holder. For the optical detection of the cantilever deflection, an optical fiber as a light source very closeto the cantilever is used. The detection of the reflected light is done by a quadrant photodiode above the samplechamber.

sensitivity is below 0.01 nm with a signal-to-noise ratio of 10, comparable to the sensitiv-ity of other optical detection techniques. Theadvantage of this method is that there are nolenses and there is no air–liquid or air–solidinterface across which the incoming lightbeam must travel; only the outgoing light hasto cross the water–air interface, because thedetector cannot be immersed in the water.

The pipette used for holding the cellis made out of a 0.8-mm borosilicat glasscapillary that is pulled to about (a) 2- to4-µm opening in three pulling steps. Itis mounted on the piezotube scanner andcoupled to a fine and flexible Teflon tube,through which the pressure in the pipettecan be adjusted by a piston or water pump.The pipette is fixed at an angle that allowsimaging of the cell without the danger oftouching the pipette with the lever. All thesecomponents are located in a container of50-ml volume. The glass plate above the

objective of the optical microscope forms thebottom of this container.

After adding several microliters of the cellsuspension, a single cell can be sucked ontothe pipette and fixed there by maintaininglow pressure in the pipette. The other cellsare removed through the pumping system.The fixed cell is placed close to the AFMlever by a rough approach with screws andfinally positioned by the piezo scanner. Whenthe cell is in close contact with the tipof the lever, scanning of the capillary withthe cell attached leads to position-dependentdeflections of the lever. The levers usedare microfabricated silicon and silicon nitritetriangles with 200-µm length and with aspring constant of 0.12 N/m. The forceswhich can be applied with these levers, andthe sensitivity of the detection system can beas low as 0.1 nN.

With such forces applied to the plasmamembrane of a cell, the stiffness of themembrane structures dominates the images.

Page 145: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

140 ADAPTING AFM TECHNIQUES FOR STUDIES ON LIVING CELLS

The scaffold of the cortical layer of actinfilaments and actin-binding proteins thatare cross-linked into a three-dimensionalnetwork and closely connected to the sur-face membrane dominate the images. Thesefilaments have a structural role and are alsoinvolved in creating the changes seen inthe AFM as the dynamics of the surfaceprotrusions.

The relatively stable arrangements of theactin filaments are responsible for theirrelatively persistent structure. But thesesurface actin filaments are not permanent.During phagocytosis or cell movement, rapidchanges of shape occur at the cell surface.These changes depend on the transient andregulated polymerization of cytoplasmic freeactin or the depolarization during the break-down of the actin filament complex. The timescale of cell surface changes on a larger scaleobserved with the AFM was about 1–2 hat room temperature. Except for small struc-tures some 10 nm in size, everything is quitestable for 1–2 h. More and larger structuresare rearranged only 10–15 min after thisperiod has elapsed.

In a series of experiments, we have stud-ied the membrane dynamics after infectionof the cell by pox viruses. On the timescaleof seconds to minutes after adding the virussolution to the chamber, the cell surface sud-denly becomes smooth and so soft that the tiptends to penetrate the cell surface even withloading force far below 1 nN. This state ofthe cell might be related to the penetration ofthe viral particles through the cell membrane.After a few minutes the cell again becomesmore rigid, and stable imaging is observedvery similar to the situation before the infec-tion. During the first hour after infection weusually did not detect any significant varia-tion in the membrane structures imaged. It isknown that within 4–6 h the first viruses arereproduced inside the cell and pass throughthe cell membrane via exocytosis (Stokes,1976). However, approximately 2.5 h afterinfection, we already observed a series ofprocesses occurring at the membrane. Single

clear protrusions become visible and growin size. The objects quickly disappear andthe original structure of the membrane isrestored. Such processes can occur severaltimes in the same area and last about 90 s forsmall protrusions of about 20 nm and last upto 10 min for larger ones of up to 100-nmsize. Each process proceeds distinctly andapparently independently of the others. Theyare never observed with uninfected cells andnever until 2 h after infection.

The fact that the growing protrusionsabruptly disappear after a while makesus believe that we observe the exocytosisof particles related to the starting virusreproduction, but not viruses. First progenyviruses are known to appear 5–8 h afterinfection, and they are clearly bigger thanthe structures described so far. It is known(Moss, 1986), however, that after 2–3 h,only the early stage of virus reproduction isfinished and the final virus assembly has justbegun. Since the protrusions are observedafter this characteristic time span, we believethat they are related to the exocytosis ofprotein agglomerates originating from thevirus assembly.

Significantly longer than 6 h after infec-tion there are even more dramatic changesobserved in the cell membrane. Large pro-trusions, with cross sections of 200–300 nm,grow out of the membrane in the vicin-ity of deep folds. These events occur muchless frequently than those after 2 h. Theseprotrusions also abruptly disappear, leavingbehind small scars at the cell surface. Con-sidering the timing and their size, we believethese protrusions are progeny viruses exit-ing the cell. Assuming that approximately20–100 viruses exit the living cell and thatroughly 1/40 of the cell surface is accessi-ble to the AFM tip, one should be able toobserve one or two of those events. We haveactually observed 2 processes exhibiting theright size and timing during one 46-h experi-ment on a single infected cell, one after 19 hand one after 35 h. It is known from elec-tron microscopy that individual viruses exit

Page 146: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

COMBINING AN AFM WITH OPTICAL MICROSCOPY AND ELECTROPHYSIOLOGICAL TECHNIQUES 141

0 min

1000 min

12 min

Figure 8.2. Exocytotic process seen about 19 h after the cell was infected by pox viruses. An unusually strongfingerlike structure is visible, and at one end a protrusion appears and disappears again after about 3 min. The sizeof the protrusion, the timing, and the evolution of the event are consistent with the interpretation that the release ofa virus at the end of a microvillus structure is observed. There is a strong similarity to electron micrographs andfluorescence-labeled optical observation of such events.

sometimes the cell at the end of microvilli.Figure 8.2 actually shows such a protrusionand an exocytotic event occurring at the end.The release of another particle shown inFig. 8.3 also occurs in a region where the cellmembrane shows an array of microvilli. Thisstriking similarity to results seen in electronand optical microscopy (Moss, 1986, (p. 40);Frischknecht et al., 1999) makes us believethat we indeed have imaged the exocytosisof a progeny virus through the membrane ofa living infected cell.

8.1.2. Integrating the AFMinto a Electrophysiological Setup

The method of fixing a cell to a pipette isflexible enough to allow integration of andcombination with well-established cell phys-iological techniques of manipulation used ininvestigations of single cells. The structuresobserved can be partially related to knownfeatures of membranes, but more importantthan just imaging structures is the possibil-ity provided by this technique to conductdynamic studies of living organisms on this

nanometer scale. In general, this techniquemakes studies of the evolution of cell mem-brane structures possible and provides infor-mation that brings us closer to understand notonly the “being” of these structures, but alsotheir “becoming.”

An AFM with the cell fixed on end of apipette, as described in the previous section,makes a combination of patch-clamp mea-surements on ion channels in the membraneof whole cells with force microscope stud-ies possible. A logical step in the devel-opment of AFM technique combined withestablished cell biological techniques there-fore was the development of a combinedpatch-clamp and AFM setup that can beused to investigate also excised membranepatches (Horber et al., 1995) and in this wayto study single ion channels in the mem-brane, which become activated by mechani-cal stress in the membrane. The importanceof these ion channels is very obvious inour senses of touching and hearing. Duringdevelopments that started in 1991, a new typeof patch-clamp setup was built being muchmore stable to satisfy the needs of AFM

Page 147: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

142 ADAPTING AFM TECHNIQUES FOR STUDIES ON LIVING CELLS

0 min

12 min

< 1000 nm >

< 1000

nm >

Figure 8.3. Exocytotic process seen about 35 h after viral infection of cultured monkey kidney cell. The protrusionin the lower part of the images over about 10 min slowly grow in size to about 200 nm in diameter. It disappearsabruptly, leaving a scar on the cell membrane.

applications (Fig. 8.4) and able to accommo-date the additional electrophysiological andoptical components. The chamber, where aconstant flow of buffer solution guaranteesthe right conditions for the experiments, con-sists of two optical transparent plastic plates;one at the top and one at the bottom, keepingthe water inside just by its surface tension.In this way, a flow cell is created, which isaccessible from two sides. The chamber ismounted on an xyz-stage together with theoptical detection of the AFM lever move-ment and a double-barrel application pipette.The latter was integrated so that the setup canalso be used for standard patch-clamp mea-surements with application of chemicals andmarkers to characterize the sample further.The patch-clamp pipette itself is mounted ona piezo-tube scanner fixed with respect to theobjective of an inverse optical microscopethat is necessary to control the approach ofthe pipette to the cell and to the AFM leverfinally. In the experiments, small membranepieces are excised from a cell inside thechamber containing none, one, or only a few

of the mechano-sensitive ion channels. Thisenables studies of currents through a sin-gle channel, which opens and closes on thetimescale of micro- to milliseconds resultingin currents in the pico-ampere range.

In such a setup with a membrane patch atthe end of the pipette the AFM can imagethe tip of the pipette with the patch ontop (Fig. 8.5). Furthermore, changes in theform of the patched membrane producedby changing pressure in the patch pipettecan be monitored, along with the reactionof the membrane to the change of theelectric potential across. In some images,cytoskeleton structures can be seen thatare excised together membrane. On thesestabilizing structures of the membrane andon the rim of the pipette, resolutions downto 10–20 nm can be achieved showingreproducible structures and changes that canbe induced by the application of force.

Voltage-sensitive ion channels changeshape in electrical fields, leading eventuallyto opening of the ion-permeable pore. Todetermine the size of this electromechanical

Page 148: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

COMBINING AN AFM WITH OPTICAL MICROSCOPY AND ELECTROPHYSIOLOGICAL TECHNIQUES 143

Test solutionControl solution

Piezo-tube

Detector

Bath electrode

Bath chamber

ApplicationpipetteBath-

perfusion

Oocyte Pipette electrode

Laser

Lever

SuctionPatch pipette

Piezo-tube

Pressure application

Figure 8.4. Schematic top view of the patch-clamp/AFM setup within its bath chamber. The patch pipette is shownin front of the cantilever. Bath and pipette electrodes are used for electrophysiological recordings. A glass plateis positioned behind the cantilever to reduce fluctuations of laser beam direction by movement of the water–airinterface. Different solutions can be applied to the excised patch by a two-barreled application pipette. If necessary,continuous perfusion of the path chamber can be stopped during AFM measurements.

Figure 8.5. Three-dimensional representation of membrane patches at the top of a pipette pulled to 2 µm in diameter.In the upper image the higher part in the background corresponds to the glass rim and the central hill correspondsto the membrane patch spanning the central hole of the pipette. It is quite remarkable to see how this inside-outpatch attaches to the glass being pulled first to the inside of the pipette, but then in the center extends outwardagain. In the image below, a slide suction is applied through the pipette. It does not pull the whole extending partof the membrane, which has a diameter of 1 µm, to the inside, but only pulls a small part in the middle. The heightdifferences seen in the picture are about 200 nm.

Page 149: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

144 ADAPTING AFM TECHNIQUES FOR STUDIES ON LIVING CELLS

transduction, we investigated with the AFMcells from a cancer cell line (HEK 293),which are kept at a certain membrane poten-tial (voltage-clamped) (Mosbacher et al.,1998). We used either normal cells as con-trols, or cells transfected with Shaker K+ ionchannels. In control cells we found move-ments of 0.5 to 5 nm normal to the planeof the membrane. These movements trackeda ±10-mV peak-to-peak AC carrier stimu-lus to frequencies >1 kHz with a phase shiftof 90–120◦, as expected of a displacementcurrent. The movement was outward withdepolarization, and the holding potential wasonly weakly influenced by the amplitude ofthe movement. In contrast, cells transfectedwith a non-inactivating mutant of Shaker K+channels showed movements that were sensi-tive to the holding potential, decreasing withdepolarization between −80 mV and 0 mV.

Further control experiments used open orsealed pipettes and cantilever placements justabove the cells. The results suggested that theobserved movement is produced by the cellmembrane rather than by artificial movementof the patch pipette, acoustic or electricalinteraction of the membrane, and the AFMtip. The large amplitude of the movementsand the fact that they occurred also incontrol cells with a low density of voltage-sensitive ion channels imply the presenceof multiple electromechanical motors. Theseexperiments demonstrate that the AFM mightbe able to exploit the voltage-dependentmovements as a source of contrast forimaging membrane proteins.

8.1.3. AFM Studies on Tissue Sectionsfrom the Inner Ear

Cochlear hair cells of the inner ear areresponsible for the detection of sound. Theyencode the magnitude and time course ofan acoustic stimulus as an electric recep-tor potential, which is generated by a stillunknown interaction of cellular components.In the literature, different models for themechanoelectrical transduction of hair cells

are discussed (Corey and Hudspeth, 1983;Furness et al., 1996; Howard and Hudspeth,1988; Hudspeth, 1983). All hypotheses havein common that a force applied to the so-called hair bundle (which consists of spe-cialized stereocilia at the apical end ofthe cells) in the positive direction, towardthe tallest stereocilia, opens transductionchannels, whereas negative deflection closesthem. For a better understanding of the trans-duction process, it is important to know whatelements of the hair bundle contribute tothe opening of transduction channels andto study their mechanical properties. Themorphology of the hair cells is preciselydescribed by scanning and transmission elec-tron microscopy. Unfortunately, this methodis restricted to fixed and dehydrated speci-men. Therefore, we have extended our AFMdevelopment to enable the study of cochlearhair cells in physiological solution. To pro-vide a clear view onto the tissue section,we built the combined AFM/patch-clampsetup onto an upright differential interfer-ence contrast (DIC) light microscope (Langeret al., 1997). A water immersion objective(40 × /0.75) provides a high resolution ofabout 0.5 µm even on organotypic cell cul-tures with thickness of about 300 µm. Usingthis setup (Fig. 8.6), it is possible to see cil-iary bundles of inner and outer cochlear haircells extracted from six- and eight-day-oldrats and to approach these structures in a con-trolled way with the AFM cantilever tip andthe patch pipette (Fig. 8.7). Images can beobtained by measuring the local force inter-action between the AFM tip and the speci-men surface while scanning the tip (Langeret al., 2000). The question whether morpho-logical artifacts occurred at the hair bun-dles during AFM investigation was clarifiedby preparing the cell cultures for the elec-tron microscope directly after the AFM mea-surements. Forces up to 1.5 nm applied inthe direction of the stereocilia axis did notchange the structure of the hair bundles.

The scan traces shown in Fig. 8.8 repre-sent a typical example of force interaction

Page 150: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

COMBINING AN AFM WITH OPTICAL MICROSCOPY AND ELECTROPHYSIOLOGICAL TECHNIQUES 145

Figure 8.6. Schematic drawing of the setup built for whole cell tissue patch-clamp/AFM applications. A piezo-tubescanner P1 moves the cantilever mounted on a titanium arm in xy and z direction. An xz-translation stage allowsthe lateral and vertical coarse positioning of the cantilever. Additionally, an xy-translation device moves the opticalmicroscope. Thus, in combination with the vertical objective translation stage of the optical microscope, the laserbeam is adjustable in all three orthogonal directions relative to the cantilever. The cantilever deflection is detectedby the laser deflection method. A parallel beam of a laser-diode propagates through the beam-splitter cube S1 and isreduced to a diameter of 1 mm by the convex lens L5 and the concave lens L4. The convex lens L3 focuses the beaminto the back-focal plane of the water immersion objective. Notch filter S2 reflects the laser light to the objectivelens L1. It transmits light below 600 nm and above 700 nm, and it reflects with 98% in between. A Zeiss Achroplan40 × /0.75-W water immersion objective lens creates a parallel laser beam, which is positioned on the cantilever.The reflected laser light propagates the same way back and is reflected by the beam-splitter cube S1 through theconcave lens L8 onto a quadrant photodiode. With the help of this optical setup the shift of the reflected laser spotcaused by a cantilever deflection or torsion is detected. For fine adjustment of the tip-sample distance the specimenchamber is moved vertically by a piezo-stack. A hydraulic micromanipulator, directly mounted on the specimenstage, is used to move the patch-clamp head stage together with the glass pipette in three orthogonal directions.The whole setup is built into an upright optical DIC-Microscope. For infrared DIC imaging the beam of a halogenlight source propagates through an edge filter F1 cutting below 700 nm. The light is focused by lenses L6 and L7

and reflected by mirror S4 on the specimen chamber. The polarizer Pol1, the Wollaston prisms W1 and W2, and theanalyzer An1 produce the DIC contrast. The imaging tubus lens L2 creates an image which is detected by a CCDcamera simultaneously viewed through the 10× eyepiece. It is possible to use all three units (optical microscope,AFM, and patch-clamp setup) simultaneously.

between the AFM tip and the stereocilia.For determination of the stereocilium posi-tion, all 200 scan lines recorded on eachhair bundle were displayed as a greyscaleimage. The central stereocilium of a hair bun-dle was defined as “0” while adjacent stere-ocilia were labeled with numbers starting

from “1” for the left half of the hair bundleand “−1” for the right half of the hair bun-dle. Traces shown represent the force interac-tion in excitatory direction. The stiffness ofthe stereocilia, when pushed, increased from0.73 mN/m and reached a steady-state levelat about 2 mN/m. The mean stiffness of 17

Page 151: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

146 ADAPTING AFM TECHNIQUES FOR STUDIES ON LIVING CELLS

Figure 8.7. Principle of simultaneous AFM and patch-clamp recording composed from three separately taken SEMimages. The central part represents the organ of Corti with V-shaped hair bundles on top. Above the front end ofan AFM cantilever with its tip is placed and below the patch pipette contacting one of outer hair cells can be seen.

200

100nm

0

3.02.5

2.01.5

1.00.5

0.0

4.0

3.0

2.0

1.0

µm

µm

Figure 8.8. The overlayed saw-tooth patterns represent a typical example of force interaction between the AFM tipand the stereocilia. For determination of the stereocilium position, all 200 scan lines recorded on each hair bundlewere displayed as a greyscale image. The central stereocilium of a hair bundle was defined as “0”. At this positionin the experiments with an additional glass capillary attached this capillary was positioned. Adjacent stereocilia werelabeled with numbers starting from “1” for the left half of the hair bundle and “−1” for the right half of the hairbundle. Traces shown represent the force interaction in excitatory direction.

Page 152: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

COMBINING AN AFM WITH OPTICAL MICROSCOPY AND ELECTROPHYSIOLOGICAL TECHNIQUES 147

investigated hair bundles of outer hair cellscan be calculated to be 2.5 m N/m in theexcitatory direction and 3.1 m N/m in theinhibitory direction.

The average force constant showed onlya weak dependence on the stereociliumposition for the excitatory direction. Somestereocilia located at the center showedexceptionally high stiffness, whereas somelocated at the outer region (positions “−7”to “−11”) revealed smaller stiffness.

In inhibitory direction, the mean forceconstant was slightly higher with a standarddeviation being 2.5 times higher comparedto the excitatory direction. The larger forceconstant might be explained by a directcontact between the taller and the adja-cent shorter stereocilia while in excita-tory direction shorter stereocilia are pulledby elastic tip links. These links connectthe rows of taller and shorter stereocilia(Furness and Hackney, 1985; Lim, 1986;Furness et al., 1989; Zine and Romand,1996). The high standard deviation might be

an effect of the variation in spatial interac-tion between taller and shorter stereocilia.Depending on the angle between scan direc-tion and the direction defined by the cen-ters of adjacent taller and shorter stereocilia,mechanical compliance may vary in a widerange.

For the excitatory direction (Fig. 8.9) twodistributions are distinguishable. One popu-lation of stiffness values is located around2.2 mN/m, and a smaller second one islocated around 3.1 mN/m. These stiffnessdata correspond to stereocilia located in thecentral region of investigated hair bundles.Comparing the stiffness plot in Fig. 8.9 andthe corresponding greyscale image (Fig. 8.8),we can find a correlation between arrange-ment and stiffness of the stereocilia. Thestereocilia standing a little apart from theirneighbors show a significantly higher stiff-ness of 3.2 mN/m and 3.1 mN/m, respec-tively.

A possible hypothesis may be that lat-eral links connecting these stereocilia with

14mN/m

12

10

8

6

4

2

0

0 2 4 6 8 10

Stiffness with fiberStiffness without fiber

Figure 8.9. Changes observed in the elastic properties of the stereocilia with a glass fiber attached to the centralstereocilium. Only direct neighbors are significantly affected, pointing to a small effect of side links to the observedstiffness.

Page 153: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

148 ADAPTING AFM TECHNIQUES FOR STUDIES ON LIVING CELLS

their direct neighbors would be oriented indirection of the exerted force. This mightallow transmission of additional force alongthese links to their neighbors.

This hypothesis implies that all otherstereocilia would display much less interac-tion with their neighbors mediated by sidelinks. This was directly addressed by a sec-ond AFM experiment. Force transmissionbetween adjacent stereocilia was examinedby AFM using a lock-in amplifier for detec-tion of the transmitted forces together witha stimulation glass fiber touching an individ-ual pair of stereocilia. The output of the AFMdetector was connected to the input of a lock-in amplifier detecting the vertical oscillationof the AFM cantilever at 357 Hz. The lateralforce FL transmitted by lateral links was cal-culated from the output signal of the lock-inamplifier.

Force interaction between stereocilia andAFM tip led to displacements of stereociliafrom zero to about 250 nm. Therefore,relative displacement between the directlystimulated stereocilium and the stereociliumdisplaced by the AFM tip is expected toresult in continuous stretching of laterallinks located in-between. This in principleallows detection of transmitted forces bylateral links for different states of stretching.Normalized forces rapidly decrease fromthe directly stimulated to the first adjacentstereocilium. Stereocilia located further awayat positions “1” to “8” reveal only a slightdecrease in relative force from 36% to 20%.Obviously, forces transmitted by lateral linksrapidly decrease from a directly stimulated toadjacent stereocilia of rats at postnatal ageday four. This result supports the hypothesisof a weak interaction between stereocilia bylateral links.

From these AFM measurements with theglass fiber attached also the stiffness of stere-ocilia can be determined as described before.We can use this information for detecting themechanical effect of the touching glass fiberon stiffness of adjacent stereocilia. If laterallinks contribute to the stiffness obtained at

individual stereocilia, we would expect tosee an increase in stiffness of adjacent stere-ocilia compared to data shown for stereociliainvestigated without a fiber (Fig. 8.9). Stiff-ness data were calculated only for the exci-tatory direction, where the AFM tip dis-places the stereocilia toward the fiber tip.Not only is the stiffness (filled circles) ofthe directly touched stereocilium at position“0” increased, but also the stiffness of stere-ocilia “1” to “4.” Mean stiffness in excita-tory direction is 4.8 mN/m. This is about 1.9times higher compared to the mean stiffnessof stereocilia not touched with a fiber (opensquarres). For position “7” the stereociliumwith and the stereocilium without a touch-ing glass fiber show approximately identicalstiffness.

These experiments proof that the AFMallows local stiffness measurement on thelevel of individual stereocilia with the mea-sured stiffness representing the local elasticalproperties of the directly touched sterocil-ium and its nearest neighbors. The resultsshow that stiffness depends on the orien-tation of links with regard to the directionof the stimulus and that a stimulating fiberhad only little mechanical effect on adja-cent stereocilia not in direct contact withthe fiber.

For a partial decoupling of the tectorialmembrane from hair bundles of outer haircells, we would therefore suppose that onlystereocilia still in contact with the tecto-rial membrane and their nearest neighborsare displaced by an incoming mechanicalstimulus. Lateral links may not compensateloss in contact with the tallest stereociliaof outer hair cells. Decoupling of the tec-torial membrane following exposure to puretones at high sound pressure levels is sup-posed to protect hair cells and to avoid dam-age (Adler et al., 1993). A strong interac-tion between the tectorial membrane and hairbundles of outer hair cells seems to be essen-tial for the efficacy of the cochlear amplifierand transduction of sound into an electricalsignal.

Page 154: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

COMBINING AN AFM WITH OPTICAL MICROSCOPY AND ELECTROPHYSIOLOGICAL TECHNIQUES 149

Many micromechanical measurementshave already been performed at entire stere-ociliary bundles of sensory hair cells usingthin glass fibers directly attached to thebundle or fluid jets. The receptor poten-tial or transduction current was measuredin response to the displacement of stere-ocilia, but to study the kinetics of a singletransduction channel over the whole rangeof its open probability requires a techniqueallowing stimulating a single stereocilium.As demonstrated already, AFM offers theopportunity to exert a force very locally to anindividual stereocilium. In the experimentsdescribed in the following after supportingcells were removed using a cleaning pipette,a patch pipette filled with intracellular solu-tion (concentrations in mM: KCl 135, MgCl23.5, CaCl2 0.1, EGTA 5, HEPES 5, Na2ATP2.5, pH 7.4) was attached to the lateral wallof one cell of the outermost row of outerhair cell. Thereby, the glass microelectrodeforming a seal on the plasma membrane ofan intact outer hair cell isolates a small patch(Fig. 8.7). This so-called “cell attached” con-figuration is the precursor of the whole-cellconfiguration where the microelectrode isin direct electrical contact with the insideof the cell. For low noise measurementsof single ion channels the seal resistanceshould be typically in the range >1 G�.A pulse of suction applied to the pipettebreaks the patch, thereby creating a hole inthe plasma membrane, and provides access tothe cell interior. During recording the elec-trical resistance between the inside of thepipette and the hair cell should be very small.Many voltage-activated K+-ion channels areembedded in the lipid membrane of outerhair cells. Opening and closing of these chan-nels increases the background noise levelduring transduction current measurements.Therefore, during transduction current mea-surements, the holding potential of the haircell was set to −80 mV, corresponding tothe reversal potential of K+-ion channels.After forming a seal, the AFM tip was movedto the top of the corresponding hair bundle

under light microscopic control. The AFMtip successively displaced each stereociliumwithin a hair bundle as described before. Incontrast to force transmission measurements,a sinusoidal voltage was added to the nor-mal AFM scan signal modulating the AFMtip in horizontal direction with 190 nm at98 Hz. Thus, the hair bundle was slightly dis-placed several times while interacting withthe lateral face of the AFM tip. The AFMtip repeatedly scanned in the same line whileapproaching the hair bundle toward the AFMtip. As expected, an inward current was notdetected until displacement of a stereociliumin excitatory direction (Fig. 8.10).

In the experiments described before only aweak transmission of force from the directlystimulated stereocilium to adjacent stere-ocilia was observed implying that only fewchannels are opened. These results of elas-ticity measurement can be confirmed byelectrophysiological findings, as shown inFig. 8.10, where a set of transduction cur-rent measurements is displayed. The tipof an AFM cantilever repeatedly scannedacross the same stereocilium of an outerhair cell. Applied horizontal forces of upto 0.8 nN (upper graphs) resulted in stere-ocilia displacements of about 350 nm inexcitatory and 250 nm in inhibitory direc-tion. The AFM tip displaces the stereocil-ium in excitatory direction between 89 and131 ms, thereby opening transduction chan-nels (middle graph). The current amplitudewas determined for the period of interactionwith the AFM tip, and the values for excita-tory and inhibitory direction are put togetherin the histograms shown in Fig. 8.10 (lowergraph). The distribution around 0 pA corre-sponds to the closed state of the channels,while the distribution around 19.1 pA corre-sponds to the maximal activation. Compar-ing this current amplitude to the maximumtransduction current of about 462 pA mea-sured for entire hair bundles of postnatalmice (Geleoc et al., 1997) confirms that theAFM is well-suited for local stimulation ofindividual stereocilia.

Page 155: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

150 ADAPTING AFM TECHNIQUES FOR STUDIES ON LIVING CELLS

0.6

0.4

0.2

0.0

0 50 100 150 200

Time (ms)

Excitatory

For

ce [n

N]

0.6

0.4

0.2

0.0

250 300 350 400 450

Time (ms)

0 50 100 150 200

ms

250 300 350 400 450

ms

Inhibitory

For

ce [n

N]

40

30

20

10

0

−40 −20 0 20 40

Current [pA]

−40 −20 0 20 40

Current [pA]

Fre

quen

cy

70

605040

30

20

10

0

Fre

quen

cy

20 pA

Figure 8.10. The upper two graphs show the force applied to a stereocilium while scanning either in excitatory(left) or inhibitory (right) direction. The observed current response is depicted in the traces in the middle of thepicture. A reproducible opening of transduction channels between 89 ms and 131 ms can be seen in the left column.In contrast, force applications in the inhibitory direction did not result in the opening of transduction channels. In thelower section of the image, histograms of the currents measured at 89–131 ms and 366–407 ms are shown for bothscanning directions. The distribution around zero represents the current for the closed state of the channel, while thesecond, smaller distribution seen only in excitatory direction around 19 pA represents the current for the open stateof the transduction channel.

Page 156: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

PHOTONIC FORCE MICROSCOPY (PFM) 151

8.2. PHOTONIC FORCE MICROSCOPY(PFM)—REPLACING THEMECHANICAL AFM CANTILEVER BYAN OPTICAL TRAP

The AFM really turned out to be a verygood tool for studies of live cells. How-ever, cells have a rough surface, which oftenprevents the tip of a mechanical cantileverfrom following fine topographic details. Fur-thermore, forces of some 10 N are nec-essary for stable imaging, which is oftenstrong enough to cause serious deformationor even destruction of soft cellular structures.Only stiff structures, such as the membranecytoskeleton, are clearly visible in the pic-tures. Imaging inside cells is impossible dueto the mechanical connection to the imagingtip. Therefore, a scanning probe microscopewithout a mechanical connection to the tipand working with extremely small loadingforces is desirable. We developed such aninstrument, the photonic force microscope(PFM) (Florin et al., 1997, 1998; Pralk et al.,1998, 1999).

In case of the PFM the mechanicalcantilever of the AFM is replaced by the

three-dimensional trapping potential of alaser focus (Fig. 8.11). The possibility totrap small particles with high stability inthe focus of a laser was first describedby Ashkin (1986). In the PFM a nano- tomicrometer-sized particle (e.g., latex, glass,or metal bead) is used as a tip. The differencein the refractive index of the medium andthe bead, the diameter of the bead, thelaser intensity, and the intensity profile inthe focal volume determine the strengthof the trapping potential. Depending onthe application, the potential is adjusted bychanging the laser power. Usually the springconstant of the potential is two to three ordersof magnitude softer than that of the softestcommercially available cantilevers.

8.2.1. Setting Up a PFM

The PFM can make use of two position-sensing systems to determine the positionof the trapped sphere relative to that of thepotential minimum. One records the fluo-rescence intensity emitted by fluorophoresinside a trapped latex or glass sphere, whichare excited by the trapping laser via a two-photon process. The fluorescence intensity

Figure 8.11. Schematic drawing of the photonic force microscope setup on top of an inverted optical microscope.The probe, a nanometer-sized spherical particle, is trapped by optical forces, and its position is measured with respectto laser focus using the interference of the laser light with the light scattered by the particle, which is detected by aquadrant photodiode in the back focal plan of the condenser.

Page 157: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

152 ADAPTING AFM TECHNIQUES FOR STUDIES ON LIVING CELLS

provides an axially sensitive position signalwith millisecond time and 10- to 20-nm spa-tial resolution. The other position detectionsystem is based on the interference of theforward-scattered light from the trapped par-ticle with the unscattered laser light at aquadrant photodiode. This provides a fastthree-dimensional recording of the particle’sposition, and it is most sensitive perpendic-ular to the optical axis. The position of thebead can be measured with a spatial resolu-tion of better than 1 nm at a temporal reso-lution of a microsecond or better. Dependenton the application, one or the other (or both)of these detection systems is used.

At room temperature, the thermal posi-tion-fluctuations of the trapped bead in weaktrapping potentials reach several hundrednanometers. At first glance, these fluctuationsseem to be disturbing noise that limits theresolution. However, due to the speed andresolution of the position sensor based onthe forward-scattered light detection, thefluctuations of the bead can be trackeddirectly, opening new ways to analyze theinteraction of the bead with its environment.

The PFM developed is based on aninverted optical microscope with a highnumerical aperture objective lens providinga good optical control of the investigatedstructures. The laser light is coupled intothe microscope using techniques known fromlaser scanning microscopy and is focusedby the objective lens into the specimenplane. A 1064-nm Nd:YVO4 laser is usedsince neither water nor biological materialhas significant absorption at this wavelength.A dichroic mirror behind the condenserdeflects the laser light onto the quadrantphotodiode. The difference between the leftand right half of the diode provides thex position, the difference between upperand lower half provides the y position,and the sum signal provides the z-position.For displacements that are small relativeto the focal dimensions, the signals changelinearly with the position of the bead inthe trap.

The trapped particle acts as a Brownianparticle in a potential well, and its positiondistribution is therefore described by E(r) =−kT ∗ ln p(r) + E0..

The so-called Botzmann distributions p(r)

is readily measured with the high spatialand temporal resolution available of thePFM. Therefore the three-dimensional trap-ping potential E(r) can be determined justby knowing the temperature. A precision ofone-tenth the thermal energy kT is achievedwith a sufficient number of statistically inde-pendent position readings.

Scanning either the laser focus or thesample, the PFM can be used in a mannervery similar to that of a conventional AFMto image surface topographies (Fig. 8.12).The applied forces range from a few pico-newtons down to fractions of a pico-newton.The resolution is determined by the inter-action area of the sphere used with thesample and by the thermal fluctuations ofthe bead. The PFM, like an AFM, can beused in different imaging modes. Besidesthe constant height mode, implementing afeedback loop for force control, also theconstant force mode can be used. Thismode provides, besides the force and theerror signal, also the measurement of lat-eral friction forces. Furthermore, due to thelarge thermal fluctuations of the bead, anew imaging mode becomes feasible withthe PFM by observing the spatial distribu-tion of the thermal fluctuations of the bead,which avoids stationary objects. In this way,it is possible to image, for example, thethree-dimensional (3-D) structure of polymernetworks (Fig. 8.13), with a resolution nowlimited only by the precision of the detectionsystem. Additionally, detailed informationabout the interaction potential between thebead and the surface of the objects imagedcan be obtained. At present, PFM imagingof biological material is limited by nonspe-cific adhesion events between the bead andthe sample, which can lead to the loss ofthe probe due to the weak forces of thelaser focus. Although there seems to be no

Page 158: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

PHOTONIC FORCE MICROSCOPY (PFM) 153

Figure 8.12. Constant height imaging with the photonic force microscope.

Figure 8.13. Three-dimensional thermal noise image of an agar network. The volume accessible to the beadfluctuations at a certain probability is shown as black space. The nonaccessible volume corresponding to the polymerfibers is rendered as a solid body. The minimal channels observed by the bead reflect its diameter of 216 nm.

general strategy to prevent nonspecific adhe-sion to complex biological material, it is pos-sible to achieve specific binding, for instance,to membrane and other molecular structuresand to track their motion.

8.2.2. Using Thermal Fluctuationsto Measure Mechanical Propertiesof Single Molecules

Kinesins and kinesin like-proteins are ofwide interest in biology because of their

fundamental functions in the cell. Theyare responsible, for example, for targetingorganelles through the cells and for setting upthe mitotic spindle during cell division. Thedirected transport along the cytoskeletal fila-ments of microtubules is powered in an ATP-dependent way by these molecular motors.Conventional kinesin is a so-called plus-end-directed motor, able to transport organellesin a defined direction over a distance ofup to several microns as a single molecule.

Page 159: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

154 ADAPTING AFM TECHNIQUES FOR STUDIES ON LIVING CELLS

Structural and biochemical data of kinesinreveal a heavy chain folded as a globularN-terminal motor domain containing an ATPand a separate microtubule-binding site. Thecatalytic domain is followed by a neck regionthat consists of two short β-sheets and acoiled-coil helical structure probably respon-sible for dimerization. Behind this region,a hinge of variable length that is not pre-dicted to form a coiled-coil is followed bythe kinesin stalk that apparently is a α-helicalcoiled-coil interrupted by another hinge. Thestalk finally connects to a poorly conservedtail structure that interacts with its lightchain, binding to the cargo. Several studieson kinesins (e.g., Grummt et al., 1998) haveshown that the neck and the first hinge regionof the motor play an important role in kinesindirectionality, velocity, and ATPase activity.Most of the molecules dimerize in vivo andtherefore have two enzymatic head domains.The mechanism of movement and force gen-eration required for intracellular transportsis not yet fully understood. Several authors(e.g., Howard et al., 1989) suggest a hand-over-hand model where both heads wouldalternate to “walk” along the filaments. Hav-ing all its enzymatic and binding machineryin its heavy chain, the kinesin motor hasthe unique property of operating completelyon its own, either as a dimer or even as amonomer. Earlier studies on single moleculeshave shown that the maximum force gener-ated by the kinesin molecule to transport amicrosphere along a microtubule is in therange of 5 pN (Svoboda and Block, 1994).

Obviously, characterizing mechanical pro-perties of molecular motors is essential for abetter understanding of how nano-machines,like kinesin, convert chemical energy intoa mechanical movement. So far, attemptsto explain in a satisfying way the functionof kinesin using the knowledge gained bydynamical and structural studies have notbeen very successful. A major problem isthat the high-resolution structural data areobtained on an ensemble of molecules in arigid state and cannot resolve the dynamic

of intermediate states required for the kinesinmotility. Furthermore, the molecules used forstructural analysis are truncated constructsmissing the stalk. On the other hand, forin vitro motility assays, full-length constructsof the motors are used in an environmentrather different from a protein crystal.

We focused our studies with the PFMon intrinsic mechanical properties of kinesinlike its elasticity, comparing the mechan-ical behavior of two different full-lengthkinesin constructs by adsorbing them ontoglass microspheres and letting them inter-act with a microtubule in the presence ofdifferent nucleotides. One type of motorwe study is the wild type from drosophilawith two head groups. Another type is achimera missing one of the heads. To deter-mine the mechanical properties of the kinesinmolecule bound to a bead and to a micro-tubule in thermal equilibrium, the thermalposition fluctuations of the bead are mea-sured using the PFM. Boltzmann’s equationcan be used to analyze these fluctuations in3-D and to determine the energy landscapedefined by the kinesin as a molecular linkerbetween bead and microtubule structure fixedto the surface. Finally, from the energy land-scape measured (Fig. 8.14), the mechani-cal properties of single kinesin moleculesin 3-D for different types of motors andtheir different nucleotide binding states canbe extracted (Jeney et al., 2001). The mea-sured stiffness along the tether axis can bethen explained as elasticity of this structure.The rotational stiffness corresponds to restor-ing forces of the molecular hinges againstthe lateral bending due to thermal energy.The restoring forces the molecules developagainst lateral bending are dependent of themicrotubule orientation and are influencedby the microtubule’s presence. A comparisonbetween one- and two-headed motors showsthat a two-headed motor behaves stiffer inall three directions. It points to the influ-ence of the second head, which restricts thebead movement due to steric interactionswith the microtubule and perhaps in cases

Page 160: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

PHOTONIC FORCE MICROSCOPY (PFM) 155

Microtubule

Microtubule

Laser focus

Figure 8.14. The isoenergy surfaces determined by the Boltzmann equation using the position probability data of aparticle in the laser focus is shown in the left part of the figure. The schematic drawing shows the binding of thisparticle via a kinesin molecular motor to a microtubule structure fixed on the coverslip. In the upper right part of thefigure, the change of the isoenergy surface after the binding event is shown in scale to the surface before binding.

of the observed high stiffness values in theATP-free state even binds to it.

The stiffness of the motor bound with thenonhydrolyzable ATP analogue AMPPNPare much higher than the ones in the ADPbound states for one- and two-headed motors.This suggests that the binding strengthbetween kinesin and tubulin influences theelasticity of the whole kinesin/bead/micro-tubule-system. It corresponds well with thefinding that different nucleotides influencethe position of the free domain relative tothe bound one (Hirose et al., 1995; Arnal andWade, 1998).

8.2.3. Using Thermal Fluctuations toDetermine Local Membrane Viscosity

The motion of a thermally fluctuating parti-cle in a harmonic potential like that of thelaser trap can be characterized to a certainlevel by an exponentially decaying positionautocorrelation function. This function has acharacteristic autocorrelation time τ = γ/κ,

where γ denotes the viscous drag on thesphere and κ denotes the force constant ofthe optical trap. Thus, the local viscous dragand the diffusion coefficient D = kT /γ of asphere in a harmonic potential can be calcu-lated from the measured autocorrelation timeof the motion and the stiffness of the trappingpotential. The stiffness of the trapping poten-tial is determined as described before usingBoltzmann’s equation. To measure diffusioncoefficient with a statistical error smaller than10%, the observation interval must be about1000 times longer than the autocorrelationtime. Hence, the motion of the bead limits thetemporal resolution of the viscosity measure-ment, and not the bandwidth of the detectionsystem. Near surfaces (e.g., glass surfacesor cell membranes), the diffusion is reducedbecause of the spatial confinement (Happeland Brenner, 1965). Within an obstacle-freelipid bilayer, diffusion has been described bySaffman and Delbruck (1975) using a hydro-dynamic model treating the bilayer as a con-tinuum and assuming weak coupling to the

Page 161: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

156 ADAPTING AFM TECHNIQUES FOR STUDIES ON LIVING CELLS

surrounding liquid medium. The viscous dragγm on a cylindrical particle with radius r in ahomogeneous lipid bilayer of thickness h isγm = 4πηmh/(ln(ηmh/ηwr) − ε), where ηw

denotes the viscosity of the surrounding fluid,ηm is the viscosity of the lipid bilayer, andε is Euler’s constant. This approximation isvalid for proteins with radii large comparedto the size of the lipid molecules and forηm � ηw, which is true for cellular mem-branes. Thus, the membrane viscosity canbe determined by binding a bead to a mem-brane structure of known diameter and mea-suring the total viscous drag on the bead asdescribed above (Fig. 8.15). Such measure-ments can be performed with a temporal res-olution of 0.3 s and within areas of 100 nmin diameter.

The measured viscous drag γ for thebead connected to a membrane componentis the sum of the Stokes drag of thesphere γs = 6πηwr and the viscous dragγm of, for example, a single protein inthe lipid bilayer. The Stokes drag of thesphere near the cell membrane increases dueto the confinement and has to be takeninto account for the correct calculation ofγm. The observation of the strength of thelateral potential ensures that the observedmembrane component diffuses freely.

Using the PFM technique on a singletransmembrane protein of known size, it ispossible to determine the membrane viscos-ity in living cells in areas about 100 nm in

diameter, which translates into a diffusioncoefficient of D = 3.9 × 10−8 cm2/s at 36◦C.The obtained local diffusion coefficients ofproteins in the plasma membrane in intactcells agree for the first time well withthe Saffman–Delbruck model validated byPeters and Cherry (1982) in dimyristoylphos-phatidylcholine (DMPC) bilayers usingFRAP to measure the diffusion coefficientof bacteriorhodopsin to be between 0.15 ×10−8 cm2/s and 3.4 × 10−8 cm2/s, depend-ing on the protein/lipid ratio.

Once the membrane viscosity has beenobtained, the technique allows us to deter-mine the diameter of other membrane struc-tures of unknown size and to monitorcontinuously their diffusion characteristics.We performed such experiments to determinethe size of membrane structures called “rafts”(Pralle et al., 2000). Lipid rafts are involvedin the polarized sorting of proteins and cel-lular signaling (Simons and Ikonen, 1997;Keller and Simons, 1998). According to ourstudy in the plasma membrane of fibroblast-like cells, these structures are indeed quitestable on the timescale of minutes, justifyingthe model of “rafts” as lipid–protein com-plexes floating raft-like in the membrane.Their diameter was determined to be about50 nm. The broad distribution of viscousdrag values observed for the rafts studiedindicates that even one raft type in one cellmight have a distribution of sizes and thatthe size of a single raft might be dynamic.

Bead coated with Ab and FSG

HA-trimer

20 nm

20 nm0

Figure 8.15. Scaled model of the experimental situation, when a sphere (r = 108 nm) is bound via an adsorbedantibody to a transmembrane protein. The lipid bilayer is symbolized by the gray double line.

Page 162: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 157

Transferring these values to other cell typesand non-GPI–anchored proteins might notbe possible since raft size and stability aremost likely dependent on the lipid and pro-tein constituents. Indeed, recent data suggestthat rafts in the apical membrane of MDCKbehave differently from rafts in fibroblasts(Verkade et al., 2000).

According to our study, rafts in the plasmamembrane of fibroblast-like cells diffuse asa rather stable platform with an averagearea of 2100 nm2. The size estimate allowsan assessment of the maximal contents ofone raft. If these were composed purely oflipid molecules, having a radius compara-ble to phosphoethanolamine (r = 0.44 nm),one raft would contain almost 3500 lipidmolecules. How many proteins a raft con-tains depend on how densely packed theproteins would be. If they were as denselypacked as rhodopsin molecules are in frogrods (Blasie and Worthington, 1969), or asdensely packed as the spikes in the enve-lope of Semliki Forest virus (Cheng et al.,1995), a raft would contain 55–65 proteins,respectively. Clearly, the packing density in alipid raft in a mammalian plasma membranewould be lower.

The consequences of a small raft sizeand stable association is that proteins withinrafts are restricted in their interactions withother proteins. To use rafts as platforms inmembrane trafficking (e.g., in apical trans-port from the Golgi complex) would implythat these rafts have to be clustered togetherby an apical sorting machinery to form acontainer comprising several rafts. A grow-ing body of evidence implicates lipid raftsin signal transduction. Well-studied examplesinclude IgE receptor signaling and T- and B-cell activation. If rafts normally contain onlya limited set of proteins, then clustering ofrafts would be necessary to achieve the con-centration of interacting molecules requiredto elicit a signal above the activating thresh-old. There is ample indication that clusteringis an essential feature of signal transductionprocesses involving rafts.

In these experiments that finally led to aphysical proof of the existence of rafts, thePFM has proven to be a powerful tool tostudy biophysical properties of the plasmamembrane as well as interaction of singlemolecules or complexes of molecules withinthe membrane. Future applications of thePFM will cover a wide range from 3-Din vivo imaging of biological samples toa detailed investigation of single-moleculeproperties.

REFERENCES

Adler, H. J., Poje, C. P., and Saunders, J. C.(1993). Hearing Res. 71:214.

Arnal, I., and Wade, R. H. (1998). Structure 6:33.

Ashkin, A., (1986). Opt. Lett. 11:288.Binnig, G., Quate, C. F., and Gerber, C. (1986).

Phys. Rev. Lett. 56:930.

Blasie, J. K., and Worthington, C. R. (1969).J. Mol. Biol. 39:407.

Cheng, R. H., Kuhn, R. J., Olson, N. H., Ross-man, M. G., Choi, H.-K., and Baker, T. S.(1995). Cell. 80:621.

Corey, D. P., and Hudspeth, A. J., (1983).J. Neurosci., 3:962.

Drake, B., Prater, C. B., Weisenhorn, A. L.,Gould, S. A. C., Albrecht, T. R., Quate, C. F.,Cannell, D. S., Hansma, H. G., and Hansma,P. K. (1989). Science 243:1586.

Florin, E.-L., Pralle, A., Horber, J. K. H., andStelzer, E. H. K. (1997). J. Struct. Biol. 119:202.

Florin, E.-L., Pralle, A., Stelzer, E. H. K., andHorber, J. K. H. (1998). Applied Phys. A,66:S75.

Frischknecht, F., Moreau, V., Rottger, S., Gon-floni, S., Reckmann, I, Superti-Furga, G., andWay M. (1999). Nature 401:926.

Furness, D. N., and Hackney, C. M. (1985).Hearing Res. 18:177.

Furness, D. N., Richardson, G. P., and Rus-sell, I. J. (1989). Hearing Res. 38(1–2):95.

Furness, D. N., Hackney, C. M., and Benos, D. J.(1996). Hearing Res. 93:136.

Geleoc, G. S. G., Lennan, G. W. T., Richardson,G. P., and Kros, C. J. (1997). Proc. R. Soc.Lond. B 264:611.

Page 163: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

158 ADAPTING AFM TECHNIQUES FOR STUDIES ON LIVING CELLS

Grummt, M., Woehlke, G., Henningsen, U.,Fuchs, S., Schleicher, M., and Schliwa, M.(1998). EMBO J. 17:5536.

Haberle, W., Horber, J. K. H., and Binnig, G.(1991). J. Vac. Sci. Technol. B9:1210.

Haberle, W., Horber, J. K. H., Ohnesorge, F.,Smith, D. P. E., and Binnig, G. (1992). Ultra-microscopy 42–44:1161.

Happel, J., and Brenner H. (1965). Low ReynoldsNumber Hydrodynamics, Prentice Hall, Engle-wood Cliffs, NJ.

Hirose, K., Lockhart, A., Cross, R. A., and AmosL. A. (1995). Nature 376:277.

Horber, J. K. H., Haberle, W., Ohnesorge, F.,Binnig, G., Liebich, H. G., Czerny, C. P.,Mahnel, H., and Mayr, A. (1992). ScanningMicroscopy 6:919.

Horber, J. K. H., Mosbacher, J., Haberle, W.,Ruppersberg, P., and Sakmann, B. (1995).Biophys. J. 68:1687.

Howard, J., and Hudspeth, A. J. (1988). Neuron,1:189.

Howard, J., Hudspeth, A. J., and Vale, R. D.(1989). Nature 342:154.

Hudspeth, A. J. (1983). Annu. Rev. Neurosci.6:187.

Jeney, S., Florin, E.-L., and Horber, J. K. H.(2001). Methods in Molecular Biology; KinesinProtocols, (I. Vernos, ed.), Humana Press,Totowa, NJ.

Keller, P., and Simons, K. (1998). J. Cell Biol.140:1357.

Langer, M. G., Offner, W., Wittmann, H., Flos-ser, H., Schaar, H., Haberle, W., Pralle, A.,Ruppersberg, J. P., and Horber, J. K. H.(1997). Rev. Sci. Instrum. 68:2583.

Langer, M. G., Koitschev, A., Haase, H., Rex-hausen, U., Horber, J. K. H., and Ruppers-berg, J. P. (2000). Ultramicroscopy 82:269.

Lim, D. J. (1986). Hearing Res. 22:117.

Mosbacher, J., Langer, M., Horber, J. K. H., andSachs, F. (1998). J. Gen. Physiol. 111:65–74.

Moss, B. (1986). Replication of poxviruses.In: Fundamental Virology (B. N. Fields andD. M. Knape, eds.), Raven Press, New York.

Ohnesorge, F. M., Horber, J. K. H., Haberle, W.,Czerny, C.-P., Smith, D. P. E., and Binnig, G.(1997). Biophys. J. 73:2183.

Peters, R., and Cherry, R. J. (1982). Proc. Natl.Acad. Sci. USA 79:4317.

Pralle, A., Florin, E.-L., Stelzer, E. H. K., andHorber, J. K. H. (1998). Applied Phys. A66:S71.

Pralle, A., Prummer, M., Florin, E.-L., Stelzer,E. H. K., and Horber, J. K. H. (1999). Microsc.Res. Technique 44:378.

Pralle, A., Keller, P., Florin, E.-L., Simons, K.,and Horber, J. K. H. (2000). J. Cell Biol.,148:997.

Saffman, P. G., and Delbruck, M. (1975). Proc.Natl. Acad. Sci. USA 72:3111.

Simons, K., and Ikonen, E. (1997). Nature387:569.

Stokes, G. V. (1976). J. Virol. 18(2):636.

Svoboda, K., and Block, S. M. (1994). Cell77:773.

Verkade, P., Harder, T., Lafont, F., andSimons, K. (2000). J. Cell Biol. 148:727.

Zine, A., and Romand., R. (1996). Brain Res.721:49.

Page 164: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

CHAPTER 9

INTERMOLECULAR FORCES OF LEUKOCYTEADHESION MOLECULESXIAOHUI ZHANG AND VINCENT T. MOYDepartment of Physiology and Biophysics, University of Miami School of MedicineMiami, Florida

9.1. INTRODUCTION

Leukocytes (i.e., white blood cells) are foundin the systemic circulation. Their primaryroles are to aid in tissue repair and hostdefense against invading pathogens. How-ever, when left unchecked, the accumulationof leukocytes may damage the tissue as inthe case of inflammatory diseases such asrheumatoid arthritis, asthma, and atheroscle-rosis. In order for leukocytes to fulfill theirfunctions, they must leave the circulation andmigrate through the tissue toward their tar-gets. This process, termed extravasation, ismediated by leukocyte adhesion molecules(Springer, 1990).

In order to leave the bloodstream, leuko-cytes must first attach to and subsequentlypermeate through the wall of blood ves-sels. Leukocyte extravasation involves fourstages: tethering or rolling, cell activation,firm adhesion, and finally transmigration(Fig. 9.1). Each stage engages differentsets of adhesion molecules (Springer, 1994;Kubes and Kerfoot, 2001). Leukocyte rollingis mediated by selectin molecules thatare expressed on vascular endothelial cellsand by their adhesive partners (e.g., P-selectin glycoprotein ligand-1 (PSGL-1)) on

leukocytes. Adhesion mediated by selectinsis relatively weak and transient. These bondsare not strong enough to stop leukocytesfrom the blood flow. Rather, a continu-ous cycle of the formation and dissocia-tion of selectins/ligand complexes permitsthe leukocyte to roll along the endothelium.Rolling enables leukocytes to survey theendothelial surface for chemotactic signals.When appropriate signals are encountered,these signals activate a second class of leuko-cyte adhesion molecule, the integrins. Theactivated integrins bind tightly to their adhe-sive partners, which are molecules of theimmunoglobulin superfamily (IgSF) that areexpressed on the endothelial surface. Theseinteractions arrest the rolling leukocytes.Integrins important for leukocyte firm adhe-sion include leukocyte function-associatedantigen-1 (LFA-1) and very late antigen-4(VLA-4) (Kubes and Kerfoot, 2001). Theirmajor endothelial ligands are intercellularadhesion molecule-1 (ICAM-1) and vascularcell adhesion molecule-1 (VCAM-1), respec-tively (Fig. 9.1).

Leukocyte adhesion has been extensivelystudied for more than two decades. Sequenceand structure of many leukocyte adhesionmolecules have been determined. However,

Force Microscopy: Applications in Biology and Medicine, edited by Bhanu P. Jena and J.K. Heinrich Horber.Copyright 2006 John Wiley & Sons, Inc.

159

Page 165: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

160 INTERMOLECULAR FORCES OF LEUKOCYTE ADHESION MOLECULES

vascular endothelium

tether/roll activationfirm adhesion

transmigration

Blood Flow

PSGL-1−P-selectin VLA-4−VCAM-1

Leukocyte

VLA-4−VCAM-1 LFA-1−ICAM-1

Figure 9.1. Recruitment of leukocytes in inflamed tissues. Adhesion molecules including selectins, immunoglobulinsuperfamily members, and integrins, mediate the initial tethering/rolling, firm adhesion, and transmigration of leuko-cytes. Leukocytes are captured from the circulating blood by transient VLA-4–VCAM-1 or selectin-glycan-basedinteractions. Following activation by chemokines, the leukocytes adhere firmly to the endothelial cells via inte-grin-mediated (i.e., VLA-4 or LFA-1) adhesion. The selectins (P-, E-, and L-selectins) are a family of mem-brane-bound glycoproteins that bind a carbohydrate structure called sialyl Lewis X presented on their ligands.Integrins are αβ heterodimeric adhesion molecules that bind the immunoglobulin superfamily cell adhesion molecules(CAMs).

it is not known how these molecular struc-tures give rise to the mechanical strength ofthe leukocyte adhesion complexes. Recently,we applied the atomic force microscopy(AFM) (Binnig et al., 1986) to determinethe effects of a pulling force on selectinand ingetrin–ligand complexes. Similar app-roaches, including the biomembrane forceprobe (BFP) (Evans, 2001), have beenused to study the unbinding of the strep-tavidin–biotin complex (Florin et al., 1994;Merkel et al., 1999), complementary strandsof DNA (Lee et al., 1994), and otheradhesion systems (Merkel, 2001). Thesedirect force measurements have provided themeans to probe the dissociation pathwayof biomolecular complexes. In this review,we discuss the application of AFM on theunbinding of cell adhesion complexes.

9.2. SINGLE-MOLECULE UNBINDING:THEORY AND APPLICATIONS

9.2.1. Theory

The theoretical framework for understandinghow a pulling force affects the dissociationof an adhesion complex was first formulatedby Bell (1978) and later expanded on byEvans and other researchers (Evans andRitchie, 1997). The Bell model describes the

influence of an external force on the rate ofbond dissociation. This model is based on theconventional transition state theory, in whicha molecular complex needs to overcomean activation energy barrier before finalseparation. If only a single barrier dominatesthe dissociation process, the dissociation rateof this interaction is given by

koff = αkBT

hexp

{−�G∗

kBT

}(9.1)

where �G∗ is the activation energy, T

is absolute temperature, kB is Boltzmann’sconstant, h is Planck’s constant, and α isa prefactor that characterizes the potentialwell. When the complex is exposed to apulling force, the applied force adds a −f x

term to the potential of the system. If thepotential barrier is steep, adding this term tothe free energy reduces the activation barrierby approximately f γ, where γ is the distancebetween the bound state and the transitionstate along the reaction coordinate. Thus,the force dependent dissociation rate of thecomplex is given by

koff(f ) = αkBT

hexp

{−(�G∗ − f γ)

kBT

}

= k0 exp

{f γ

kBT

}(9.2)

Page 166: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

SINGLE-MOLECULE UNBINDING: THEORY AND APPLICATIONS 161

where k0 is the unstressed dissociation rate.Hence, the Bell model predicts that thedissociation rate of the complex increasesexponentially with a pulling force. The twoparameters k0 and γ are often referred to asthe Bell model parameters. These two param-eters characterize the energy potential of theprotein–ligand complex. k0 characterizes thedepth of the potential, and γ characterizesthe width of the potential and dictates theforce resistance of a molecular complex. Ifa complex has a small γ (i.e., the activationpotential is narrow), then an external forcewill have less effect on its force dependentdissociation rate koff(f ). On the other hand, ifthe activation potential is wide, the complexwill be sensitive to an external force sincef γ adds a larger term to the intermolecularpotential.

Equation (9.1) describes how bond dis-sociation is changed by a constant pullingforce. However, a constant pulling force isdifficult to maintain in an AFM experiment.Instead, a dynamic force approach is gener-ally used to characterize the forced dissoci-ation of ligand–receptor complexes (Evansand Ritchie, 1997). Under conditions of aconstant loading rate rf, the probability den-sity function for the forced unbinding of an

adhesion complex is given by

P(f ) = k0 exp

{γf

kBT

}exp

{k0kBT

γrf

×[

1 − exp

(γf

kBT

)]}. (9.3)

From Evans and Ritchie (1997), the mostprobable unbinding force f ∗ (i.e., the maxi-mum of the distribution ∂P (f )/∂f = 0) is

f ∗ = kBT

γln

k0kBT

}+ kBT

γln

{rf

}(9.4)

Equation (9.4) shows that f ∗ is a linearfunction of the logarithm of the loading rate.The Bell model parameters are obtained fromthe plot of f ∗ versus ln(rf ), the dynamicforce spectrum (DFS) of the complex (Evansand Ritchie, 1997).

The energy landscape of a complex mayconsist of multiple sharp activation barriers(Fig. 9.2A) (Evans and Ritchie, 1997). Inthis case, the DFS is predicted to have mul-tiple linear regimes with ascending slopes,as shown in Fig. 9.2B. The increase inslope from one regime to the next indicatesthat an outer barrier has been suppressed

TS1

TS2

Ene

rgy

Reaction coordinate

force = 0

force > 0

force >> 0

bound complex dissociated complex

For

ce

Loge (Loading Rate)

A B

G*

Figure 9.2. Dynamic force spectroscopy. (A) Effects of an applied force on a protein–ligand interaction potentialconsisting of two transition states: TS1 and TS2. In the absence of an applied force (top trace), the dissociation kineticsof the complex are determined by the outer energy barrier (i.e., TS2). An external force tilts the energy potential andsuppresses the outer barrier (middle trace). Further increase in force results in a potential that is governed by theproperties of the inner energy barrier (i.e., TS1) (bottom trace). (B) Two linear regimes are predicted for a cascadeof two sharp energy barriers. The increase of slope indicates that the outer barrier has been suppressed and that theinner barrier has become the dominant kinetic impedance to detachment.

Page 167: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

162 INTERMOLECULAR FORCES OF LEUKOCYTE ADHESION MOLECULES

by force and that an inner barrier dom-inates the dynamic response of the com-plex (Fig. 9.2A). This theory is supportedby recent experiments from different groups(Merkel et al., 1999; Evans et al., 2001;Zhang et al., 2002; Li et al., 2003). Multi-ple activation barriers were found in a num-ber of ligand–receptor systems, including the(strep)avidin/biotin complexes and all testedintegrin–ligand complexes. A partial list ofthese studies is tabulated in Table 9.1.

9.2.2. Force Unbinding of IndividualSelectin/sLeX Complexes

DFS has been employed by us and oth-ers to characterize the mechanical prop-erties of selectin/ligand complexes (Evanset al., 2001; Zhang et al., 2002, 2004; Liet al., 2003). A series of representative AFMforce scans (i.e., force versus piezo dis-placement) are shown in Fig. 9.3A. In thesemeasurements, the two interacting surfaceswere the AFM tip, functionalized with sia-lyl Lewis X, a carbohydrate ligand for allselectins, and a substrate coated with P-selectin. Figure 9.3B presents the DFS ofthe P-selectin–sLeX complex (Zhang et al.,2004). The force spectrum can be dividedinto two loading regimes: The unbindingforce increased exponentially with loadingrate from 70 to 9000 pN/s. Beyond 9000pN/s, a faster exponential increase is clearlyevident. Fitting Eq. (9.4) to both the slow andfast loading regimes yielded the Bell modelparameters for the outer and inner activa-tion barriers of the complex, respectively.For the P-selectin–sLeX complex, the bar-rier widths γ for the outer and inner barrierswere 4.5 A, and 0.80 A, respectively. Similarresults were reported for the forced unbind-ing of L-selectin–PSGL-1 complex (Evanset al., 2001).

9.2.3. Probing Integrin Activationon Live Leukocytes

The regulation of integrin affinity plays animportant role in leukocyte extravasation.

The strength of the integrin–ligand bondis drastically increased when the integrinmolecule is activated by intracellular signals.In order to access the bond strength of indi-vidual integrin–ligand complexes in differ-ent affinity states, it is best to maintain theintegrins in their native environments—thatis, on the surface of live cells. To achievethis goal, we have developed an experimen-tal protocol to couple a live leukocyte ontothe AFM cantilever (Figs. 9.4A and 9.4B)(Zhang et al., 2002; Li et al., 2003) based onan earlier work by Benoit et al. (2000). Thisapproach allows us to measure the unbindingforces on the surface of live leukocytes.

As shown in Figs. 9.4A and 9.4B, theAFM force measurements were carried outwith an LFA-1 expressing T-cell hybridoma(i.e., 3A9) coupled to the AFM cantilever.Purified ICAM-1 was immobilized on a tis-sue culture dish. High-affinity state of LFA-1 was induced by the addition of 5 mMMg2+ and 1 mM ethylene glycol bis(β-aminoethylether)-N ,N ,N ′,N ′-tetraacetic acid(EGTA). Figure 9.4C plots the most prob-able unbinding force of individual LFA-1–ICAM-1 complexes formed between 3A9 andimmobilized ICAM-1 over three orders ofmagnitude change in loading rate. As shown,the LFA-1–ICAM-1 force spectrum can bedivided into two loading regimes. Induction ofhigh-affinity LFA-1 by Mg2+/EGTA resultedin stronger LFA-1–ICAM-1 unbinding forcesin the slow loading regime (<10,000 pN/s).However, the unbinding forces of restingand Mg2+ –EGTA-activated complexes werenearly identical in the fast loading regime(>10,000 pN/s).

An analysis of Fig. 9.4C revealed that theunbinding of the LFA-1–ICAM-1 complexinvolves overcoming at least two activationbarriers. Fitting Eq. (9.4) to the slow and fastregimes gives the Bell model parameters forthe outer and inner barriers of the complex,respectively. The γ values for the inner andouter barriers are approximately 0.2 and 2 A,respectively. Moreover, our analysis revealedthat the observed increase in unbinding

Page 168: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

SINGLE-MOLECULE UNBINDING: THEORY AND APPLICATIONS 163

TABLE 9.1. Summary of Reported AFM Unbinding Studies.

Ligand–Receptor PairLoading Rate

(pN/s)Rupture Forces

(pN)Bell Parameter

γ (A) References

Leukocyte AdhesionComplexesLFA-1–ICAM-1 50–50,000 20–320 0.2, 2 Zhang et al. (2002)VLA-4–VCAM-1 30–60,000 15–130 1, 5.5 Zhang et al. (2003)P-selectin–sLeX 70–100,000 20–220 0.8, 4.5 Zhang et al. (2004)E-selectin–sLeX 200–100,000 40–160 0.9, 5 Zhang et al. (2004)L-selectin–sLeX 300–100,000 20–140 0.8, 4.5 Zhang et al. (2004)P-selectin–PSGL-1 100–10,000 30–220 1.4 Hanley et al. (2003)P-selectin–PSGL-1 —a 110–170 2.5 Fritz et al. (1998)L-selectin–sLeXb 10–100,000 10–180 0.6, 4 Evans et al. (2001)L-selectin–PSGL-1b 10–100,000 20–160 0.6, 4 Evans et al. (2001)Other Ligand–ReceptorComplexesα5β1 –fibronectin 50–50,000 40–170 0.9, 4 Li et al. (2003)Streptavidin–biotin 0.05–60,000 5–170 1.2, 5 Merkel et al. (1999)Avidin–biotin 0.05–60,000 5–170 1.2, 3, 30 Merkel et al. (1999)Con A–D-mannose 400–5000 80–125 2.7 Chen and Moy (2000)Plant lectin–asialofetuin 100–30,000 37–65 4–6 Dettmann et al. (2003)GTPase Rap–impβc 300–80,000 40–90, 75–160d N/A Nevo et al. (2003)VE-cadherin pair —a 30–50 5.9 Baumgartner et al. (2000)Anti-γ-GT–γ-GTe N/A 131 ± 44 N/A Wielert-Badt et al. (2002)Anti-ferritin–ferritin N/A 49 ± 10 N/A Allen et al. (1997)Anti-HSA–HSAf N/A 240 ± 48 N/A Hinterdorfer et al. (1996)Anti-βhCG–βhCGg N/A 100 ± 47 N/A Allen et al. (1999)Glycoprotein csA pair N/A 23 ± 8 N/A Benoit et al. (2000)GroELh pair N/A 420 ± 100 N/A Vinckier et al. (1998)ICAM-1–anti-ICAM-1 N/A 100 ± 50 N/A Willemsen et al. (1998)Insulin pair N/A 1340–1350 N/A Yip et al. (1998)Meromyosin–actin N/A 15–25 N/A Nakajima et al. (1997)Ocular mucin pair N/A 100–4000 N/A Berry et al. (2001)Proteoglycan pair N/A 40 ± 15 N/A Dammer et al. (1995)α5β1 –GRGDSP peptide N/A 32 ± 2 N/A Lehenkari and Horton (1999)αvβ3 –echistatin N/A 97 ± 15 N/A Lehenkari and Horton (1999)αvβ3 –GRGDSP peptide N/A 42 ± 4 N/A Lehenkari and Horton (1999)αvβ3 –osteopontin N/A 50 ± 2 N/A Lehenkari and Horton (1999)

a Authors reported unbinding forces versus different pulling velocities; we were unable to convert the velocity toloading rates.bStudies using biomembrane force probe.c impβ: nuclear import receptor importin β1.d Authors reported two populations of unbinding forces, reflecting the existence of two conformation states in theRap–impβ complexes.eγ-GT: γ-glutamyl-transpeptidase.f HSA: Human serum albumin.gβhCG: β subunit of human chorionic gonadotrophin.h GroEL: Chaperonin GroEL from E. coli.

force following LFA-1 activation in the slowloading regime stemmed from an increase inthe outer activation energy barrier, which ismanifested in a lowering of the dissociationrate constant from 4.6/s to 0.15/s. The

inner activation barrier was unaffected bythe integrin activation, since no significantchange was found in the dynamic strengthof the complex in the fast loading regimefollowing LFA-1 activation.

Page 169: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

164 INTERMOLECULAR FORCES OF LEUKOCYTE ADHESION MOLECULES

Loading rate (pN/s)

Unb

indi

ng fo

rce

(pN

)

102 103 104 105

200

150

100

50

0

50 nm50 pN

fu

Piezo displacement

For

ce ks

A B

Figure 9.3. Dynamic force spectra of P-selectin–sLeX interactions. (A) A series of three consecutive AFM forcetraces of the P-selectin–sLeX interaction. Gray and black traces were recorded during the approach and withdrawalof the cantilever, respectively. In the top trace, the force fu is the unbinding force. The loading rate of the unbindingforce was derived from the system spring constant ks . To study individual bonds, proteins were immobilized in lowsurface densities; and the interaction between tip and substrate was further reduced by lowering contact duration.Under these conditions, some force scans showed no adhesion (middle and bottom traces). The specificity of theP-selectin–sLeX interactions was confirmed by a reduction in the frequency of adhesion following the addition of afunction blocking antibody against P-selectin, as well as by the low adhesion frequency measured between the sLeXfunctionalized cantilever and immobilized bovine serum albumin. (B) Dynamic force spectra of the P-selectin–sLeXinteractions. The best-fit curves (solid lines) were obtained using Eq. (9.4) applied for each of the two loadingregimes. Error bars are the standard error of the mean (SEM). Some error bars are smaller than the symbols.

RestingEGTA/Mg2+

Loading rate (pN/s)

300

200

100

0

102 103 104

Unb

indi

ng fo

rce

(pN

)

Loading rate (pN/s)

101 102 103 104

Unb

indi

ng fo

rce

(pN

) Resting

140

120

100

80

60

40

20

0

TS 2/16

DC

cell

cantilever

LFA-1

ICAM-1

3A9 Cell

AFM cantileverBA

Figure 9.4. Dynamic force spectra of integrin–ligand interactions on live leukocytes. (A) Schematic diagram of theAFM experiments using live 3A9 cells. (B) Micrograph of a live leukocyte attached to the AFM cantilever. The baris 20 µm. (C) DFS of the low- and high-affinity LFA-1–ICAM-1 complexes. (D) DFS of the low- and high-affinityVLA-4–VCAM-1 complexes on live U937 cells.

Page 170: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

SINGLE-MOLECULE UNBINDING: THEORY AND APPLICATIONS 165

(B) P-selectin−sLeX

(C) LFA-1−ICAM-1

(A) VLA-4−VCAM-1

250200150100500

Pulling force (pN)

104

Off-

rate

(1/

s)100

102

10−2

Figure 9.5. Kinetic profiles of the (A) VLA-4–VCAM-1, (B) P-selectin–sLeX, and (C) LFA-1–ICAM-1 complexes.

Similarly, the DFS of the VLA-4–VCAM-1 complex was measured using liveU937 cells (Fig. 9.4D). The high-affinitystate of VLA-4 was induced by the addi-tion of an activation antibody TS2/16. Afteractivation, the unbinding forces of the VLA-4–VCAM-1 complex were elevated overthe range of loading rates between 100and 20,000 pN/s, but did not change thedynamic response of the complex at loadingrates greater than 20,000 pN/s. The dissocia-tion rates of the outer barrier for low- andhigh-affinity VLA-4–VCAM-1 complexesare 1.4/s and 0.035/s, respectively. However,no significant difference was found in dis-sociation rates (∼80/s) for the inner bar-rier of both affinity states. Thus, the VLA-4–VCAM-1 system shows similarities tothe LFA-1–ICAM-1 system. In both inte-grin systems, induction of high-affinity stateselevated the energy potential of their outeractivation barriers, but had minimal effect onthe inner barriers.

For a molecular complex with two acti-vation barriers, the force-dependent dissoci-ation rate is given by

koff = 1/{k0−1

1 exp[−f γ1/kBT ]

+ k0−1

2 exp[−f γ2/kBT ]} (9.5)

where the superscripts 1 and 2 refer to theinner and outer activation energy barriers,respectively (Evans et al., 2001). Figure 9.5presents the kinetic profiles of theP-selectin–sLeX, high-affinity VLA-4–VCAM-1, and high-affinity LFA-1–ICAM-1interactions. The kinetic profiles revealed theprofound impact of a pulling force on the dis-sociation rate of these complexes. As shown,the dissociation rate constants are sensitiveto weak pulling forces, but are less respon-sive to stronger pulling forces (>∼50 pN forboth P-selectin and VLA-4; >∼100 pN forLFA-1). This resistance to stronger pullingforce can be explained by the Bell–Evansmodel. At weak pulling forces, the disso-ciation rates of these complexes are deter-mined by the properties of the outer barriers,which are relatively wide and, hence, moresensitive to the applied forces. However, atstronger pulling forces, the outer barriers aresuppressed, and the dissociation rates aregoverned by the properties of the inner bar-rier, which is steep and less responsive to apulling force.

It is interesting to compare the dynamicresponses of these three interactions andrelate them to their functions. Previous stud-ies revealed that the P-selectin–sLeX and

Page 171: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

166 INTERMOLECULAR FORCES OF LEUKOCYTE ADHESION MOLECULES

LFA-1–ICAM-1 interactions mediate leuko-cyte rolling and firm adhesion, respectively,and that the VLA-4–VCAM-1 interactioncan mediate both leukocyte rolling and firmadhesion (Kubes, 2002). A comparison ofkinetic profiles of these complexes revealedthat both the P-selectin–sLeX complex andthe VLA-4–VCAM-1 complexes are moresensitive to a pulling force than the LFA-1–ICAM-1 complex (Fig. 9.5). This findingsuggests that the P-selectin–sLeX and VLA-4–VCAM-1 interactions are more suited forcell rolling, because during cell rolling, theadhesive complexes need to dissociate read-ily by shear force (Orsello et al., 2001). Themore stable complex, the LFA-1–ICAM-1,is more suitable for mediating firm adhesion,in line with observations by Lawrence et al.(1991). In their flow chamber experiments,leukocytes rolled only on P-selectin but noton ICAM-1-coated surfaces.

9.3. SUMMARY

In this review, we have presented exampleson how to use the AFM to probe theintermolecular forces of leukocyte adhesionmolecules. AFM studies on three majorleukocyte adhesion complexes revealed thata common characteristic of these complexesis the presence of at least two activationbarriers. The inner barriers are crucial forthese complexes to resist strong pullingforces. In addition, experiments using liveleukocytes revealed that integrin activationenhances only the energy of their outeractivation barriers.

The methods described herein are notlimited to the chosen system, and has beenused to study a broad range of protein–ligandinteractions (see Table 9.1 for a partiallist). With the completion of the humangenome project, there is a compelling needto characterize the biophysical and functionalfeatures of a tremendous amount of protein-protein interactions. It is predictable thatin the near future, the AFM will play animportant role in such studies.

ACKNOWLEDGMENTS

We thank A. Chen, E. Wojcikiewicz, F. Li,and D. Bogorin for insightful discussionsand thank C. Freites for technical support.This work was sponsored by grants fromthe AHA, the NSF-BITC and the NIH(GM55611). XZ was a Predoctoral Fellow(0215139B) of the AHA.

REFERENCES

Allen, S., Chen, X., Davies, J., Davies, M. C.,Dawkes, A. C., Edwards, J. C., et al. (1997).Detection of antigen–antibody binding eventswith the atomic force microscope. Biochemistry36(24):7457–7463.

Allen, S., Davies, J., Davies, M. C., Dawkes,A. C., Roberts, C. J., Tendler, S. J., et al.(1999). The influence of epitope availabil-ity on atomic-force microscope studies ofantigen–antibody interactions. Biochem. J.341(Pt 1):173–178.

Baumgartner, W., Hinterdorfer, P., Ness, W.,Raab, A., Vestweber, D., Schindler, H., et al.(2000). Cadherin interaction probed by atomicforce microscopy. Proc. Nat. Acad. Sci. USA97(8):4005–4010.

Bell, G. I. (1978). Models for the specific adhe-sion of cells to cells. Science 200:618–627.

Benoit, M., Gabriel, D., Gerisch, G., and Gaub,H. E. (2000). Discrete interactions in celladhesion measured by single-molecule forcespectroscopy. Nature Cell Biol. 2(6):313–317.

Berry, M., McMaster, T. J., Corfield, A. P., andMiles, M. J. (2001). Exploring the molecularadhesion of ocular mucins. Biomacromolecules2(2):498–503.

Binnig, G., Quate, C. F., and Gerber, C. (1986).Atomic force microscope. Phys. Rev. Lett.56:930–933.

Chen, A., and Moy, V. T. (2000). Cross-linkingof cell surface receptors enhances cooper-ativity of molecular adhesion. Biophys. J.78(6):2814–2820.

Dammer, U., Popescu, O., Wagner, P., Ansel-metti, D., Guntherodt, H. J., and Misevic, G. N.(1995). Binding strength between cell adhe-sion proteoglycans measured by atomic forcemicroscopy. Science 267(5201):1173–1175.

Page 172: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 167

Dettmann, W., Grandbois, M., Andre, S., Be-noit, M., Wehle, A. K., Kaltner, H., et al.(2003). Differences in zero-force and force-driven kinetics of ligand dissociation from beta-galactoside-specific proteins (plant and animallectins, immunoglobulin G) monitored by plas-mon resonance and dynamic single moleculeforce microscopy. Arch. Biochem. Biophys.383(2):157–170.

Evans, E. (2001). Probing the relation betweenforce–lifetime–and chemistry in single molec-ular bonds. Annu. Rev. Biophys. Biomol. Struct.30:105–128.

Evans, E., and Ritchie, K. (1997). Dynamicstrength of molecular adhesion bonds. Biophys.J. 72(4):1541–1555.

Evans, E., Leung, A., Hammer, D., and Simon, S.(2001). Chemically distinct transition statesgovern rapid dissociation of single L-selectinbonds under force. Proc. Natl. Acad. Sci. USA98(7):3784–3789.

Florin, E. L., Moy, V. T., and Gaub, H. E. (1994).Adhesion forces between individual ligand–re-ceptor pairs. Science 264(5157):415–417.

Fritz, J., Katopodis, A. G., Kolbinger, F., andAnselmetti, D. (1998). Force-mediated kineticsof single P-selectin/ligand complexes observedby atomic force microscopy. Proc. Natl. Acad.Sci. USA 95(21):12283–12288.

Hanley, W., McCarty, O., Jadhav, S., Tseng, Y.,Wirtz, D., and Konstantopoulos, K. (2003).Single molecule characterization of P-selectin/ligand binding. J. Biol. Chem. 278(12):10556–10561.

Hinterdorfer, P., Baumgartner, W., Gruber, H. J.,Schilcher, K., and Schindler, H. (1996). Detec-tion and localization of individual antibody–an-tigen recognition events by atomic forcemicroscopy. Proc. Nat. Acad. Sci. USA 93(8):3477–3481.

Kubes, P. (2002). The complexities of leukocyterecruitment. Semin. Immunol. 14(2):65–72.

Kubes, P., and Kerfoot, S. M. (2001). Leuko-cyte recruitment in the microcirculation: therolling paradigm revisited. News Physiol. Sci.16:76–80.

Lawrence, M. B., and Springer, T. A. (1991).Leukocytes roll on a selectin at physio-logic flow rates: Distinction from and pre-requisite for adhesion through integrins. Cell65(5):859–873.

Lee, G. U., Chrisey, L. A., and Colton, R. J.(1994). Direct measurement of the forcesbetween complementary strands of DNA. Sci-ence 266(5186):771–773.

Lehenkari, P. P., and Horton, M. A. (1999). Sin-gle integrin molecule adhesion forces in intactcells measured by atomic force microscopy.Biochem. Biophys. Res. Commun. 259(3):645–650.

Li, F., Redick, S. D., Erickson, H. P., and Moy,V. T. (2003). Force measurements of thealpha(5)beta(1) integrin–fibronectin interaction.Biophys. J. 84(2):1252–1262.

Merkel, R. (2001). Force spectroscopy on sin-gle passive biomolecules and single biomolec-ular bonds. Phys. Rep. Rev. Sect. Phys. Lett.346(5):344–385.

Merkel, R., Nassoy, P., Leung, A., Ritchie, K.,and Evans, E. (1999). Energy landscapes ofreceptor–ligand bonds explored with dynamicforce spectroscopy. Nature 397(6714):50–53.

Nakajima, H., Kunioka, Y., Nakano, K., Shi-mizu, K., Seto, M., and Ando, T. (1997). Scan-ning force microscopy of the interaction eventsbetween a single molecule of heavy mero-myosin and actin. Biochem. Biophys. Res. Com-mun. 234(1):178–182.

Nevo, R., Stroh, C., Kienberger, F., Kaftan, D.,Brumfeld, V., Elbaum, M., et al. (2003). Amolecular switch between alternative confor-mational states in the complex of Ran andimportin beta1. Nature Struct. Biol. 10(7):553–557.

Orsello, C. E., Lauffenburger, D. A., and Ham-mer, D. A. (2001). Molecular properties in celladhesion: A physical and engineering perspec-tive. Trends Biotechnol. 19(8):310–316.

Springer, T. A. (1990). Adhesion receptors of theimmune system. Nature 346:425–434.

Springer, T. A. (1994). Traffic signals for lym-phocyte recirculation and leukocyte emigration:The multistep paradigm. Cell 76(2):301–314.

Vinckier, A., Gervasoni, P., Zaugg, F., Ziegler, U.,Lindner, P., Groscurth, P., et al. (1998). Atomicforce microscopy detects changes in the inter-action forces between GroEL and substrate pro-teins. Biophys. J. 74(6):3256–3263.

Wielert-Badt, S., Hinterdorfer, P., Gruber, H. J.,Lin, J. T., Badt, D., Wimmer, B., et al. (2002).Single molecule recognition of protein binding

Page 173: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

168 INTERMOLECULAR FORCES OF LEUKOCYTE ADHESION MOLECULES

epitopes in brush border membranes by forcemicroscopy. Biophys. J. 82(5):2767–2774.

Willemsen, O. H., Snel, M. M., van der Werf,K. O., de Grooth, B. G., Greve, J., Hinter-dorfer, P., et al. (1998). Simultaneous heightand adhesion imaging of antibody–antigeninteractions by atomic force microscopy. [com-ment]. Biophys. J. 75(5):2220–2228.

Yip, C. M., Yip, C. C., and Ward, M. D. (1998).Direct force measurements of insulin mono-mer–monomer interactions. Biochemistry37(16):5439–5449.

Zhang, X., Wojcikiewicz, E., and Moy, V. T.(2002). Force spectroscopy of the leuko-cyte function-associated antigen-1/intercellularadhesion molecule-1 interaction. Biophys. J.83(4):2270–2279.

Zhang, X., Craig, S. E., Humphries, M. J., andMoy, V. T. (2003). Unpublished data.

Zhang, X., Bogorin, D. F., and Moy, V. T. (2004).Molecular basis of the dynamic strength ofthe sialyl Lewis X–selectin interaction. Chem.Phys. Chem. 4:100–107.

Page 174: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

CHAPTER 10

MECHANISMS OF AVIDITY MODULATIONIN LEUKOCYTE ADHESION STUDIED BY AFMEWA P. WOJCIKIEWICZ AND VINCENT T. MOYDepartment of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami,Florida

10.1. INTRODUCTION

Integrins play an important role in the reg-ulation of leukocyte adhesion to target cells.Integrins are αβ heterodimeric glycoproteinsderived from the noncovalent associationof one of 18 α chains and one of 8 β

chains (Hynes, 1992). In leukocytes, αL com-bines with β2 to form leukocyte function-associated antigen-1 (LFA-1), the majorleukocyte integrin (Sanchez-Madrid et al.,1983). LFA-1 facilitates leukocyte adhesionby binding to its ligands, which include inter-cellular adhesion molecule-1 (ICAM-1), amember of the immunoglobulin superfam-ily, expressed on the target cell (Marlin andSpringer, 1983; Springer, 1990).

Integrins possess the ability to modulatethe adhesive states of cells, which is a crit-ical process necessary for proper immunefunction (Dustin and Springer, 1991). LFA-1is expressed in its inactive form on rest-ing lymphocytes and binds to ICAM-1 withlow affinity. In vivo, LFA-1 activation takesplace following the engagement of the Tlymphocyte by an antigen presenting cell(APC). This process results in the strength-ening of the transient interaction between thetwo cells that lasts long enough for the T

lymphocyte to become activated before thecells separate.

Both receptor affinity modulation andreceptor clustering (avidity modulation) con-tribute to the regulation of integrin-mediatedleukocyte adhesion (Lollo et al., 1993). Themechanism for affinity modulation involvesthe cytoplasmic engagement of the LFA-1 β

subunit. This “inside-out” signal, transmittedthough the β subunit, crosses over to the α

subunit via the β-propeller–β I-like domaininterface. The β propeller of the α subunitthen activates the I domain (Lu et al., 2001)by inducing a downward displacement of theI domain α7 helix and the subsequent open-ing of the ICAM-1 binding site. This result-ing conformational change quickly stabilizesthe LFA-1–ICAM-1 complex (Hughes et al.,1996; Shimaoka et al., 2001; Lupher et al.,2001). The need for an inside-out signalfor the induction of high-affinity LFA-1 canbe circumvented by high concentration ofextracellular Mg2+ and by certain antibod-ies directed against the β chain (Shimaokaet al., 2001; Lupher et al., 2001; McDowallet al., 1998).

The usage of the term “avidity modula-tion” most frequently refers to redistribu-tion or clustering of receptors that results

Force Microscopy: Applications in Biology and Medicine, edited by Bhanu P. Jena and J.K. Heinrich Horber.Copyright 2006 John Wiley & Sons, Inc.

169

Page 175: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

170 MECHANISMS OF AVIDITY MODULATION IN LEUKOCYTE ADHESION STUDIED BY AFM

in augmented cell adhesion. The mechanismof avidity modulation remains ill-definedand may include polarization of receptorsto the zone of cell–cell contact, clusteringof receptors to form focal adhesion sites,and dimerization of receptors. The dimer-ization of receptors may result in the for-mation of a dimeric complex that functionsas a cooperative unit that ruptures simulta-neously. Although the relative contributionsof these underlying mechanisms to adhe-sion still needs to be resolved, it is well-established that the initial event following theengagement of the antigen receptor of T lym-phocytes in the avidity modulation of leuko-cytes is the release of LFA-1 from cytoskele-tal constraints (Stewart et al., 1998). Recentstudies have shown the mobilization ofLFA-1 on T lymphocytes in contact withAPCs. Once released from the cytoskele-ton, the receptor molecules form ring-likestructures termed supramolecular activationclusters (SMACs). The SMACs consist ofantigen receptors localized at the center ofcell–cell contact and LFA-1–ICAM-1 com-plexes in the periphery (Monks et al., 1998).

Normally avidity modulation in the leuko-cyte is brought about as a result of T-cellreceptor signaling. Experimentally, changesin avidity can be induced by pharmaco-logical agents that exert their effects onintracellular pathways, bypassing the require-ment of surface receptor engagement (Berryand Nishizuka, 1990). One such agentthat promotes the mobilization of LFA-1and enhanced adhesion is phorbol myris-tate acetate (PMA) (Rothlein and Springer,1986). PMA is a potent activator of pro-tein kinase C (PKC). The activation ofPKC leads to an increase of intracellu-lar Ca2+ concentrations, Ins(1,4,5)P3- kinaseactivity, and phosphorylation of Rack1, Mac-MARCKS (macrophage-enriched myristoy-lated alanine-rich C kinase substrate), andL-plastin (van Kooyk and Figdor, 2000;Zhou and Li, 2000; Jones et al., 1992).Rack1 functions as a scaffold protein thatrecruits PKC to the site of action (van Kooyk

and Figdor, 2000). MacMARCKS and L-plastin are PKC substrates and are involvedin releasing LFA-1 from cytoskeletal con-straints. This takes place through the actionof the Ca2+-dependent protease, calpain,which is activated due to the increased lev-els of intracellular Ca2+ and releases LFA-1from the cytoskeleton (Stewart et al., 1998).PMA also acts indirectly on the activityof cytohesin-1, resulting in its associationwith LFA-1. Cytohesin-1 has been shown toinduce cytoskeletal reorganization and sub-sequently promote cell spreading (Kolanuset al., 1996).

Avidity changes can also be brought aboutexperimentally through the use of the Ca2+ionophore ionomycin and Ca2+ mobilizerssuch as thapsigargin, which increase intra-cellular Ca2+ more directly than PMA (Thas-trup et al., 1990; Thomas and Hanley, 1994).Thapsigargin functions by inhibiting theATPase pumps of Ca2+ storage organelles.These pumps normally pump Ca2+ fromthe cytosol into Ca2+ stores. When theyare inhibited, there is a resulting influx ofintracellular Ca2+ that not only comes fromthe internal Ca2+ stores but also occursas a result of capacitative fluxing throughthe plasma membrane (Stewart et al., 1998).Both Ca2+ mobilizers have been shown tofunction in a similar way as PMA. Clusteringhas been shown as a result of both ionomycinand thapsigargin application, and their mech-anism of action had been shown to dependon the calpain protease. The use of calpeptin,a calpain protease blocker, resulted in max-imal inhibition of T-cell adhesion (Tsuji-naka et al., 1988). No affinity modulationhad been reported following the use of Ca2+mobilizers using more traditional cell adhe-sion studies (Stewart et al., 1998).

Our previous work showed that cellspreading was part of the activation mech-anism of PMA (Wojcikiewicz et al., 2003).Using the AFM, we were able to quan-tify direct force measurements of PMA-stimulated 3A9 cells, which are murinehybridoma cells that express the LFA-1

Page 176: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

METHODS 171

integrin but no other receptor for ICAM-1(Lollo et al., 1993). LFA-1 of 3A9 cells isconstitutively inactive but can be activatedto a high avidity state by PMA. Our AFMdata revealed that PMA-stimulated cells didnot exhibit a strengthening of individualbonds between LFA-1/ICAM-1, but insteadexhibit an increase in the number of bondsthat were formed as well as an increasein the work of de-adhesion (Wojcikiewiczet al., 2003). This is thought to be largely aresult of cell spreading, which we were ableto visualize through reflection interferencecontrast microscopy (unpublished data). Thecells were found to become more compliantfollowing PMA stimulation as well, whichwas shown in our AFM compliance mea-surements. The combination of cell spread-ing and increased cell compliance appearsto be a multiplicative effect leading to a10-fold increase of leukocyte adhesion (Woj-cikiewicz et al., 2003)

The first part of this report details workthat was done in order to separate thecontribution of cell spreading from that ofLFA-1 redistribution or clustering towardenhanced leukocyte adhesion to ICAM-1.This was accomplished by restricting thelateral diffusion of LFA-1 by cross-linkingthe cell surface and then stimulating the cellsby PMA. Under these conditions, we are ableto investigate the role of cell spreading in celladhesion without contribution from receptorredistribution.

The second part of this report focuses onintegrin activation through a direct increaseof intracellular Ca2+. This was accomplishedby stimulating the 3A9 T cells with thap-sigargin and ionomycin. Thapsigargin actsby inhibiting the ATPase pumps on theCa2+ storage organelles and ionomycin isa Ca2+ ionophore. Both methods increasedleukocyte adhesion to ICAM-1 at levels thatwere comparable to the increases previouslyreported with PMA. However, unlike PMA,which only acts through avidity modulation,other mechanisms may be involved whenintracellular Ca2+ is increased directly.

10.2. METHODS

10.2.1. Cells and Reagents

The 3A9 cell line was maintained in con-tinuous culture in RPMI 1640 medium sup-plemented with 10% heat-inactivated fetalcalf serum (Irvine Scientific, Santa Ana,CA), penicillin (50 U/ml, Gibco BRL, GrandIsland, NY), and streptomycin (50 µg/ml,Gibco BRL) and were expanded on a 3-daycycle (Kuhlman et al., 1991).

ICAM-1/Fc chimera consisted of all fiveextracellular domains of murine ICAM-1(Met 1-Ala 16), and the Fc fragment ofhuman IgG1 and was purchased from R & DSystems, Inc. (Minneapolis, MN). The abilityof this protein to bind LFA-1 was confirmedusing ELISA and adhesion assays. Fromthese experiments, we were able to con-clude that the LFA-1 binding epitope D1 ofICAM-1 is available for binding. Antibodiesagainst LFA-1 (i.e., M17/4.2 and FD441.8)and against ICAM-1 (i.e., BE29G1) werepurified from culture supernatant by pro-tein G affinity chromatography (Sanchez-Madrid et al., 1983; Kuhlman et al., 1991).Stock solutions of PMA (10,000X) (Sigma,St. Louis, MO) were prepared at 1 mMin DMSO. Stimulation of cells was carriedout at 37◦C for 5 min. Alternatively, thap-sigargin (Calbiochem, La Jolla, CA) andionomycin, calcium salt, S. congobatus (Cal-biochem) were used for cell stimulation at5 µM and 0.7 µM, respectively (Stewartet al., 1998). Stimulation of cells was carriedout at 37◦C for 30 min.

10.2.2. Cross-Linking of 3A9 Cells

The 3A9 cells were exposed to either disuc-cinimidyl suberate (DSS) or bis(sulfosuccini-midyl)suberate (BS3), (Pierce, Rockford, IL).Both cross-linkers were prepared at 20 mM,DSS in DMSO and BS3 in water. The cross-linking reaction was prepared at room tem-perature in PBS. One-tenth of a milligram of3A9 cells was incubated for 30 min in a finalconcentration of 2 mM of either DSS or BS3.

Page 177: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

172 MECHANISMS OF AVIDITY MODULATION IN LEUKOCYTE ADHESION STUDIED BY AFM

The reaction mixture was quenched with 1 MTris, pH 7.5, added to a final concentrationof 10 mM for 15 min.

10.2.3. Protein Immobilization

Twenty-five microliters of ICAM-1/Fc at50 µg/ml in 0.1 M NaHCO3 (pH 8.6) wasadsorbed overnight at 4◦C on the center ofa 35-mm tissue culture dish (Falcon 353001,Becton Dickinson Labware, Franklin Lakes,NJ). Unbound ICAM-1/Fc was removed andbovine albumin (Sigma) at 100 µg/ml inPBS was used to block the exposed surfaceof the dish. A similar protocol was used toimmobilize the anti-LFA-1 antibodies (i.e.,FD441.8 and M17/4.2).

10.2.4. AFM Measurementsof Adhesive Forces

The AFM used in our laboratory wasa homemade modification of the standardAFM and was designed to be operated inthe force spectroscopy mode (Fig. 10.1A)(Benoit et al., 2000; Heinz and Hoh, 1999;Willemsen et al., 2000; Wojcikiewicz et al.,2004). 3A9 cells were attached to theAFM cantilever by concanavalin A (con A)-mediated linkages (Zhang et al., 2002). To

prepare the con A-functionalized cantilever,the cantilevers were soaked in acetone for5 min, UV irradiated for 30 min, and incu-bated in biotinamidocaproyl-labeled bovineserum albumin (biotin-BSA, 0.5 mg/ml in100 mM NaHCO3, pH 8.6; Sigma) overnightat 37◦C. The cantilevers were then rinsedthree times with phosphate buffered saline(PBS, 10 mM PO4

3−, 150 mM NaCl, pH7.3) and incubated in streptavidin (0.5 mg/mlin PBS; Pierce; Rockford, IL) for 10 minat room temperature. Following the removalof unbound streptavidin, the cantilevers wereincubated in biotinylated Con A (0.2 mg/mlin PBS; Sigma) and then rinsed with PBS.

To attach the 3A9 cell to the cantilever,the end of the Con A-functionalized can-tilever was positioned above the center ofa cell and carefully lowered onto the cellfor approximately 1 s. When attached, thecell is positioned right behind the AFM can-tilever tip. To obtain an estimate of thestrength of the cell-cantilever linkage, weallowed the attached cell to interact witha substrate coated with Con A for 1 min.Upon retraction of the cantilever, separationalways (N > 20) occurred between the celland the Con A-coated surface. The aver-age force needed to induce separation wasgreater than 2 nN. Thus, these measurements

A B

Figure 10.1. (A) Complete schematic diagram of the AFM. (B) Schematics of AFM force measurements. Theprincipal events of cell–substrate interaction during an AFM force versus displacement measurement are: approachof the 3A9 cell onto a surface coated with ICAM-1, contact between cell and substrate, retraction of the 3A9 cell,and separation of the cell from the substrate. Arrows indicate the direction of cantilever movement.

Page 178: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

RESULTS 173

revealed that the linkages supporting cellattachment to the cantilever is greater than2 nN and much larger than the detach-ment force required to separate the bound3A9 cell from immobilized ICAM-1 (Zhanget al., 2002).

A piezoelectric translator was used tolower the cantilever/cell onto the sample.During the acquisition of a force scan, thecantilever was bent (Fig. 10.1B), causing thebeam of a 3-mW diode laser (Oz Optics;emission 635 nm) that was focused on topof the cantilever to be deflected, which wasmeasured by a position-sensitive 2-segmentphotodiode detector. AFM cantilevers werepurchased from TM Microscopes (Sunny-vale, CA). The largest triangular cantilever(320 µm long and 22 µm wide) from a setof five on the cantilever chip was used in ourmeasurements. These cantilevers were cali-brated by analysis of their thermally inducedfluctuation to determine their spring con-stant (Hutter and Bechhoefer, 1993). Theexperimentally determined spring constantswere consistent with the nominal value of10 mN/m given by the manufacturer (Woj-cikiewicz et al., 2003).

10.2.5. AFM Measurements of CellElasticity

In this report, the AFM also served as amicroindenter that probes the mechanicalproperties of the cell. The bare AFM tip islowered onto the cell surface at a set rate,typically 2 µm/s, to obtain the cell elasticitymeasurements. After contact, the AFM tipexerts a force against the cell that is propor-tional to the deflection of the cantilever. Thedeflection of the cantilever was recorded as afunction of the piezoelectric translator posi-tion during the approach and withdrawal ofthe AFM tip. The force–indentation curvesof the cells were derived from these recordsusing the surface of the tissue culture dishto calibrate the deflection of the cantilever.Estimates of Young’s modulus were made onthe assumptions that the cell is an isotropic

elastic solid and the AFM tip is a rigidcone (Wu et al., 1998; Hoh and Schoe-nenberger, 1994; Radmacher et al., 1996).According to this model, initially proposedby Hertz, the force (F )–indentation (α) rela-tion is a function of Young’s modulus ofthe cell, K , and the angle formed by theindenter and the plane of the surface, θ,as follows:

F = K

2(1 − ν2)

4

π tan θα2 (10.1)

Young’s modulus was obtained by least-square analysis of the force–indentationcurve using routines in the Igor Pro (Wave-Metrics, Inc., Lake Oswego, OR) softwarepackage. The indenter angle, θ, and Poissonratio, ν, were assumed to be 55◦ and 0.5,respectively.

Measurements of cell adhesion and elas-ticity were carried out at 25◦C in fresh tissueculture medium supplemented with 10 mMHEPES buffer. Cells were stimulated by5 mM MgCl2 plus 1 mM EGTA or 100 nMPMA. The activation of 3A9 by Mg2+ wasimmediate. 3A9 cells were exposed to PMAfor ∼5 min at 37◦C prior to the start ofthe experiments. All experiments involvedmaking contact with the same cell up to 50times. There was no dependence on previ-ous contacts observed in either the elastic-ity or adhesion AFM studies (Wojcikiewiczet al., 2003).

10.3. RESULTS

10.3.1. Contribution of Integrin LateralRedistribution in Leukocyte Adhesion

We have carried out studies aimed atresolving the contribution of cell spreadingfrom LFA-1 redistribution on the cell surface(clustering). This was accomplished throughthe use of both an external and an internalcross-linking agent, which allowed us toexamine leukocyte adhesion to ICAM-1under conditions where the movement ofreceptors on the cell surface is retarded

Page 179: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

174 MECHANISMS OF AVIDITY MODULATION IN LEUKOCYTE ADHESION STUDIED BY AFM

and cell spreading is prevented. 3A9 Tcells were exposed to bis(sulfosuccinimidyl)suberate (BS3), a water-soluble, membrane-impermeable cross-linker that restricts thelateral diffusion of LFA-1 but should notcross-link the cytoskeleton. The BS3 cross-linked cells were then activated by PMA,which induced the cell to spread, whilethe lateral diffusion of membrane receptorsremained retarded. Under these conditions,we were able to investigate the role of cellspreading in cell adhesion while limiting thecontribution from receptor redistribution.

AFM force measurements were carriedout to determine the adhesive propertiesof the cross-linked cells. As illustrated inFig. 10.1B, a 3A9 cell, coupled to the endof an AFM cantilever, was lowered onto asubstrate coated with ICAM-1. The workof de-adhesion was obtained by integrat-ing the adhesive force over the distancetraveled by the cantilever upon its retrac-tion from the substrate. The AFM adhe-sion data revealed that both BS3 cross-linkedand untreated 3A9 cells had similar levelsof adhesion. On average, the work of de-adhesion to detach resting 3A9 cells fromimmobilized ICAM-1 was 3.16 × 10−16 Jfor untreated cells and 2.89 × 10−16 J forthe BS3 cross-linked (Fig. 10.2). This isto be expected, because clustering is notthought to occur until the cell is stimu-lated. Following PMA stimulation, both theuntreated and the BS3 cross-linked cellsshowed a significant increase in adhesion(Fig. 10.2). The work of de-adhesion ofthe BS3 cross-linked cells following PMAstimulation was ∼20% smaller on averagethan that of the untreated cells. In con-trast, 3A9 cells that were internally cross-linked with disuccinimidyl suberate (DSS)exhibited diminished adhesion to immobi-lized ICAM-1, even following PMA stimu-lation. These observations are attributed tothe cross-linked cell’s inability to spread orinduce LFA-1 clustering.

To establish that the cytoskeleton ofthe cell is not cross-linked by BS3, we

Figure 10.2. Work of de-adhesion of resting (whitebars) and PMA-stimulated (gray bars) 3A9 cells. Thecells compared were either not cross-linked or cross-linked with the water-soluble, membrane-impermeablecross-inker (BS3) or the lipid-soluble, membrane-permeable cross-linker (DSS). The calculated areas ofde-adhesion were acquired with a compression force of200 pN, 5-s contact, and a cantilever retraction speedof 2 µm/s. The error bar is the standard error.

measured the elasticity of the cross-linkedcells by AFM. The Young’s modulus val-ues for both the unstimulated and PMA-stimulated cells that have been cross-linkedwith BS3 are similar to those of cells thathave not undergone cross-linking. As shownin Fig. 10.3, the Young’s modulus valuesof the cross-linked cells were 1.6 ± 0.08kPa for unstimulated cells (1.4 ± 0.4 kPa,without cross-linking) and 0.3 ± 0.02 kPafor PMA-stimulated cells (0.3 ± 0.01 kPa,without cross-linking). Elasticity measure-ments conducted on the DSS cross-linkedcells showed Young’s modulus values of5.1 ± 0.3 kPa for the unstimulated cells and4.6 ± 0.5 kPa PMA for the PMA stim-ulated cells. These values are consider-ably higher than both the untreated andBS3 cross-linked cells and are indicative ofstiffer, less compliant cells that are also lesslikely to undergo spreading following PMA-stimulation (Fig. 10.3). The ability of bothgroups of cross-linked cells to spread wasfurther verified using reflection interferencecontrast microscopy, where spreading wasonly observed for the PMA-stimulated cells

Page 180: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

RESULTS 175

Figure 10.3. Young’s modulus of resting (white bars)and PMA-stimulated (gray bars) 3A9 cells. The cellscompared were either not cross-linked or cross-linkedwith the water-soluble, membrane-impermeable cross-linker (BS3) or the lipid-soluble, membrane-permeablecross-linker (DSS). The error bar is the standard error.

that had been cross-linked with BS3 (datanot shown).

10.3.2. Effects of Ionomycinand Thapsigargin Stimulationon Leukocyte Adhesion

The next series of experiments focusedon the effects of direct intracellular Ca2+increase on the adhesion of the 3A9 cellsto ICAM-1. We examined the effects of twoagents, ionomycin and thapsigargin, whichhave been shown to enhance leukocyte adhe-sion through increases in [Ca2+

i ] (Stewartet al., 1998). Figure 10.4 presents a seriesof AFM force measurements carried outwith resting and thapsigargin-stimulated 3A9cells. The adhesive interaction of the can-tilever bound cell and the ICAM-1-coatedsubstrate was determined by the work doneby the cantilever to stretch the cell and todetach it from immobilized ICAM-1. Thework of de-adhesion was derived by inte-grating the adhesive force over the distancetraveled by the cantilever. The work of de-adhesion, as can be seen in the second forcescan in Fig. 10.4, is much greater following

Figure 10.4. Force versus displacement traces ofthe interaction between 3A9 cells and immobilizedICAM-1. The measurements were carried out with aresting cell (first trace) and a thapsigargin-stimulatedcell (second trace). The measurements were acquiredwith a compression force of 200 pN, 5-s contact,and a cantilever retraction speed of 2 µm/s. The thirdtrace corresponds to a measurement acquired from athapsigargin-stimulated cell in the presence of an LFA-1(20 µg/ml FD441.8) function-blocking antibody. Theshaded area estimates the work of de-adhesion. Arrowspoint to breakage of LFA-1–ICAM-1 bond(s).

stimulation of the cell with thapsigargin.The work of de-adhesion increased from2.82 × 10−16 J in the resting state to 1.31 ×10−15 J following thapsigargin stimulation(Fig. 10.6). Treatment of the thapsigarginstimulated cells with an antibody againstLFA-1 (FD441.8) resulted in a work of de-adhesion that was greatly reduced (5.95 ×10−17 J), indicating the specificity of thisinteraction.

Stimulation of the 3A9 cells with 0.7 µMof ionomycin also resulted in a signifi-cant increase of the work of de-adhesion.Figure 10.5 shows a series of measurementsobtained with ionomycin-stimulated cells.The work of de-adhesion following ion-omycin stimulation is significantly largerthan that of the resting cell as can beseen in Fig. 10.5. The work of de-adhesion

Page 181: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

176 MECHANISMS OF AVIDITY MODULATION IN LEUKOCYTE ADHESION STUDIED BY AFM

Figure 10.5. Force versus displacement traces ofthe interaction between 3A9 cells and immobilizedICAM-1. The measurements were carried out with aresting cell (first trace) and an ionomycin-stimulatedcell (second trace). The measurements were acquiredwith a compression force of 200 pN, 5 s contactand a cantilever retraction speed of 2 µm/s. The thirdtrace corresponds to a measurement acquired from anionomycin-stimulated cell in the presence of an LFA-1(20 µg/ml FD441.8) function-blocking antibody. Theshaded area estimates the work of de-adhesion. Arrowspoint to breakage of LFA-1–ICAM-1 bond(s).

Figure 10.6. Work of de-adhesion of resting and stimu-lated 3A9 cells bound to immobilized ICAM-1/Fc. Cellswere stimulated with 5 µM thapsigargin or 0.7 µMionomycin. The inhibitory monoclonal antibody usedwas FD441.8 (anti-LFA-1; 20 µg/ml). The numbersgiven here were derived from measurements that wereacquired with a compression force of 200 pN, 5-s con-tact, and a cantilever retraction speed of 2 µm/s. Theerror bar is the standard error.

increased from 2.82 × 10−16 J in the restingstate to 1.94 × 10−15 J following ionomycinstimulation (Fig. 10.6). The ionomycin-sti-mulated cells that were treated with antibodyagainst LFA-1 had a greatly reduced area ofde-adhesion of 8.64 × 10−17 J (Fig 10.6).

10.4. DISCUSSION

The main goal of these studies was to elabo-rate on the contribution of integrin clusteringto the enhanced adhesion of 3A9 cells toICAM-1. As was reported earlier in Woj-cikiewicz et al. (2003), our force data didnot suggest the formation of large clus-ters of LFA-1 on the cell surface. Thiswould have been observed as irregularitiesin the adhesion force measurements follow-ing PMA-stimulation due to areas on thecell surface exhibiting high and low recep-tor densities (Wojcikiewicz et al., 2003). Itis still conceivable that microclustering istaking place; and although no cooperativ-ity of bonds was observed in this or thepreviously published data, clustering maystill aid in enhancing adhesion by distribut-ing forces among clustered LFA-1–ICAM-1 complexes.

Retarding the mobility of LFA-1 receptorson the cell surface through BS3 cross-linking resulted in a ∼20% decrease ofthe area of de-adhesion following PMAstimulation as compared to the untreatedcells (Fig. 10.2). Both RICM and elasticitystudies revealed that the cells were stillspreading following PMA stimulation. Weattribute the drop in the area of de-adhesionfollowing BS3 cross-linking to be due tothe absence of clustering. This indicatesthat the rearrangement of LFA-1 on thecell surface is an important part of PMA-stimulation of 3A9 T cells, but it is notthe major mechanism responsible for theobserved enhanced adhesion.

The 3A9 cells that were exposed tothe lipid-soluble cross-linker, DSS, exhib-ited low levels of adhesion. This cross-linker

Page 182: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

DISCUSSION 177

was expected to prevent receptor mobilityon the cell surface as well as any cytoskele-tal changes leading to cell spreading due toits internal cross-linking capacity. Eliminat-ing cell spreading in addition to clusteringresulted in a very significant drop in the levelof adhesion. Even following stimulation withPMA, these cells had lower areas of de-adhesion than the resting, untreated 3A9 Tcells (Fig. 10.2).

Our previous study highlighted the impor-tance of cell spreading as a key mechanismfor enhanced adhesion of T cells to ICAM-1 following PMA stimulation (Wojcikiewiczet al., 2003). The cross-linking studies rein-force the importance of cell spreading. Theacquired elasticity measurements can be usedto estimate the area of contact for the 3A9cells. The Young’s modulus of the unstim-ulated BS3 cross-linked cells was ∼1.6 kPa(Fig. 10.3). An estimate of contact area Ac

for a given compression force F , cell radiusR, and Young’s modulus K is given by theHertz model, that is, Ac = π × 3

√(RF/K)2

(Israelachvili, 1992). For F = 200 pN, R =5 µm, and K = 1600 Pa, the estimated con-tact area is ∼2.3 µm2. When K is reduced to0.3 kPa following PMA stimulation, the esti-mated area of contact is ∼7.0 µm2. Thesedata suggest that cell spreading can con-tribute to a 300% increase in cell adhe-sion for the BS3 cross-linked cells. Thearea of contact for the DSS cross-linkedcells changed little and remained smallerthan for resting cells that had not beencross-linked. The estimated contact area was∼1.06 µm2 for the DSS cross-linked cells(for K = 5100 Pa) and ∼1.14 µm2 for thePMA-stimulated cells (for K = 4600 Pa).These small contact areas for this groupof cells largely contributed to their lowadhesion.

Cell clustering has been examined inmany studies and multiple systems. It is animportant mechanism that has been impli-cated in adhesion enhancement. Our stud-ies focused on examining the contributionof this mechanism through the use of PMA

stimulation that has been shown to inducereceptor clustering. Based on this method-ology, we can conclude in these studiesthat cell compliance changes following PMAstimulation provide a major contribution tothe increase in the area of de-adhesion.Clustering of receptors on the cell surfaceappears to contribute to the adhesion pro-cess most likely through the re-distributionof force among the receptor micro-clustersand could be responsible for up to 20%of the enhanced adhesion effect that isobserved following PMA stimulation of 3A9cells.

The latter part of this study focusedon two other methods of cell stimula-tion, both of which involved increasingthe levels of [Ca2+

i ]. The first involvedstimulating the 3A9 T cells with 5 µMthapsigargin, which resulted in an ∼5-foldincrease in the work of de-adhesion. Stim-ulation of the cells with 0.7 µM ionomycinresulted in an ∼7-fold increase in the workof de-adhesion. These changes are simi-lar to those observed in our earlier stud-ies using PMA-stimulation, where an ∼5-fold increase in the work of de-adhesionwas observed following stimulation. In allthree cases, the enhancement in adhesionof the 3A9 cells to ICAM-1 was simi-lar in magnitude, with ionomycin stimu-lation having the greatest effect in theseexperiments.

The mechanism of PMA stimulation isbetter defined than the mechanisms of iono-mycin and thapsigargin stimulation. In addi-tion to the intracellular signaling changestaking place following PMA stimulation,the resultant Ca2+ fluxes following treat-ment of cells with ionomycin or thapsi-gargin have also been shown to activateactin-binding proteins, causing actin dis-sociation and cytoskeletal rearrangements(Stossel, 1989). Both compounds have beenshown to induce LFA-1 clustering on T cellsthrough a mechanism involving protease cal-pain (Stewart et al., 1998).

Page 183: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

178 MECHANISMS OF AVIDITY MODULATION IN LEUKOCYTE ADHESION STUDIED BY AFM

As in stimulation with PMA, both cellclustering and cell spreading most likely playa role in the observed enhancement of adhe-sion following stimulation with thapsigar-gin and ionomycin. However, other mech-anisms of enhanced adhesion may also beinvolved. Upon closer examination of theionomycin data, it becomes clear that theydiffer from the PMA data. The individualLFA-1–ICAM-1 ruptures are greater inmagnitude than those observed followingPMA stimulation and resemble those follow-ing treatment with Mg2+/EGTA (Fig. 10.5)(Wojcikiewicz et al., 2003; Zhang et al.,2002). This may be due to affinity modu-lation taking place alongside of cell spread-ing and receptor clustering. Affinity changeslead to the strengthening of the individ-ual LFA-1–ICAM-1 bonds. It is also pos-sible that the LFA-1–ICAM-1 bonds arebreaking in a cooperative manner—that is,simultaneously. The mechanism that allowsfor the enhancement of adhesion follow-ing thapsigargin stimulation may also bedifferent from what takes place followingPMA stimulation. The strength of the indi-vidual LFA-1–ICAM-1 rupture forces isnot greater than following PMA stimula-tion. However, we did observe large stepsduring the initial unbinding of the cellfrom ICAM-1, as can be seen in the sec-ond force scan of Fig. 10.4. This could bean indication of receptor cooperativity. Theobserved differences between PMA, thap-sigargin, and ionomycin stimulation mostlikely stem from the differences in the acti-vation process that results following theirapplication: PMA enhances adhesion throughthe activation of PKC, while both thapsi-gargin and ionomycin do so by increas-ing [Ca2+

i ] directly. There still remain manyunanswered questions regarding these mech-anisms of activation.

ACKNOWLEDGMENTS

This work was supported by grants from theAHA (Florida/Puerto Rico Affiliate), NSF-BITC and the NIH (GM55611).

REFERENCES

Benoit, M., Gabriel, D., Gerisch, G., and Gaub,H. E. (2000). Discrete interactions in celladhesion measured by single-molecule forcespectroscopy. Nature Cell Biol. 2:313–317.

Berry, N., and Nishizuka, Y. (1990). Proteinkinase C and T cell activation. Eur. J. Biochem.189:205–214.

Dustin, M. L., and Springer, T. A. (1991). Roleof lymphocyte adhesion receptors in transientinteractions and cell locomotion. Annu. Rev.Immunol. 9:27–66.

Heinz, W. F., and Hoh, J. H. (1999). Relativesurface charge density mapping with the atomicforce microscope. Biophys. J. 76:528–538.

Hoh, J. H., and Schoenenberger, C. A. (1994).Surface morphology and mechanical proper-ties of MDCK monolayers by atomic forcemicroscopy. J. Cell Sci. 107:1105–1114.

Hughes, P. E., Diaz-Gonzalez, F., Leong, L.,Wu, C., McDonald, J. A., Shattil, S. J., andGinsberg, M. H. (1996). Breaking the inte-grin hinge. A defined structural constraintregulates integrin signaling. J. Biol. Chem.271:6571–6574.

Hutter, J. L., and Bechhoefer, J. (1993). Calibra-tion of atomic-force microscope tips. Rev. Sci.Instrum. 64:1868–1873.

Hynes, R. O. (1992). Integrins: Versatility, mod-ulation, and signaling in cell adhesion. Cell69:11–25.

Israelachvili, J. N. (1992). Intermolecular andSurface Forces, Academic Press, London.

Jones, S. L., Wang, J., Turck, C. W., and Brown,E. J. (1992). A role for the actin-bundlingprotein L-plastin in the regulation of leukocyteintegrin function. Proc. Natl. Acad. Sci. USA95:9331–9336.

Kolanus, W., Nagel, W., Schiller, B., Zeitlmann,L., Godar, S., Stockinger, H., and Seed, B.

Page 184: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 179

(1996). Alpha L beta 2 integrin/LFA-1 bindingto ICAM-1 induced by cytohesin-1, a cytoplas-mic regulatory molecule. Cell 86:233–242.

Kuhlman, P., Moy, V. T., Lollo, B. A., and Brian,A. A. (1991). The accessory function of murineintercellular adhesion molecule-1 in T lympho-cyte activation. Contributions of adhesion andco-activation. J. Immunol. 146:1773–1782.

Lollo, B. A., Chan, K. W., Hanson, E. M., Moy,V. T., and Brian, A. A. (1993). Direct evidencefor two affinity states for lymphocyte function-associated antigen 1 on activated T cells. J.Biol. Chem. 268:21693–21700.

Lu, C., Shimaoka, M., Zang, Q., Takagi, J., andSpringer, T. A. (2001). Locking in alternateconformations of the integrin alpha Lbeta 2 Idomain with disulfide bonds reveals functionalrelationships among integrin domains. Proc.Natl. Acad. Sci. USA 98:2393–2398.

Lupher, M. L., Jr., Harris, E. A., Beals, C. R.,Sui, L. M., Liddington, R. C., and Staunton,D. E. (2001). Cellular activation of leukocytefunction-associated antigen-1 and its affinityare regulated at the I domain allosteric site.J. Immunol. 167:1431–1439.

Marlin, S. D., and Springer, T. A. (1987). Purifiedintercellular adhesion molecule-1 (ICAM-1) isa ligand for lymphocyte function-associatedantigen 1 (LFA-1). Cell 51:813–819.

McDowall, A., Leitinger, B., Stanley, P., Bates,P. A., Randi, A. M., and Hogg, N. (1998).The I domain of integrin leukocyte function-associated antigen-1 is involved in a confor-mational change leading to high affinity bind-ing to ligand intercellular adhesion molecule 1(ICAM-1). J. Biol. Chem. 273:27396–27403.

Monks, C. R., Freiberg, B. A., Kupfer, H.,Sciaky, N., and Kupfer, A. (1998). Three-dimensional segregation of supramolecular acti-vation clusters in T cells. Nature 395:82–86.

Radmacher, M., Fritz, M., Kacher, C. M., Cleve-land, J. P., and Hansma, P. K. (1996). Mea-suring the viscoelastic properties of humanplatelets with the atomic force microscope. Bio-phys. J. 70:556–567.

Rothlein, R., and Springer, T. A. (1986). Therequirement for lymphocyte function-associatedantigen 1 in homotypic leukocyte adhesion

stimulated by phorbol ester. J. Exp. Med.163:1132–1149.

Sanchez-Madrid, F., Simon, P., Thompson, S.,and Springer, T. A. (1983). Mapping of anti-genic and functional epitopes on the alpha- andbeta-subunits of two related mouse glycopro-teins involved in cell interactions, LFA-1 andMac-1. J. Exp. Med. 158:586–602.

Shimaoka, M., Lu, C., Palframan, R. T., vonAndrian, U. H., McCormack, A., Takagi, J.,and Springer, T. A. (2001). Reversibly lockinga protein fold in an active conformation with adisulfide bond: Integrin alpha L I domains withhigh affinity and antagonist activity in vivo.Proc. Natl. Acad. Sci. USA 98:6009–6014.

Stewart, M. P., McDowall, A., and Hogg, N.(1998). LFA-1-mediated adhesion is regulatedby cytoskeletal restraint and by a Ca2+-dependent protease, calpain. J. Cell Biol.140:699–707.

Springer, T. A. (1990). Adhesion receptors of theimmune system. Nature 346:425–434.

Stossel, T. P. (1989). From signal to pseudopod.How cells control cytoplasmic actin assembly.J. Biol. Chem. 264:18261–18264.

Thastrup, O., Cullen, P. J., Drobak, B. K., Han-ley, M. R., and Dawson, A. P. (1990). Thap-sigargin, a tumor promoter, discharges intra-cellular Ca2+ stores by specific inhibition ofthe endoplasmic reticulum Ca2+-ATPase. Proc.Natl. Acad. Sci. USA 87(7):2466–2470.

Thomas, D., and Hanley, M. R. (1994). Pharma-cological tools for perturbing intracellular cal-cium storage. Methods Cell Biol. 40:65–89.

Tsujinaka, T., Kajiwara, Y., Kambayashi, J.,Sakon, M., Higuchi, N., Tanaka, T., and Mori, T.(1988). Synthesis of a new cell penetrating cal-pain inhibitor (calpeptin). Biochem. Biophys.Res. Commun. 153(3):1201–1208.

van Kooyk, Y., and Figdor, C. G. (2000). Avidityregulation of integrins: The driving force inleukocyte adhesion. Curr. Opin. Cell Biol.12:542–547.

Willemsen, O. H., Snel, M. M., Cambi, A.,Greve, J., De Grooth, B. G., and Figdor, C. G.(2000). Biomolecular interactions measuredby atomic force microscopy. Biophys. J. 79:3267–3281.

Page 185: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

180 MECHANISMS OF AVIDITY MODULATION IN LEUKOCYTE ADHESION STUDIED BY AFM

Wojcikiewicz, E. P., Zhang, X., Chen, A., andMoy, V. T. (2003). Contributions of molecularbinding events and cellular compliance to themodulation of leukocyte adhesion. J. Cell Sci.116:2531–2539.

Wojcikiewicz, E. P., Zhang, X., and Moy, V. T.(2004). Force and compliance measurementson living cells using atomic force microscopy(AFM). Biol. Proc. Online 6:1–9.

Wu, H. W., Kuhn, T., and Moy, V. T. (1998).Mechanical properties of L929 cells mea-sured by atomic force microscopy: Effects of

anticytoskeletal drugs and membrane crosslink-ing. Scanning 20:389–397.

Zhang, X., Wojcikiewicz, E., and Moy, V. T.(2002). Force spectroscopy of the leukocytefunction-associated antigen-1/intercellular ad-hesion molecule-1 interaction. Biophys. J. 83:2270–2279.

Zhou, X., and Li, J. (2000). Macrophage-enrichedmyristoylated alanine-rich C kinase substrateand its phosphorylation is required for the phor-bol ester-stimulated diffusion of beta 2 integrinmolecules. J. Biol. Chem. 275:20217–20222.

Page 186: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

CHAPTER 11

RESOLVING THE THICKNESS ANDMICROMECHANICAL PROPERTIES OF LIPIDBILAYERS AND VESICLES USING AFMGUANGZHAO MAO AND XUEMEI LIANGDepartment of Chemical Engineering and Materials Science, Wayne State University, 5050Anthony Wayne Drive, Detroit, Michigan

11.1. INTRODUCTION

Lipid bilayers and vesicles serve as modelsfor biomembranes and cells (Blumenthalet al., 2003) and show increasing appli-cations in medical and nonmedical fields(Barenholz, 2001; Lasic, 1998). A number ofmethods have been established to make two-dimensional (2-D) lipid bilayers (Cooper,2004): (1) supported lipid bilayers by vesi-cle fusion or Langmuir–Blodgett (LB) dipcoating, (2) tethered bilayer membranes byhydrophilic linkers or biotinylated recep-tors, (3) polymer-supported lipid layers, (4)micro-arrayed lipid layers, and (5) blacklipid membranes spanning the holes ofporous supports. Small unilamellar vesiclescan be prepared from multilamellar vesi-cle dispersions by either sonication (Huang,1969) or extrusion (Hope et al., 1985). Thestudy of structure, morphology, and stabil-ity of lipid bilayers and vesicles is importantto the understanding of membrane fusion aswell as in drug delivery, gene therapy, andbiosensor design.

AFM images a surface by scanning asharp tip attached to a cantilever at a close

distance to the surface (Quate, 1994). AFMis one of the newest and most importanttools for biomembrane analysis because itprovides unrivaled molecular-level under-standing of structure, stability, and layerinteractions. AFM provides surface topo-graphical images with spatial resolution closeto 1 A and force–distance curves with detec-tion limit close to 10−12 N. AFM has becomethe preferred method for imaging soft mate-rials such as molecular crystals, proteins, andliving cells (Radmacher et al., 1992). Theideal way to image lipid layers and biomem-branes is to conduct scanning in the natu-ral solution environment of the constituents.Minimizing image force to below a thresh-old breakthrough force further ensures min-imum disturbance of the surface structure.Force measurement has become an indis-pensable part of AFM analysis of biomem-branes and biomaterials. AFM force curvesprovide information on the structure and sur-face charges, stability, and surface interac-tions of lipid layers in addition to the bilayerthickness. AFM imaging and force measure-ment are combined to provide a microme-chanical map of the biomembrane or cell

Force Microscopy: Applications in Biology and Medicine, edited by Bhanu P. Jena and J.K. Heinrich Horber.Copyright 2006 John Wiley & Sons, Inc.

181

Page 187: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

182 RESOLVING THE PROPERTIES OF LIPID BILAYERS AND VESICLES

surface in force mapping or force–volumeimaging (A-Hassan et al., 1998; Heinz andHoh, 1999).

This chapter describes various experimen-tal methods used to determine the structureof lipid layers, specifically the bilayer thick-ness and elastic constants using EggPC as anexample.

11.2. SUPPORTED LIPID BILAYER(SLB) THICKNESS

The structure and stability of SLBs withor without biological or synthetic additiveshave been studied extensively by AFM(Dufrene and Lee, 2000). The ideal way toimage fluid-like lipid bilayers with the leastdisturbance is the soft contact or tappingimaging mode, which uses the standardsilicon nitride (Si3N4) tips in the naturalsolution environment. After a solid substrate,such as mica, graphite, glass, silicon waferwith or without surface modification, isbrought to a close distance to the AFM tip,the lipid vesicle solution is injected directlyinto an AFM liquid cell through inlet tubing.The liquid cell is often sealed by an O-ring.Sometimes an open-cell configuration ispreferred. SLBs are often formed in situ asa result of vesicle rupture and fusion after

sufficient incubation time. Excess vesiclescan be removed by flushing the liquidcell with buffer solutions. A different pHor salt solution can be introduced duringmeasurement by solution exchange throughthe inlet/outlet tubing.

Bilayer thickness can be determined bythe step height at the bilayer domain edges.An example of bilayer thickness determina-tion is given in Fig. 11.1 of egg yolk phos-phatidylcholine or EggPC (Mao et al., 2004;Liang et al., 2004a). Multilamellar EggPCvesicle solution is extruded through a poly-carbonate membrane with an average poresize of 200 nm using a LiposoFast extruderfrom Avestin. The extrusion method pro-duces larger vesicles that rupture into bilay-ers upon adsorption to produce the EggPCSLB. AFM imaging and force measurementare conducted using a Nanoscope IIIa AFM(Digital Instruments) and an E scanner (max-imum scan area = 14.2 × 14.2 µm2). AFMsectional height analysis of the SLB stepedges in water yields a value of 6.3 ± 0.6 nm.The bilayer film thickness agrees with thehydrated bilayer thickness measured by x-raydiffraction (McIntosh et al., 1987). The valueis larger than the headgroup-to-headgroupbilayer distance (∼4 nm) due to hydration ofthe headgroup. Sackmann (1996) points outthat freely supported lipid–protein bilayers

10.0

−10.0

0

0 0.25

nm

0.50 0.75 µm

Figure 11.1. The AFM amplitude image and sectional height analysis of the corresponding height image of EggPCSLB patches on mica in water. The thickness of the EggPC bilayer patches is 6.27 ± 0.57 nm, which agrees with6.58 ± 0.37 nm from force-curve analysis.

Page 188: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

SUPPORTED LIPID BILAYER (SLB) THICKNESS 183

are separated from the substrate by a 1-nm-thick water layer. 1H-NMR estimates that theaverage thickness of the water layer betweenthe single bilayer and the glass bead surfaceis 1.7 ± 0.5 nm (Bayerl and Bloom, 1990).

In addition to three-dimensional (3-D)topographical imaging, AFM enables directforce measurements between the tip andsurface by moving an AFM tip up anddown at one point on the sample surface.The force–distance curves, or short forcecurves, yield not only bilayer thickness butalso its micromechanical properties. It hasbeen shown that film thickness from forcecurves matches precisely the value fromsectional height analysis of bilayer stepedges if similar image force is used inboth cases. AFM force calibration plots areconverted to force curves by defining thepoint of zero force and the point of zeroseparation (Prater et al., 1995). Zero forceis determined by identifying the region ata large separation, where the deflection isconstant. Zero separation is determined fromthe constant compliance region at high forcewhere deflection is linear with the expansionof the piezoelectric crystal or by the end of

the jump-in process. Discontinuity, called thejump-in point, has been a typical feature inforce curves measured on lipid bilayers andadsorbed surfactant films above a thresholdforce (Patrick et al., 1997; Dong and Mao,2000; Dufrene et al., 1998; Loi et al., 2002;Franz et al., 2002). This threshold forcehas been used as the upper limit of theimage force in the soft-contact AFM imagingmode most useful for organic, polymeric, andbiological samples (Manne et al., 1994). Thejump-in process during tip approach is due tospring instability. The process corresponds tothe rupture and removal of the bilayer portionfrom the tip/substrate gap, most probably bya lateral push-out mechanism. The thresholdforce (or maximum steric barrier) is reportedto be proportional to the surface excess ofthe film, and it can be used to comparepacking density within similar adsorptionclass (Eskilsson et al., 1999).

Figure 11.2 is a representative force curvemeasured on the EggPC SLB. Only forcevalues obtained with the same AFM tip arecompared. The radius of the contact tip (=33.2 ± 6.6 nm) is calibrated by imaging theTGT01 gratings (Mikro-Masch) (Villarrubia,

2.5

For

ce (

nN)

Distance (nm)

0−5 3020 2510 1550

0.5

1

1.5

2

Jump-in

Figure 11.2. Approaching force curve on EggPC SLB on mica surface in water. It shows that a repulsive forcestarts at 6.58 ± 0.37 nm and the jump-in distance near 4.57 ± 0.27 nm on the EggPC bilayer patch.

Page 189: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

184 RESOLVING THE PROPERTIES OF LIPID BILAYERS AND VESICLES

1997). The spring constant of the cantileveris calibrated using the deflection methodagainst a reference cantilever (Park Scien-tific Instruments) of known spring constant(0.157 N/m) (Tortonese, 1997). The cali-brated spring constant (0.17 ± 0.05 N/m) isused. Force curves are obtained in liquid con-tact mode only. Multiple force curves areobtained on the same SLB. In Fig. 11.2,the repulsion starts at 6.6 ± 0.4 nm andthe jump-in occurs at 4.6 ± 0.3 nm with amaximum force = 1.6 ± 0.3 nN. The repul-sion up to the jump-in point can bedescribed as a combination of hydration,steric force, and mechanical deformationfor bilayer films. The force curves mea-sured on the lower, flat background in bothextruded and sonicated vesicle adsorptioncases show characteristics of tip–mica inter-actions in water. It can be concluded thatmica is not fully covered by the SLB inthis case.

Next let’s look at the SLB film thicknessvariation due to the incorporation of amacromolecular additive as studied by AFM

force measurement (Liang et al., 2004b).Four Pluronic copolymers (L81, L121, P85,F87) (BASF) are used to prepare Pluronics-modified EggPC SLBs. The Pluronics areincorporated into EggPC vesicles accordingto literature procedures (Kostarelos et al.,1995, 1999). The concentration of Pluronicsis kept at 0.02% w/w well below thecritical micelle concentration (CMC). Thenominal structures and the number of EO(hydrophilic) and PO (hydrophobic) unitsare listed in Table 11.1. Pluronic copolymersare known to enhance the stability of lipidvesicles (Woodle et al., 1995; Kostareloset al., 1998; Johnsson et al., 1999).

TABLE 11.1. Molecular Structure and Propertiesof Pluronic Copolymers Provided by BASF.

Pluronic CompositionMolecular

Weight

L81 (PEO)2(PPO)40(PEO)2 ∼2666L121 (PEO)4(PPO)60(PEO)4 ∼4000P85 (PEO)26(PPO)39.5(PEO)26 4600F87 (PEO)61.1(PPO)39.7(PEO)61.1 7700

7

For

ce (

nN)

−1−2 3 8

Distance (nm)13

Pure EggPC

EggPC/L81

EggPC/L121

EggPC/P85

EggPC/F87

18

1

3

5

Figure 11.3. Force curves on EggPC, EggPC/L81, EggPC/L121, EggPC/P85, and EggPC/F87 bilayer. The forcecurves on the bilayer systems are characterized by one repulsive regime and one jump-in point.

Page 190: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

SUPPORTED LIPID BILAYER (SLB) THICKNESS 185

EggPC bilayer

0

18

16

14

12

10

8

6

4

2

EggPC/L81 EggPC/L121

Bilayer system

Onset point Jump-in point

Dis

tanc

e (n

m)

EggPC/P85 EggPC/F87

Figure 11.4. Comparison of onset point (square) and jump-in point (circle) distances for EggPC, EggPC/L81,EggPC/L121, EggPC/P85, and EggPC/F87 bilayer films. The error bar is calculated based on the standard deviationof jump-in distance and onset point from a number of force curves.

Fused bilayers are observed for Pluronicswith short PEO chain length. Fig. 11.3compares the force curve of EggPCSLB with those treated with Pluronics(EggPC/L81, EggPC/L121, EggPC/P85, andEggPC/F87). The approaching force curvesshow typical features of SLBs including thesteric repulsion up to a threshold followedby the jump-in to contact. The distancesof the repulsive force onset and jump-inpoints are plotted in Fig. 11.4. The forceonset distance corresponds to an unperturbedbilayer thickness at a small contact force(∼0.2 nN). Fig. 11.4 shows that the onsetdistance increases with increasing PEOchain length, which suggests that Pluroniccopolymers have been integrated into theSLB with PEO chain protruding into thesolution. Similar chain-length dependenceof adsorbed Pluronic copolymer layers ona self-assembled monolayer (SAM) of n-octadecyltrichlorosilane has been found byWang et al. (2002). A likely structure ofthe Pluronics-modified SLB is drawn in

Fig. 11.5. On the other hand, the jump-indistances remain unaffected by the PEOchain length. This observation is consistentwith the interpretation of the force gap as thetip jumping across the interior of the bilayer.

The PEO chains sticking out of theEggPC lipid bilayer can be either randomcoils or extended chains. Fig. 11.6 comparesour experimental data of the force onsetdistances with calculations based on randomcoil and fully extended chain configurations,respectively. The following equations areused. Fully extended chain length is Lf =a × N . Random coil chain length is Lr =a × N3/5, where a is the EO monomer chainlength (0.35 nm) and N is the number ofEO units. Pluronics-modified EggPC bilayerthickness is L

bilayerf = L0 + 2Lf (nm) if the

chain is fully extended and Lbilayerr = L0 +

2Lr (nm) if the chain is a random coil. L0

is the undisturbed EggPC bilayer thickness.Fig. 11.6 shows that experimental data arein good agreements with calculated valuesbased on the random coil chain.

Page 191: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

186 RESOLVING THE PROPERTIES OF LIPID BILAYERS AND VESICLES

Undisturbed bilayer thickness(Onset point of repulsive force)

PEO chain

PPO chain

bilayer thickness(Jump-in point)

Figure 11.5. (Top) Scheme of onset point of repulsive force on bilayer (undisturbed bilayer thickness). (Bottom)Scheme of jump-in point (bilayer thickness). The onset point of the repulsive force increased with increasing PEOchain length and the jump-in point is around 4–5 nm.

0

10

20

30

40

50

60

EggPC EggPC/L81(2) EggPC/L121(4) EggPC/P85(26) EggPC/F87(61)

Ons

et p

oint

(nm

)

Extended brush (theoretical value)

Random Coil (theoretical value)

Experimental value

Figure 11.6. Comparison of Pluronics-modified SLB thickness based on AFM force curves and theoreticalcalculations. The number in the parentheses in x axis is the PEO chain length.

Page 192: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

SONICATED UNILAMELLAR VESICLE (SUV) BILAYER THICKNESS AND MORPHOLOGY 187

11.3. SONICATED UNILAMELLARVESICLE (SUV) BILAYER THICKNESSAND MORPHOLOGY

Compared to other micromechanical meth-ods such as shape fluctuation method(Servuss et al., 1976; Schneider et al.,1984a, b), magnetic-field-induced orienta-tion (Sakurai and Kawamura, 1983), andmicropipette aspiration method (Evans andRawicz, 1990; Waugh et al., 1992), AFMcan provide information on (1) bilayer thick-ness, (2) adsorbed liposomes of small sizesbetween 20 and 100 nm, (3) mechanicalproperty variation at nanoscale, (4) localdeformation and rupture events induced bythe tip, and (5) surface and adhesive forces.AFM measurement focuses on the proper-ties of vesicle population with the small-est sizes because large vesicles rupture intobilayers upon adsorption. Small vesicleswith size between 50 and 150 nm havebeen found to passively target several dif-ferent tumors because this size range isa compromise between loading efficiency(which increases with increasing size), sta-bility (which decreases with increasing size),and ability to extravasate in tissues withenhanced permeability (which decreases withincreasing size) (Lasic, 1998). Size controlmay be a simple way for targeted drug deliv-ery because vesicle distribution among dif-ferent organs and tissues is related to its size.

Compared to studies on SLBs, muchfewer articles address the structure ofadsorbed yet unruptured vesicles. Shapeinstability, size, and softness of vesiclesoften prevent unambiguous AFM imaging.Perhaps the earliest images of intact lipo-somes are obtained by Shibata-Seki et al.(1996) from dipalmitoyl phosphatidylcholine(DPPC) and cholesterol mixture. Others sub-sequently confirm the characteristic featuresof adsorbed vesicles as first reported. Thefeatures include (1) flattened spheres withlength to width ratio less than 1 (about 0.4 intheir case) and (2) image quality dependenceon contact pressure either by changing imageforce or using tips of different curvature.

Egawa and Furusawa (1999) reporteda conical relief image of sonicated phos-phatidylethanolamine (PE) vesicles withdiameter around 100 nm adsorbed on itsown bilayer. The conical image has beenattributed to unruptured yet flattened vesiclewith height to diameter ratio of 0.175. Thom-son et al. (2000) imaged liposomes 70 nmin diameter made from p-ethyldimyristoylphosphatidylcholine (EDMPC) and choles-terol mixture on aminopropylsilane modifiedmica. Closely packed liposomes have beenimaged but can be easily disturbed by thescanning process. The article by Raviakineand Brisson (2000) provides many detailsabout vesicle adsorption and fusion by AFMimaging. In the article, supported vesicularlayer (SVL) is used to differentiate adsorbedand intact vesicles from adsorbed and single-bilayer disks as in SLBs. It is found thatvesicles of all sizes adsorb on mica, butonly vesicles with sizes below a critical rup-ture radius remain intact. The bilayer disksexhibit constant height of 5 nm, while theintact vesicles exhibit height variation from10 to 40 nm. The shape of liposomes arefound to change both with applied imageforce and adhesion between biotin and strep-tavidin that are incorporated into vesicleand substrate layer, respectively (Pignataroet al., 2000). Kumar and Hoh (2000) reportedintact vesicles of phospholipid and choles-terol mixture that exhibited saucerlike struc-ture in addition to the usual rounded pro-trusion (domelike structure). The saucerlikestructure is attributed to wetting and fusionnear the edge of the vesicle. A force curveis obtained on the intact vesicle that showsmonotonic repulsion with onset at 15 nm.The flattening from the outer edges towardthe center has been studied as a function oftime by Jass et al. (2000) and is described asthe first step in a multistep processes leadingtoward the formation of SLBs.

Here we describe an AFM study ofEggPC SUVs with diameter less than 50 nm,adsorbed on mica (Mao et al., 2004; Lianget al., 2004). AFM tip is moved on top of

Page 193: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

188 RESOLVING THE PROPERTIES OF LIPID BILAYERS AND VESICLES

individual vesicles so that mechanical prop-erties of the smallest vesicles can be mea-sured at the nanoscale. The EggPC vesi-cle undergoes reversible shape changes fromconvex to flattened and concave shape withincreasing image force. In addition to themonotonic repulsion due to vesicle resistanceto the AFM tip advancement and compres-sion, there exist several characteristic breaksin the force versus distance curves. Hertziananalysis of the slope of the repulsion gives ameasure of the vesicle elastic properties. Thebreaks in the force curve are interpreted asthe tip jumping across the bilayer and allowthe determination of bilayer thickness.

A well-established recipe is used to pre-pare EggPC SUVs (Huang, 1969). Multil-amellar vesicle (MLV) solution is obtainedby dissolving appropriate amounts of EggPClipids in chloroform/methanol (2:1 v/v) andevaporating the solvent with nitrogen. Afterdrying in a desiccator connected to a rotaryvacuum pump for 30 min, the lipids areresuspended by stirring them in an aqueousbuffer solution (20 mM NaCl) at a concen-tration of 0.5 mg/ml. SUVs are producedfrom the MLV suspension by sonication

to clarity (about 1 h) in a sonicator bath(Branson 2200). The suspension is kept inan ice bath during the sonication process.Sonicated samples are centrifuged for 1 h at16,000 rpm to remove large lipid fragmentsby Sorvall OTD70B Ultraspeed Centrifuge.

Images of EggPC vesicles are obtainedin both liquid contact mode and liquid tap-ping mode. Fig. 11.7 shows the amplitudeand deflection images for tapping and con-tact, respectively, and the cross-sectionalheight profiles from the corresponding heightimages. The images consist of sphericalobjects on a flat background. The lateralwidth or diameter of the spheres is measuredto be 48.6 ± 11.4 nm and 69.3 ± 12.8 nm onthe sectional height profiles of tapping andcontact, respectively. The diameter is takenfrom the width of the peak at the baselinein sectional height profiles. The lateral widthvalues are higher than the vesicle diame-ter in solution 37.0 ± 7.9 nm by dynamiclight scattering. The discrepancy is gener-ally attributed to the flattening of vesicleson surface and tip casting its shadow overthe object, so-called tip convolution effect.The height of vesicles from sectional height

∼13-

15 n

m

A B

∼4 n

m

Figure 11.7. AFM images and cross-section profiles of EggPC vesicles on mica by tapping mode (A) and contactmode (B). The image size is 1 × 1 µm2. The height profile belongs to the particle pointed by the arrow obtained inheight image. (A) Amplitude image with Z scale = 20 nm. (B) Deflection image with Z scale = 5 nm.

Page 194: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

SONICATED UNILAMELLAR VESICLE (SUV) BILAYER THICKNESS AND MORPHOLOGY 189

profiles in Tapping and Contact Mode isdetermined to be 13.9 ± 2.2 nm and 3.9 ±0.4 nm, respectively. The height value fromtapping mode is about 40% to 50% of thevesicle diameter in solution. Causes for theunreasonably low height values from con-tact mode include vesicle movement duringcontact mode imaging and the vesicles beingseverely compressed by the tip. The inter-mittent contact motion of the tapping tipis known to reduce frictional and adhesiveforces on the sample surface. By minimizingimage force, it is possible to obtain stableimages of intact vesicles in either tappingor contact mode. Tapping causes less defor-mation of the vesicle, and it can maintain asmaller image force than contact mode.

With increasing image force, EggPC vesi-cles exhibit three distinctive morphologies:convex-shaped vesicles where the high-est point is at the center as shown inFig. 11.8A, disk-shaped vesicles with uni-form height across much of the vesicle asshown in Fig. 11.8B, and concave-shapedvesicles where the center is depressed asshown in Fig. 11.8C. The lateral diameterof the vesicle increases from 48.6 ± 11.4 nmfor convex shape to 73.7 ± 11.5 nm fordisk shape and to 89.8 ± 6.6 nm for con-cave shape. The morphological change is

reversible under varying image force. Theconvex-shaped vesicles are similar to theconical relief and dome-like structure inother studies (Egawa and Furusawa, 1999;Kumar and Hoh, 2000). The disk-shapedvesicles are similar to the saucer-like vesicles(Kumar and Hoh, 2000; Jass et al., 2000).While the saucer-like vesicles are cited asan intermediate state of vesicle fusion dueto adhesion between vesicle and substrate,the morphological changes observed here canonly be attributed to the compressive tip. Thesmall size of the SUVs means that the adhe-sion alone is not enough to collapse the vesi-cle edge to form saucer-like vesicles (Seifertand Lipowsky, 1996). The morphologicalchange due to tip compression is schemati-cally illustrated in Fig. 11.9. A convex shapeis obtained in the lowest indentation regionof the force curve; a planar shape is obtainedat an intermediate indentation, and a concaveshape is obtained at the highest indentation.For the concave shape, when the tip movesacross the vesicle with an applied force heldmore or less constant, the amount of inden-tation is largest at the center of the vesicle.Several factors may contribute to maximumdepression at the center:

1. It is known that the mechanicalresponse of thin films couples to

0

10.0

nm

0

10.0nm

A B C

0

7.5nm

Figure 11.8. AFM height images obtained by tapping mode with different shapes. The image size is0.75 × 0.75 µm2. Z scale = 20 nm. The height profile across the dotted line is given. (A) Convex vesicles. (B) Planarvesicles. (C) Concave vesicles.

Page 195: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

190 RESOLVING THE PROPERTIES OF LIPID BILAYERS AND VESICLES

A

B C D

AFM Tip

Figure 11.9. Tip compression scheme on vesicle morphology: (A) Vesicle in solution. (B) Convex vesicle imagedwith minimum image force. (C) Planar vesicle imaged with intermediate indentation. (D) Concave vesicle imagedwith highest indentation.

the substrate, which results in anincrease (50%) of the apparent mod-ulus (Domke and Radmacher, 1998).The coupling increases with decreas-ing film thickness. Therefore, the tipindents more in the central region of asessile droplet.

2. A high volume percentage of materialis bound by the surface near the edgeof a droplet and spreads less uponcompression of the tip. Contrarily, atthe center of the vesicle, less resistance

exists against squeezing of the trappedliquid portion.

Figure 11.10 represents a typical forcecurve captured on EggPC vesicles. Whileit is impossible to place the tip exactly atthe center of the vesicle, the force curvewith the largest onset repulsion distance wasselected from a set of force curves obtainedin the vicinity of a vesicle, and it is used torepresent the interaction between the apexesof AFM tip and vesicle. The onset forcedistance of 32.2 nm in Fig. 11.10 falls in

−2

−1

−5 5 15 25 45

Approach

Distance (nm)

For

ce (

nN)

Retraction

35 55 65 750

1

2

Figure 11.10. Force versus distance curves on the EggPC vesicle. Two jump-in events during approach and twojump-out events during retraction are marked by arrows.

Page 196: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

SUV MICROMECHANICAL PROPERTIES 191

A B C D

Figure 11.11. Scheme of tip effect on vesicle during approach force measurement. (A) Elastic compression ofvesicle top fraction. (B) Tip penetration of the upper bilayer. (C) Further compression with tip bridging the gap.(D) Tip penetration of the lower bilayer.

the range of measured vesicles size between30 and 40 nm. The repulsion is not con-tinuous, but displays characteristic breaks.During approach, one or two breaks areobserved, around 19 and 7 nm respectively.While the exact locations of the breaks varysomewhat, the gap distance between thebeginning and the end point of the jump-inis constant at 4.8 ± 0.4 nm. During retrac-tion, similar jump-out gap is also observedthough apparently not as well defined. Thisunique gap distance coincides with lipidbilayer thickness.

We hypothesize that during approach,the bilayer portion at the top of vesiclegives away to the pressing tip and slidesaside, followed by continuous compressionof the tip on the vesicle, until tip approachesand breaks through the bottom portion ofthe bilayer enclosure. It is surprising thatvesicle maintains its enclosed shape whilethe tip bridges across different parts ofthe bilayer shell. The stepwise deformationduring approach is shown in Fig. 11.11:(1) compression of vesicle, (2) breakthroughof top portion, (3) further compression ofvesicle with tip bridging top portion, and(4) breakthrough of bottom portion with tipspanning the whole vesicle. The process isreversed during retraction except that thevesicle is stretched to 150% of its intrinsicdiameter before the final detachment of tipfrom vesicle. This adhesive interaction mayalso contribute to the shape stability ofvesicle bridged by the AFM tip. Vesicles areknown to self-heal after perforation due tohigh line tension.

11.4. SUV MICROMECHANICALPROPERTIES

The mechanical properties of phospholipidmembranes can be related to many vesiclebehaviors, such as their formation, stability,size, shape, fusion, and budding processes.Bending rigidity is a fundamental and char-acteristic mechanical property of vesicles andis related to the stability and strength ofbilayer (Svetina and Zeks, 1996).

Force curves can be used to extractquantitative micromechanical constants ofSLBs and SUVs. The force curves can befitted to the Hertzian model δ = AF b (δ is theindentation on the vesicle and F is the loadforce) by assuming a spherical shape for thetip. The indentation, δ, from the differencebetween the cantilever distance z − z0 andcantilever deflection d − d0 is described inEq. (11.1):

|z − z0| − (d − d0) = δ = A(d − d0)2/3

= 0.825

[k2(Rtip + Rves)(1 − υves

2)2

Eves2RtipRves

]1/3

× (d − d0)2/3 (11.1)

Eves is the Young’s modulus of the vesicle,Rtip and Rves are the radii of the tip andvesicle, respectively, υves is the Poisson’sratio of the vesicle, and k is the cantileverspring constant. υves is assumed to be0.5 (Laney et al., 1997; Radmacher et al.,1996). The spring constant and tip radiusare calibrated to be 0.17 N/m and 33 nm,respectively. Rves is taken to be equal toz0/2. Experimental data can be fitted to

Page 197: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

192 RESOLVING THE PROPERTIES OF LIPID BILAYERS AND VESICLES

Eq. (11.1) with two fitting parameters A andz0. In general, the fitted z0 value is foundto be consistent with the visually examinedcontact point. Thus, z0 in our experimentis determined by the onset point of therepulsive force. Eves is then computed fromA by least-square fitting.

Bending modulus kc is deduced fromYoung’s modulus based on Eq. (11.2)(Evans, 1974):

kc = Evesh3

12(1 − υves2)

(11.2)

h is the bilayer thickness.Laney et al. (1997) have calculated elas-

tic properties from averaged data of approachand retraction force curves, and they dis-card the force plots with discontinuities.The approach part of the force curvesis suitable for the indentation calculationbecause significant adhesive forces in retrac-tion can affect the measurement of indenta-tion (Vinckier and Semenza, 1998). Gener-ally, the force curves on vesicles are charac-terized by two repulsive regimes.

Figure 11.12A shows a typical deflec-tion versus z position plot on a vesiclecontaining cholesterol (EggPC 80:cholesterol20) (Liang et al., 2004c). Fig. 11.12B is aforce curve converted from Fig. 11.12A bydefining points of zero force and zero sep-aration. The force curve can be dividedinto four regions as labeled. In region I,the noncontact region, the tip is far awayfrom vesicle and the force between the tipand the vesicle is zero. Region II illus-trates the elastic deformation of the vesi-cle under tip compression and thereforecan be used to calculate Young’s modulus.Region III corresponds to further tip com-pression after the tip penetrates the vesicle’stop bilayer. Region IV reflects the cantileverdeflection when it is in contact with thehard mica substrate after penetrating throughthe vesicle’s bottom bilayer. Theoretically,the slope of region IV in Fig. 11.12Ashould be 1.0 because the deflection of

the cantilever is identical to the z direc-tion movement of sample on hard sur-face (Weisenhorn et al., 1993). Based onthe fitted data, a slope of 0.9967 ± 0.0036is obtained.

By fitting our experimental data (regionII and region III) to the Hertzian model weobtain exponent b of 0.6632 (region II) and0.9227 (region III), respectively. The poor fiton the high loading force (region III) (b =0.9227) suggests that the Hertzian modelseverely fails in region III. The exponent b =0.6632, which is close to the 2/3 in region II,is remarkable. Although the Hertzian modeldescribes the contact between two solid bod-ies on the basis of continuum elasticitytheory without adhesion force (Radmacheret al., 1994), the good fit in region II sug-gests that Hertzian model may also be appli-cable to describing the elastic deforma-tion between the tip and the vesicle withinthe limit of small indentation. Region IIillustrates the elastic deformation of thevesicle under tip compression, and it isused in subsequent calculations for deter-mining the micromechanical properties ofthe vesicle. Fig. 11.12C is the indentationversus load force converted from data inFig. 11.12A, region II. It shows that in thebeginning of the compression (indentation),the Hertz model can simulate the exper-imental findings very well. A deviationfrom the model is observed at high loadforce (larger indentation, the second repul-sive regime), illustrating the limitation of themodel.

Young’s modulus is calculated usingRtip = 33 nm, Rves = z0/2 (the onset pointof repulsive force is regarded as the size ofthe vesicle), υves = 0.5, and k = 0.17 N/m.Bending modulus is a characteristic propertyof the vesicles, which is closely related tothe activities of liposomes and the gel–liquidphase transition of liposome’s bilayer mem-brane. According to solid-state mechan-ics, Young’s modulus is related to bend-ing modulus as Eves = kc/I where I is thecross-sectional moment. The value I for a

Page 198: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

SUV MICROMECHANICAL PROPERTIES 193

−5

5

15

25

35

45

55

65

0 20 40 60 80 100 120 140z position (nm)

Def

lect

ion

(nm

)

I

II

III

IV

(A)

−2

0

2

4

6

8

−5 0 5 10 15 20 25 30 35 40 45

Distance (nm)

For

ce (

nN)

(B)

0

2

4

6

8

10

12

0 0.2 0.4 0.6 0.8 1 1.2Load force (nN)

Inde

ntat

ion

(nm)

Experimental dataHertzian model

y = 9.9291x^0.6632

(C)

Figure 11.12. (A) Deflection versus z position approaching curve. (B) Force curve converted Part A.(C) Force-curve data fit with Hertzian model for region II in Part A. The squares are experimental data. Experimentaldata can be described by power equation δ = 9.9291F 0.6632. The solid line is the Hertzian model δ = AF 2/3 withA = 9.9291. The measured compression (indentation) versus loading force agrees with the Hertzian model in thecase of the first repulsive force region.

Page 199: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

194 RESOLVING THE PROPERTIES OF LIPID BILAYERS AND VESICLES

three-dimensional, isotropic planar surface ish3/[12(1 − υ2)], where h is the thicknessof the bilayer and υ is Poisson’s ratio(Laney et al., 1997; Radmacher et al., 1996;Evans, 1974). The bending modulus kc iscalculated using Eq. (11.2) with h = 4.57 ×10−9 m and υ = 0.5. The Young’s modulusand the bending modulus for pure EggPC arefound to be 1.97 ± 0.75 MPa and (0.21 ±

0.08) × 10−19 J, respectively. The calculatedvalues are compared with literature valuesin Tables 11.2 and 11.3. The Young’s mod-ulus of the EggPC vesicle from force curvesis one order of magnitude smaller than thereported value (∼ 107 MPa) (Hantz et al.,1986). The discrepancy is probably dueto different measurement environment (Maoet al., 2004).

TABLE 11.2. Young’s Modulus of Biological Samples.

MaterialYoung’s Modulus

E (MPa) Method Remarks References

Synaptic vesicles 0.2–1.3 Force mapping Size: 90–150 nmadsorbed on mica

38a Laney et al.(1997)

DMPC vesicles 15 Osmotic swelling Size: 160–180 nm in 44 Hantz et al.(1986)

DOPC vesicles solutionEggPC vesicles 1.97 ± 0.75 Force plot Size: < 60 nm

adsorbed on mica Mao et al. (2004),Liang et al.,(2004a)

a DMPC, dimyristoylphosphatidylcholine; DOPC, dioleoylphosphatidylcholine.

TABLE 11.3. Comparison of Bending Modulus of Egg Yolk Phosphatidylcholine.

Method Size/ShapeBending Modulus

kc(×10−19 J) T (◦C) References

Phase contrastmicroscopy

Long unilamellar tubularvesicle (11 µm < L<34 µm, 17 < L/r <

83)

2.3 ± 0.3 22.0 Servuss et al. (1976)

Cylindrical vesicle(>10 µm)

1–2 25 Schneider et al.(1984a)

Quasi-spherical vesicle(>10 µm)

1–2 Schneider et al.(1984b)

Spherical (>10 µm) 0.4–0.5 46a, bMagnetic-field-

inducedorientation

Cylindrical rods (5 to30-µm diameter,<200 µm long)

0.4 25 Sakurai andKawamura (1983)

AC electricfield

Spherical vesicle (diameter>20 µm)

0.247 Kummrow andHelfrich (1991)

Spherical vesicle (∼15 to70-µm diameter)

0.66 ± 0.06, 0.45 ± 0.05 Angelova et al.(1992)

AFM forcecurve

Spherical vesicle (diameter< 60 nm) on micasubstrate

0.21 ± 0.08 22 ± 1 Mao et al. (2004),Liang et al. (2004a)

Page 200: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

SUV MICROMECHANICAL PROPERTIES 195

The calculated bending modulus is in thesame range as that reported in literaturebetween 10−19 and 10−20 J (Table 11.3).Variations in bending modulus measuredhave been reported, and the reason forthe discrepancy still needs further analysis(Kummrow and Helfrich, 1991; Angelovaet al., 1992; Niggemann et al., 1995).

Table 11.4 lists the bending moduli ofpure, cholesterol-modified, and Pluronics-modified EggPC vesicles. The data showthat EggPC vesicles are stiffened by addingcholesterol and Pluronic copolymers.

There is a significant increase in bend-ing modulus when cholesterol is incorporatedinto EggPC vesicles. It has been reported thatthe phospholipid bilayer packing geometricalstructures are changed by cholesterol inser-tion and thus by changing fluidity and intrav-esicle interaction (Liu et al., 2000). After thecholesterol is incorporated into phospholipidbilayers, the small hydrophilic 3β-hydroxylheadgroup of cholesterol is located in thevicinity of the lipid ester carbonyl groups,and the hydrophobic steroid ring orients itselfparallel to the acyl chains of the lipid (Lad-brooke et al., 1968). Thus, the movement ofthe acyl chains of the phospholipid bilayerhas been restricted. Below the gel phasetransition temperature (Tm) of lipids, choles-terol addition increases the fluidity of lipids,while above Tm the mobility and fluidityof the lipid chains are restricted (Tm ofEggPC = −15◦C) (New, 1990). Moreover,

hydrogen bonding between cholesterol’s β-OH and the carbonyl groups of the lipidenhances the stability of the bilayer (Prestiet al., 1982). The interaction between thecholesterol and phospholipid bilayer resultsin an increase in membrane cohesion, asshown by increases in the mechanical stiff-ness of the membranes.

The slope of the long-range repulsiveforce regime in Fig. 11.3 can be used to cal-culate the elastic properties of the Pluronics-modified EggPC SUVs based on Eqs. (11.1)and (11.2). The slopes of EggPC/F88 andEggPC/F127 are found to be lower than thatof EggPC/F108. The slope of EggPC/F127is close, but slightly higher than that ofEggPC/F88. Both F127 and F88 have simi-lar PEO size, while the PPO chain of F127 islonger. Table 11.3 lists the calculated bend-ing modulus values. The bending modulus ofPluronics-modified EggPC SUVs increasesby an order of magnitude from that of pureEggPC. The magnitude of the increase iscomparable with cholesterol/EggPC vesicles.These data show that the bending moduli ofPluronics-modified EggPC SUVs are a func-tion of PEO and PPO chain lengths. The dra-matic increase can be attributed to PPO blockincorporation and PEO chain on the surface.For Pluronics-modified SUVs, the physicalstability is improved by the PEO steric repul-sion (Kostarelos et al., 1998, 1999). Thebending modulus obtained on F127 vesiclesis (1.64 ± 0.28) × 10−19 J, slightly largerthan that of F88 ((1.50 ± 0.40) × 10−19 J).

TABLE 11.4. Bending Modulii of Pure, Cholesterol-Modified, and Pluronics-Modified EggPC Vesicles.

Sample Bending Modulus References

Pure EggPC 0.27 ± 0.10 Mao et al. (2004), Liang et al. (2004a)EggPC:Chol (85:15) 1.68 ± 0.21 Liang et al. (2004c)EggPC:Chol (80:20) 1.49 ± 0.09EggPC:Chol (70:30) 1.44 ± 0.56EggPC:Chol (50:50) 1.81 ± 0.41EggPC/F88 1.50 ± 0.40 (103 EO, 40 PO) Liang et al. (2004b)EggPC/F127 1.64 ± 0.28 (100 EO, 65 PO)EggPC/F108 2.06 ± 0.38 (133 EO, 50 PO)

Page 201: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

196 RESOLVING THE PROPERTIES OF LIPID BILAYERS AND VESICLES

The increase can be attributed to longerPPO chain length of F127. While comparingbending moduli of F127 and F108, the cir-cumstances are somewhat complicated sinceboth have different PPO and PEO chainlengths. The bending modulus of F108 ishigher than that of F127, although the PPOchain length of F108 is smaller. However,F108 has longer PEO chain, and stiffer shell-like “coating” on the vesicles seems to con-tribute to bending modulus increase.

It is well known that the stability ofpolymer-coating liposome is improved interms of steric repulsive force betweenthe polymer chains dangling outside ofthe liposome. In earlier studies, the stericstabilization effect has been described byaggregation and fusion among vesicles insolution (Kostarelos et al., 1998, 1999).Kostarelos et al. (1998) draw a similarconclusion by comparing prolonged vesi-cle durability against flocculation and ζ-potential change. It has been reported thatthe bending and mechanical properties ofvesicles from diblock polymer exceeds those

of phospholipid liposomes by a factor of5 or more (Discher et al., 1999). Thus, thesignificant bending modulus improvementcan be attributed to PEO (incorporated) andPPO chains (dangling outside of EggPC vesi-cles). Fig. 11.13 shows a schematic of thesteric stabilization effect of copolymer onliposome. A block copolymer may aggregateto form micelles (core/shell structure) con-sisting of a swollen core of insoluble blockssurrounded by a flexible fringe of solubleblock (Tuzar et al., 1993). As an analogy tothe polymer micelle, the PEO chains dan-gling outside the vesicle surface may exhibita shell-like effect to provide steric stabiliza-tion to the vesicle. The shell-like structurealso serves as a kind of “net” or “coating”of vesicles (Ringsdorf et al., 1988). Withlonger PEO chain length (>19 units), a finitesteric barrier is formed (Wang et al., 2002).The PPO chain incorporated into the lipidbilayer acts in a similar fashion as choles-terol to restrict the lipid molecular move-ment and fluidity. For copolymer with longerPEO chain length (F88, F108, and F127),

PPO Chain PEO Chain

Figure 11.13. Scheme of the Pluronics PEO (right) and PPO effect (left) on the stability of the EggPC vesicle.(Left) SUVs with long attached PEO chains can form shell-like structure (black dotted circle) due to sufficient stericrepulsion provided by the dangling PEO chains away from the vesicle surface. (Right) The PPO chain incorporatedinside the lipid bilayer restricts the bilayer fluidity and movement, and makes the lipid bilayer membrane more rigid.

Page 202: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 197

shell-like wall structure is strong enough tokeep all EggPC vesicles in an intact formupon adsorption on mica (Liang et al., 2005).

11.5. SUMMARY

This chapter describes experimental methodsto determine the thickness and micromechan-ical properties of supported bilayers and vesi-cles using AFM imaging and force measure-ment. Fused bilayers and sonicated unilamel-lar vesicles of EggPC are used as examplesto illustrate the various approaches for filmthickness determination. The AFM imagingof lipid layers is ideally conducted in naturalbiological solutions using tapping or con-tact mode at below a threshold force. Theadsorbed vesicles of EggPC below 50 nm insize can be imaged in an intact state usingthe soft contact mechanism. The bilayerthickness can be accurately determined bythe step height at bilayer domain edges.Alternatively, the jump-in distances can beused to obtain the bilayer thickness. Thejump-in distances change when biologicalor polymeric additives are integrated intothe lipid bilayer. The distance increase canbe matched to the hydrophilic block chainlength when nonionic Pluronic copolymersare adsorbed onto the EggPC bilayer. Jump-in points are also observed in supportedintact vesicles, with one corresponding tothe upper bilayer and another correspond-ing to the lower, surface-attached bilayer.The two gaps are identical in value, whichis the lipid bilayer thickness. These gapscan be interpreted as abrupt jumps of theAFM tip across the top and bottom portionof the bilayer enclosure during approach andretraction. AFM can be also used to monitorthe shape changes at different image forces.The indentation curve converted from theforce curve can be fitted to the Hertz spher-ical contact model in order to extract theYoung’s modulus of the vesicle. The calcu-lated Young’s modulus and bending modu-lus of the EggPC SUVs are (1.97 ± 0.75) ×

106 Pa and (0.21 ± 0.08) × 10−19 J, respec-tively. The elastic constants are comparedto literature values from various microme-chanical tests of liposomes. AFM methodsare the most direct way to determine bilayerfilm thickness and offer new insights intoadsorption, spreading, fusion, self-healing,and mechanical properties of biomembranesand liposomes at the nanoscale.

REFERENCES

A-Hassan, E., Heinz, W. F., Antonik, M. D.,D’Costa, N. P., Nageswaran, S., Schoenen-berger, C.-A., and Hoh, J. H. (1998). Relativemicroelastic mapping of living cells by atomicforce microscopy. Biophys. J. 74:1564.

Angelova, M. I., Soleau, S., Meleard, P., Fau-con, J. F., and Bothorel, P. (1992). Preparationof giant vesicles by external AC electric fields,kinetic and applications. Progr. Colloid Polym.Sci. 89:127.

Barenholz, Y. (2001). Liposome application:Problems and prospects, Curr. Opin. ColloidInterface Sci. 6:66.

Bayerl, T. M., and Bloom, M. (1990). Physicalproperties of single phospholipid bilayersadsorbed to micro glass beads. A new vesicularmodel system studied by 2H-nuclear magneticresonance. Biophy. J. 58:357.

Blumenthal, R., Clague, M. J., Durell, S. R., andEpand, R. M. (2003). Membrane fusion. Chem.Rev. 103:53.

Cooper, M. A. (2004). Advances in membranereceptor screening and analysis. J. Mol.Recognit. 17:286.

Discher, B. M., Won, Y., Ege, D. S., Lee, J. C.,Bates, F. S., Discher, D. E., and Hammer, D. A.(1999). Polymersomes: Tough vesicles madefrom diblock copolymers. Science 284:1143.

Domke, J., and Radmacher, M. (1998). Measur-ing the elastic properties of thin polymer filmswith the atomic force microscope. Langmuir14:3320.

Dong, J., and Mao, G. (2000). Direct study ofC12E5 aggregation on mica by AFM imagingand force measurements. Langmuir 16:6641.

Dufrene, Y. F., and Lee, G. U. (2000). Advancesin the characterization of supported lipid films

Page 203: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

198 RESOLVING THE PROPERTIES OF LIPID BILAYERS AND VESICLES

with the atomic force microscope. Biochim.Biophys. Acta 1509:14.

Dufrene, Y. F., Boland, T., Schneider, J. W.,Barger, W. R., and Lee, G. U. (1998). Charac-terization of the physical properties of modelmembranes at the nanometer scale with theatomic force microscope. Faraday Discuss.111:79.

Egawa, H., and Furusawa, K. (1999). Liposomeadhesion on mica surface studied by atomicforce microscopy. Langmuir 15:1660.

Engelhardt, H. P., Duwe, H., and Sackmann, E.Bilayer bending elasticity measured by Fourieranalysis of thermally excited surface undula-tions of flaccid vesicles. J. Phys. Lett. France46:L395.

Eskilsson, K., Ninham, B. W., Tiberg, F., andYaminsky, V. V. (1999) Effects of adsorptionof low-molecular-weight triblock copolymerson interactions between hydrophobic surfacesin water. Langmuir 15:3242.

Evans, E. A. (1974). Bending resistance andchemically induced moments in membranebilayers. Biophys. J. 14:923.

Evans, E., and Rawicz, W. (1990). Entropy-driven tension and bending elasticity incondensed-fluid membranes. Phys. Rev. Lett.64:2094.

Faucon, J. F., Mitov, M. D., Meleard, P., Bivas, I.,and Bothorel, P. (1989). Bending elasticityand thermal fluctuations of lipid membranes.Theoretical and experimental requirements, J.Phys. France 50:2389.

Franz, V., Loi, S., Muller, H., Bamberg, E., andButt, H. J. (2002). Tip penetration through lipidbilayers in atomic force microscopy. ColloidsSurf. B 23:191.

Hantz, E., Cao, A., Escaig, J., and Taillandier, E.(1986). The osmotic response of large unil-amellar vesicles studied by quasielastic lightscattering. Biochim. Biophys. Acta 862:379.

Heinz, W. F., and Hoh, J. H. (1999). Spatiallyresolved force spectroscopy of biologicalsurfaces using the atomic force microscope.Tibtech 17:143.

Hope, M. J., Bally, M. B., Webb G., andCullis, P. R. (1985). Production of large unil-amellar vesicles by a rapid extrusion procedure.Characterization of size distribution, trappedvolume and ability to maintain a membranepotential. Biochim. Biophys. Acta 821:55.

Huang, C.-H. (1969). Studies on phosphatidyl-choline vesicles formation and physical char-acteristics. Biochemistry 8:344.

Jass, J., Tjarnhage, T., and Puu, G. (2000).From Liposomes to Supported, planar bilayerstructures on hydrophilic and hydrophobicsurfaces: An atomic force microscopy study.Biophys. J. 79:3153.

Johnsson, M., Silvander, M., Karlsson, G., andEdwards, K. (1999). Effect of PEO-PPO-PEOtriblock copolymers on structure and stabilityof phosphatidylcholine liposomes. Langmuir15:6314.

Kostarelos, K., Luckham, P. F., and Tadros, T. F.(1995). Addition of block copolymers toliposomes prepared using soybean lecithin.Effects on formation, stability and the specificlocalization of the incorporated surfactantsinvestigated. J. Liposome Res. 5:117.

Kostarelos, K., Luckham, P. F., and Tadros, T. F.(1998). Steric stabilization of phospholipidvesicles by block copolymers—vesicle floccu-lation and osmotic swelling caused by monova-lent and divalent cations. J. Chem. Soc. Fara-day Trans. 94:2159.

Kostarelos, K., Tadros, T. F., and Luckham, P. F.(1999). Physical conjugation of (tri-) blockcopolymers to liposomes toward the construc-tion of sterically stabilized vesicle systems.Langmuir 15:369.

Kumar, S., and Hoh, J. H. (2000). Direct visual-ization of vesicle–bilayer complexes by atomicforce microscopy. Langmuir 16:9936.

Kummrow, M., and Helfrich, W. (1991). Defor-mation of giant lipid vesicles by electric fields.Phys. Rev. 44:8356.

Ladbrooke, B. D., Williams, R. M., and Cha-pan, D. (1968). Studies on lecithin–choles-terol–water interactions by differential scan-ning calorimetry and x-ray diffraction. Biochim.Biophys. Acta 150:333.

Laney, D. E., Garcia, R. A., Parsons, S. M., andHansma, H. G. (1997). Changes in the elasticproperties of cholinergic synaptic vesicles asmeasured by atomic force microscopy. Biophys.J. 72:806.

Lasic, D. D. (1998). Novel applications ofliposomes. Tibtech 16:307.

Liang, X., Mao, G., and Ng, K. Y. S. (2004a).Probing small unilamellar EggPC vesicles on

Page 204: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 199

mica surface by atomic force microscopy,Colloids Surf. B: Biointerfaces 34:41.

Liang, X., Mao, G., and Ng, K. Y. S. (2005).Effect of chain lengths of PEO-PPO-PEO onsmall unilamellar liposome morphology andstability: An AFM investigation, J. ColloidInterface Sci., 285:360–372.

Liang, X., Mao, G., and Ng, K. Y. S. (2004c).Mechanical properties and stability measure-ment of cholesterol-containing liposome onmica by atomic force microscopy. J. ColloidInterface Sci. 278:53.

Liu, D., Chen, W., Tsai, L., and Yang, S. (2000).Effects of cholesterol on the release of freelipids and the physical stability of lecithinliposomes. J. Chin. Inst. Chem. Engrs. 31:269.

Loi, S., Sun, G., Franz, V., and Butt, H.-J.(2002). Rupture of molecular thin filmsobserved in atomic force microscopy. II.Experiment. Phys. Rev. E 66:031602.

Manne, S., Cleveland J. P., Gaub, H. E.,Stucky G. D., and Hansma, P. K. (1994).Direct visualization of surfactant hemimicellesby force microscopy of the electrical doublelayer. Langmuir 10:4409.

Mao, G., Liang X., and Ng, K. Y. S. (2004).Direct force measurement of liposomes byatomic force microscopy. In: The DekkerEncyclopedia of Nanoscience and Nanotech-nology (J. A. Schwarz, C. I. Contescu, andK. Putyera, eds.), pp. 923–932, Marcel Dekker,New York.

McIntosh, T. J., Magid, A. D., and Simon, S. A.(1987). Steric repulsion between phosphatidyl-choline bilayers. Biochemistry 26:7325.

New, R. R. C. (1990). In: Liposomes: A PracticalApproach (R. R. C. New, Ed.), Oxford Univer-sity Press, New York.

Niggemann, G., Kummrow, M., and Helfrich, W.(1995). The bending rigidity of phosphatidyl-choline bilayers: Dependences on experimentalmethod, sample cell sealing and temperature. J.Phys. II France 5:413.

Patrick, H. N., Warr, G. G., Manne, S., andAksay, I. A. (1997). Self-assembly structuresof nonionic surfactants at graphite/solutioninterfaces. Langmuir 13:4349.

Pignataro, B., Steinem, C., Galla, H-J., Fuchs, H.,and Janshoff, A. Specific adhesions of vesiclesmonitored by scanning force microscopy andquartz crystal microbalance. Biophys. J. 78:487.

Prater, C. B., Maivald, P. G., Kjoller, K. J., andHeaton, M. G. (1995) Probing Nano-scaleForces with the Atomic Force Microscope.Application Note No. 8.

Presti, F. T., Pace, R. J., and Chen, S. I. (1982).Cholesterol–phospholipid Interaction in mem-branes. 2. Stoichiometry and molecular pack-ing of cholesterol-rich domains. Biochemistry21:3831.

Quate, C. F. (1994). The AFM as a tool forsurface imaging. Surf. Sci. 299/300:980.

Radmacher, M., Tillmann, R. W., Fritz, M., andGaub, H. E. (1992). From molecules to cells:Imaging soft samples with the atomic forcemicroscope. Science 257:1900.

Radmacher, M., Fritz, M., Kacher, C. M., Wal-ters, D. A., and Hansma, P. K. (1994). Imag-ing adhesion forces and elasticity of lysozymeadsorbed on mica With the atomic force micro-scope. Langmuir 10:3809.

Radmacher, M., Fritz, M., Kacher, C. M., Cleve-land, J. P., and Hansma, P. K. (1996). Mea-suring the viscoelastic properties of humanplatelets with the atomic force microscope. Bio-phys. J. 70:556.

Reviakine, I., and Brisson, A. (2000). Formationof supported phospholipid bilayers fromunilamellar vesicles investigated by atomicforce microscopy. Langmuir 16:1806.

Ringsdorf, H., and Schlarb, B. Venzmer, J.(1988). Molecular architecture and function ofpolymeric oriented systems. J. Angew. Chem.Int. Ed. Engl. 27:113.

Sackmann, E. (1996). Supported membranes:Scientific and practical applications. Science271:43.

Sakurai, I., and Kawamura, Y. (1983). Magnetic-field-induced orientation and bending ofthe myelin figures of phosphatidylcholine.Biochim. Biophys. Acta 735:189.

Schneider, M. B., Jenkins, J. T., and Webb, W. W.(1984a). Thermal fluctuations of large cylindri-cal phospholipid vesicles. Biophys. J. 45:891.

Schneider, M. B., Jenkins, J. T., and Webb, W. W.(1984b). Thermal fluctuations of large quasi-spherical bimolecular phospholipid vesicles. J.Phys. France 45:1457.

Seifert, U., and Lipowsky, R. (1996). Shapesof fluid vesicles. In Handbook of Non-medical Applications of Liposomes—Theoryand Basic Sciences, Vol. I, (D. D. Lasic and

Page 205: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

200 RESOLVING THE PROPERTIES OF LIPID BILAYERS AND VESICLES

Y. Barenholz, eds.), pp. 43–85, CRC Press,Boca Raton, FL.

Servuss, R. M., Harbich, W., and Helfrich, W.(1976). Measurement of the curvature-elasticmodulus of egg lecithin bilayers. Biochim.Biophys. Acta. 436:900.

Shibata-Seki, T., Masai, J., Tagawa, T., Sorin, T.,and Kondo, S. (1996). In-situ atomic forcemicroscopy study of lipid vesicles adsorbed ona substrate. Thin Solid Film 273:297.

Svetina, S., and Zeks, B. In: (D. D. Lasic andY. Barenholz, eds.), Handbook of NonmedicalApplications of Liposomes, Chapter 1, CRCPress, Boca Raton, FL.

Thomson, N. H., Collin, I., Davies, M. C.,Palin, K., Parkins, D., Roberts, C. J.,Tendler, S. J. B., and Williams, P. M. (2000).Atomic force microscopy of cationic lipo-somes. Langmuir 16:4813.

Tortonese, M., and Kirk, M. (1997). Characteri-zation of application specific probes for SPMs.SPIE 3009:53.

Tuzar, Z., and Kratochvil, P. (1993). In: Surfaceand Colloid Science, Vol. 15 (E. Matijevic,ed.), Plenum Press, New York.

Villarrubia, J. S. (1997). Algorithms for scannedprobe microscope image simulation, surfacereconstruction, and tip estimation. J. Res. Natl.Inst. Stand. Technol. 102:425.

Wang, A., Jiang, L., Mao, G., and Liu, Y. (2002).Direct force measurement of silicone- andhydrocarbon-based ABA triblock surfactants inalcoholic media by atomic force microscopy. J.Colloid Interface Sci. 256:331.

Waugh, R. E., Song, J., Svetina, S., and Zeks, B.(1992). Local and nonlocal curvature elasticityin bilayer membranes by tether formation fromlecithin vesicles. Biophys. J. 61:974.

Weisenhorn, A. L., Khorsandi, M., Kasas, S.,Gotzos, V., and Butt, and H.-J. Deformationand height anomaly of soft surfaces studiedwith an AFM. Nanotechnology 4:106.

Woodle, M. C., Newman, M. S., and Work-ing, P. K. (1995). In: Stealth Liposomes(D. Lasic and D. F. Martin, eds), pp. 103–118,CRC Press. Boca Raton, FL.

Vinckier, A., and Semenza, G. (1998). Measuringelasticity of biological materials by atomicforce microscopy. FEBS Lett. 430:12.

Page 206: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

CHAPTER 12

IMAGING SOFT SURFACES BY SFMANDREAS JANKELeibriz-Institut fur Polymerforschung Dresden eV., Hohe Str. 6, 01005 Dresden, Germany

TILO POMPELeibriz-Institut fur Polymerforschung Dresden eV., Hohe Str. 6, 01005 Dresden, Germany

12.1. INTRODUCTION

Since its invention in 1986 (Binning et al.,1986), the atomic force microscope hasbecome a scientific tool with steadily grow-ing use. Its open modular construction allowfor the inclusion of various input parame-ters and applications. Various sensing mecha-nisms and improvements in the sensing toolspermit the microscope’s application in mate-rial research and physics as well as imag-ing techniques. Nowadays, scanning probemicroscopy (SPM) is a tool for microscopy,force spectroscopy, and nanometer manip-ulation in many fields of industry andresearch. With the scanning probe micro-scope, imaging of crystal structures withatomic resolution became as readily possi-ble as (a) the measuring and mapping of theforces of specific chemical bonds on surfacesor (b) writing nanometer-sized structures ofmono-molecular layers onto surfaces.

Most recently, scanning force techniqueshave found their way into life science appli-cations. The major reason for this may befound in SPM’s own characteristics: thehigh resolution of the scanning probe micro-scope in the nanometer range, the great

sensitivity of the probe techniques, andthe microscope’s open construction. Thislast characteristic allows the flexible adap-tation of scanning force microscopy (SFM)techniques to the environmental conditions,which is absolutely necessary for investigat-ing living systems. This adaptability is onemajor advantage using SPM and its availabletechniques over high-resolution techniquessuch as (a) scanning electron microscopyor (b) its improvement for higher-pressureconditions, environmental scanning electronmicroscopy. On the other hand, SPM opticaltechniques, which permit application underenvironmental conditions, are limited to reso-lutions of only about 0.5 µm. However, SPMoffers the opportunity to image, investigate,and manipulate soft structures with nanome-ter resolution under several controlled envi-ronmental conditions, (namely temperature,humidity, gas content) and directly in fluids.Furthermore, SPM readily allows measure-ment of properties of living tissues, such aselasticity of cell structure elements and bind-ing forces of antibodies.

Despite its usefulness as a research toolin life science, the use of the SPM requirescertain precautions and special conditions

Force Microscopy: Applications in Biology and Medicine, edited by Bhanu P. Jena and J.K. Heinrich Horber.Copyright 2006 John Wiley & Sons, Inc.

201

Page 207: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

202 IMAGING SOFT SURFACES BY SFM

because of the softness and high sensitivityof living structures. This often results indeviation from standard SFM applications.The goal of this chapter is to providean overview of the basic principles ofSPM techniques in SFM, along with itsapplications and problems, with special focuson the use of these techniques in livingstructures. Various probing tools for thesetechniques are described, and the chapterconcludes with the presentation of examplesillustrating several of the special features,problems, and artifacts that can arise usingSPM in the imaging of soft surfaces.

12.2. IMAGING SOFT SURFACES:PHYSICS AND METHODS

In this section, the setup of typical scanningforce microscopes, along with methods andprobes used for investigation, is explained.The section cannot provide a detailed appli-cation description for certain experiments.

For that the reader is referred to otherchapters of this book. However, some spe-cial features generally important for imagingsoft surfaces are outlined.

12.2.1. Experimental Setup

The first section will deal with the gen-eral setup of a scanning force microscope. Itconsists schematically of three major parts.Figure 12.1 illustrates the setup describednext. The central part is the detection unit,which is a small tip mounted on a cantilever.The cantilever itself is fixed at a stage, whichcan be moved by a piezoelectric crystal. Thispiezoelectric crystal belongs to the secondmajor part, which is the scanning unit, con-sisting of piezoelectric crystals. Two differ-ent setups are available. In one, the sampleis moved in x, y, and z direction on a stageand the cantilever stage remains fixed. In thesecond setup, the piezoelectric crystals andthe detection unit are integrated in one stageand the sample remains fixed. The scanning

Mirror

Laser diode

Beam path

Cantilever substrate

Sample

Flexiblecantilever

xyz Piezoelectricscanner

4-Quadrantphotodiode

Figure 12.1. Sketch of the experimental setup of a scanning force microscope. The xyz-scanner provides themovement of the mounted sample in the three directions. The beam deflection principle is illustrated. The laserbeam hits the cantilever. By reflection of the laser beam, distortion and bending of the cantilever are detected withvery high precision on the four-quadrant photodiode.

Page 208: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

IMAGING SOFT SURFACES: PHYSICS AND METHODS 203

range of piezoelectric crystals is limited, soappropriate scanners must be used to ascer-tain the dimensions of the range of interest(e.g., atomic resolution—small-range scan-ner; imaging of whole cells—large-rangescanner, up to 200 µm). The movement ofthe piezoelectric crystals is controlled by anelectronic and computer unit, which is thethird major part.

How is the detection of surface forcesor structures performed? The main principleis the beam deflection. A beam of a laserdiode is focused on the back of the very endof a cantilever, on which a tip is mountedon the other side. The reflected laser beamis detected by a four-quadrant photodiode.When the cantilever is bent or distorteddue to interactions between the tip and thesurface by any kind of force, the bending ordistortion gives rise to an amplified changeof the laser beam position on the photodiode.In this way, changes of the tip position downto the angstrom range can be detected. Thesignal of the photodiode is used to give afeedback to the controlling unit, so that thepiezoelectric crystals can be adjusted in thez direction to keep the bending on a constantlevel while scanning in x –y direction overthe surface. This mode is called imagingmode or constant force mode, meaning that aconstant force between tip and surface leadsto a constant bending of the cantilever. Inthis mode the z-axis position of the cantileveris plotted over the x –y plane leading to aconstant force map, which is used as thetopography image.

In the spectroscopy mode the feedbackis disabled and the tip is moved in thez direction only. The distortion of thecantilever is measured in dependence of thedistance of the tip from the surface giving theinteraction force of the tip with the samplesurface in dependence of distance.

With this general setup, SFM imagingstarted its development to a powerful toolin a wide variety of applications. Many ofthem used special modes of imaging andspectroscopy and special probing tools to

differentiate between different forces. Thenext two sections will focus on these modesand tools.

12.2.2. Modes

The operation modes of SPM are determinedby the type of tip scanning.

In contact mode the tip is in a repulsivecontact with the surface and is dragged with aconstant velocity over the sample. Scanningis possible in two different variations ofthis mode: constant force mode, alreadymentioned above, and constant height mode.The constant force mode is useful forimaging rough surfaces (minimizing artifactsdue to variable load) and for friction forcemicroscopy (a constant normal load is arequirement for good friction imaging). Adisadvantage of this mode, especially forimaging very rough samples, is the limitedscan velocity by the time constants of thefeedback loop; typical scan frequencies arein the range of 1–6 Hz per scan line. In thedeflection mode the piezo-tube extension (inz direction) is constant and the cantileverdeflection is recorded. This mode is usefulfor flat samples, and in this case the load isonly nearly constant and high scan speeds areallowed because the feedback gains has beenset to zero. Typical scan frequencies range upto 60 Hz. A high scan speed can be usefulfor scanning small areas below 300 nm tominimize the influence of thermal drift ofsample and sample holder.

Under ambient conditions (humid air),only the contact mode provides the highestlateral resolution of 0.1–0.2 nm down tothe true atomic resolution under appropriateconditions (Ohnesorge and Binnig, 1993).However, scanning under ambient conditionshas disadvantages for the contact mode: highlocal pressure and shear stresses on thesurface. Each surface in air is covered withfluid, in most cases water; the thickness ofthis layer depends on the hydrophobicityof the surface. In contact mode the fluidlayers form a meniscus between surfaceand tip, and the resulting capillary forces

Page 209: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

204 IMAGING SOFT SURFACES BY SFM

Figure 12.2. Contact mode image (7 µm × 7 µm) of the surface of a ternary copolyester. The inner region showsthe result of scanning a 4-µm × 4-µm image (scan angle 45◦) with the same normal load and with the same scanfrequency, but the smaller image size results in lower tip velocity; the tip is now able to move a certain amount ofmaterial and can generate an artificial structure.

Plexiglas

"O" ring Sample Liquid

Figure 12.3. Sketch of a liquid cell for a “scanning sample” setup.

are the main contribution to the contactforces in a range from 5 to 1000 nN.With the small contact area (10–100 nm2),even very low normal forces in the nano-newton-range result in very high normalpressure and shear stresses in the rangeof 0.1–100 GPa (Tsukruk, 1997). In most

cases, such high local stresses damage softsurfaces, an example of which is shownin Fig. 12.2.

Imaging in fluids (water, buffer solu-tion) can circumvent this drawback. In thiscase, both sample surface and cantilever areimmersed in the liquid (Fig. 12.3), resulting

Page 210: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

IMAGING SOFT SURFACES: PHYSICS AND METHODS 205

in no capillary forces. The remaining contactforces are in the range of a few hundredpico-newtons.

For topographic imaging, the fast scandirection is normally parallel to the can-tilever symmetry axis and the photodiodesignal generated by the vertical movementof the cantilever is used for the feedbacksystem of a z-scanner and for topographyinformation. With the fast scan direction per-pendicular to the cantilever symmetry axis,another form of SPM in contact mode can beperformed: friction force microscopy (FFM),also called lateral force microscopy (LFM).The signal at the four-quadranted photodi-ode is generated by two kinds of cantilevermotion (Fig. 12.4), the vertical bending pro-vides information about the topography (=(A + C) − (B + D)), and the torsion and thehorizontal bending of the cantilever are a mea-sure of the friction force (= (A + B) − (C +D)). Topography and friction images are takensimultaneously, making it possible to investi-gate material contrast alongside surface mor-phology. Small height steps that are hardlyvisible in topography images can be detectedin friction images. Figures 12.5 and 12.6 showthe principles and an example of FFM.

To overcome surface damage problemsdue to the high local pressure in con-tact mode, the dynamic mode (often called“tapping mode” or “intermittent contactmode”) has been developed (Zhong et al.,

1993; Spatz et al., 1995; Winkler et al.,1996; Tamayo and Garcia, 1996). In thismode, the instant tip-surface contact isreplaced with short contacts of an oscillat-ing tip (Fig. 12.7). The cantilever with thetip is mounted on a piezo stack in a specialcantilever holder that drives the cantilevernear or at its resonance frequency (typicallyin the range of 70–300 kHz). The oscilla-tion is detected as RMS signal (amplitude) atthe photodiode. In the vicinity of the surface,weak interactions between tip and surfacecan significantly reduce the amplitude of can-tilever oscillation: The tip slightly “taps” onthe sample surface during scanning, contact-ing the surface at the bottom of its oscil-lation turning point. The amplitude of theoscillation is reduced by two mechanisms:(1) shifting of the effective resonance fre-quency of the cantilever-interacting surfacesystem and (2) damping due to energy dis-sipation. The feedback loop of the controllermaintains the reduced amplitude constant byvariation of the z distance of the scanner ateach (x, y) data point (like the constant forcemode in contact mode AFM), while also pro-viding information about the topography. Ifthe free amplitude (with the tip far away fromthe surface) is high enough (i.e., greater than20 nm), the absorbed fluid layer on the sur-face will be penetrated without getting stuck.At sufficient low free amplitudes (<5 nm)the fluid layer can be imaged.

Cantilever

Lateralcomponent

Verticalcomponent

Detector

A

B D

C

Figure 12.4. Cantilever motion in contact mode. The vertical component (signal (A + C) − (B + D)) givesinformation about topography, and the lateral component (signal (A + B) − (C + D)) results from friction forces.

Page 211: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

206 IMAGING SOFT SURFACES BY SFM

Sample

LFM image

Different material

(a)

Sample

LFM image

(b)

Figure 12.5. Principle of lateral force microscopy (LFM). (A) Material contrast. (B) Detection of small edges.

Figure 12.6. Arachine acid adsorbed from chloroform solution on mica. (Left) Height image—brighter parts arehigher regions. (Right) Simultaneously taken friction image (dark—low friction; bright—high friction). The arachineacid shows a significantly lower friction than mica, and the coverage is more clearly seen as in the height image.

Page 212: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

IMAGING SOFT SURFACES: PHYSICS AND METHODS 207

“Free” amplitude

Amplitude reduced

Fluid layer“Tapping”

Figure 12.7. Principle of tapping mode. The reduced amplitude will be maintained during scanning.

Maintaining a constant reduced amplitudemeans also a constant tip–sample interactionis maintained during scanning. Compared tocontact mode, the typical tip–surface interac-tion forces are reduced by at least one orderof magnitude (like the forces in fluid celloperation in contact mode). Only compressionforces—and most soft samples are insensi-tive to such forces—have any effect on thesample surface; shear forces are absent. There-fore, dynamic mode is the first choice forimaging soft sample surfaces under ambientconditions. The scan velocity in the dynamicmode is slightly lower than in contact mode;scan frequencies in tapping mode should notexceed 1.5 Hz per scan line (Digital Instru-ments, 1993). The lateral resolution on mostsamples (depending on scan size and tipradius) is in the range of 1–5 nm.

The next technique of force reductionbetween tip and sample surface involvesthe dynamic mode in fluids. In this way,forces down to tens of pico-newton can bereached. Furthermore, active electronic de-damping of the cantilever oscillation (calledQ control) can enhance the sensitivity ofthe dynamic mode by orders of magni-tude due to the enhancement of the Q fac-tor. The quality factor Q determines howfast an oscillating system (cantilever + tip)can respond to a change of the interaction

forces acting on the tip. The quality fac-tor is defined as Q = �ω/ω0, where ω0

is resonance frequency and �ω is halfof full maximum width of the resonancecurve. At low Q (Fig. 12.8A), a relativelystrong interaction (change in frequency) isneeded to reach the chosen reduced ampli-tude. At higher Q (Fig. 12.8B), the systemis much more sensitive: The reduced ampli-tude can be reached by tiny changes of thetip–surface interaction. The quality factorcan be changed by Q control (Anczykowskiet al., 1998; Humphris et al., 2000), an extrafeedback circuit with a phase shifter and anadjustable amplifier added to the standardSFM setup. This circuit applies an addi-tional force via the standard piezo, which isused to drive the cantilever base. The fre-quency range of modern Q-control devices(5 kHz to 1 MHz) allows applications fromliquid dynamic mode (low frequency) tohigh frequency dynamic mode in air. Anexample of Q control is shown in Fig. 12.9:tapping mode images of the surface of apoly(hydroxybutyrate) film taken in air witha used, broken tip; without Q control (left)an artifact appears due to a “double” tip, withQ control (right), only the nearest tip touchesthe surface resulting in an artifact free image.

Apart from amplitude detection, thedynamic mode also allows the detection

Page 213: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

208 IMAGING SOFT SURFACES BY SFM

Figure 12.8. Amplitude and phase curve in a frequency sweep of a 125-µm-long dynamic mode silicon cantilever(Nanosensors, Germany) in air. (A) Without Q control, Q = 195. (B) With Q control, Q = 2542.

Figure 12.9. An example of Q control: Surface of poly(hydroxybutyrate) film on silicon wafer imaged by tappingmode in air with a used broken tip. (Left) Without Q control, artifacts resulting from a “double” tip are clearly seen(arrows). (Right) With Q control, only the nearest tip of the “double” tip touches the surface, the tip artifacts arevanished.

Page 214: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

IMAGING SOFT SURFACES: PHYSICS AND METHODS 209

Figure 12.10. Principle of phase shift detection. The electronics calculate the phase shift between the phase signalof the cantilever drive voltage and the phase signal of the real cantilever oscillation detected at the photodiode.

Figure 12.11. Phase shift depending on the kind of tip–surface interaction (explanation in the text).

of phase shift between the oscillating volt-age at the cantilever drive piezo andthe real cantilever oscillation detected atthe photodiode (Fig. 12.10). Interactions ofthe tip with the surface can reduce theamplitude, and the phase shift can bechanged by the influence of different mate-rial properties—for example, viscoelasticity,

stiffness, and electric and magnetic fields.The mechanism of phase imaging is depictedin Fig. 12.11: The phase of the freevibrating cantilever at its resonance fre-quency ω0 is � = 90◦; under the influenceof attractive forces, the effective spring con-stant decreases, leading to lower resonancefrequency, but the cantilever will be further

Page 215: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

210 IMAGING SOFT SURFACES BY SFM

driven at ω0, with the result of a phase shift��a . This behavior is reversed in the caseof repulsive interaction, leading to a phaseshift ��r . For practical phase imaging it isimportant to ensure scan conditions for eitherrepulsive interaction (stiffness or viscoelas-ticity contrast) or attractive interaction (adhe-sion contrast). According to Magonov et al.(1997a, b), the scan conditions for repulsiveinteraction are a free amplitude greater than100 nm and a setpoint ratio (reduced ampli-tude/free amplitude) of 0.5, the scan con-ditions for attractive interaction are a freeamplitude lower than 20 nm and in mostcases a setpoint ratio of 0.9; furthermore,the phase shift depends on the cantilever’sQ factor and spring constant k: �� ∼ Q

and 1/k.The above-mentioned phase shift, which

is due to the influence of long-range forces(electric, magnetic fields), can be bestdetected in the so-called lift mode (DigitalInstruments, 1993). In this mode, one linewill be scanned twice (Fig. 12.12). After thefirst pass, which provides the topography,the tip will be lifted a certain value, andthen in the second pass it will follow thetopography in a constant distance, with onlythe long-range forces acting to the tip. Fordetecting electric fields, it is possible to applyvoltage to the tip; for detecting magnetic

fields, special magnetic tips are necessary.The second pass is performed without truematerial contact, like a noncontact regime.Under ambient conditions this noncontactmode can only be performed for very long-ranging forces (>50 nm), because otherwisenear to the surface the fluid layer will formmaterial contact by liquid bridge formationincluding action of strong attractive capillaryforces to the tip.

12.2.3. Cantilevers and Probes

As mentioned at the beginning of the chapter,a very fine tip mounted on a cantilever isthe central part of the SPM. It embodies thesensing tool for imaging and spectroscopy.Because of that importance a wide variety ofcantilevers and probes exist. For many exper-iments, however, the choice of cantilever andtip is absolutely crucial for the success of theexperiments.

Most commercially available cantileversare made of silicon nitride or silicon. Gen-erally, silicon nitride cantilevers are softercompared with those made of silicon. Fur-thermore, V-shaped and beam-shaped can-tilevers are the two major constructions(Fig. 12.13). The material properties and themeasures of the cantilever provide two essen-tial parameters for SPM: (1) The force con-stant k and (2) the Q factor (quality factor).

Figure 12.12. Principle of the lift mode.

Page 216: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

IMAGING SOFT SURFACES: PHYSICS AND METHODS 211

+ +

Figure 12.13. Sketch of the two different basic con-struction forms of cantilevers: (Left) Beam-shaped and(right) V-shaped. The tip is placed in the center of theturning point of the V-shaped cantilever or near the edgeof the beam-shaped cantilever.

The force constant is defined by Hooke’s lawas the ratio between the applied force F andthe cantilever deflection �d:

k = F

�d

When low forces should be applied forimaging soft surfaces, cantilevers with lowforce constants should be used. For dynamicmode the resonance frequency (which isrelated to the force constant) and the Q factorof the resonance are important measures. TheQ factor is a measure of the sharpness of theresonance of the cantilever, thus providing aindication about the sensitivity to changes inoscillation of the cantilever due to externalforces.

In addition, cantilevers with a one-sidedmagnetic coating are available, which areused for dynamic mode with a magneticdrive, in contrast to the standard drive bya piezoelectric crystal.

The position of the tip on the cantileveris a further difference between the availablecantilevers. Mostly the tip is placed near theend of the beam-shaped cantilever or in thecenter of the turning point of the V-shapedcantilever (Fig. 12.13). However, there arealso cantilevers available where the tip is atthe very end of the cantilever. That shapeallows for optical control at the exact pointof imaging during the imaging process fromabove, and it allows an easier way to achieveexact positioning, too.

To differentiate between various tips, onehas to look at the aspect ratio and the tipradius that are necessary for the experiments.Standard tips are available with tip radii from10 nm to 50 nm and cone angles around40◦. However, many procedures may be usedto alter these tip characteristics. A tip canbe oxide-sharpened, and a fine carbon tipcan be grown on it by electron beam depo-sition. Furthermore, carbon nanotubes canbe attached to a tip, rendering it very fineindeed (Wong et al., 1998). Larger micro-spheres or beads can be glued to the tip, tocreate a more blunt sensing tool.

Once the geometry of the tip is knownor established, its surface properties canbe further altered to sense different signalsfrom the sample surface. An electric fieldcan be applied to a conducting tip tomeasure varying electric surface properties.Capacitance, magnetic, and thermal gradientscan be measured by modified tips. Chemicalmodification of the tip is often used toattach certain molecules and special chemicalgroups to the tip, or just alter the surfacetension of the tip.

Before an experiment and while imag-ing, one should take care about the shape ofthe tip. Contamination can result in doubletip images or reduction in lateral resolution.There are a few good ways to clean tips;mostly by oxidation of the organic layerson the tip by UV light, or by using oxi-dizing acid solutions. Other cleaning meth-ods include the use of oxygen or argonplasma (Luginbuhl et al., 2000) or carbondioxide snow (Hills, 1995).

For biological applications such as theimaging of biomolecules, cells, tissues, andbiomaterials, the choice of cantilevers andprobes may be summarized as follows.Silicon tips are often used in dynamic modein air. Silicon nitride tips or very soft silicontips are used in contact and dynamic modein liquids. For imaging biomolecules, mostlysharp tips are used, while for imaging livingcells blunt tips should chosen.

Page 217: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

212 IMAGING SOFT SURFACES BY SFM

12.3. EXAMPLES AND SPECIALPROBLEMS

12.3.1. Liquid Droplets

This section provides an example showingsome basic principles that are importantfor imaging soft liquid-like surfaces orsurfaces with a liquid layer on top. Itspurpose is to show the high sensitivity ofthe scanning force microscope, as well asdescribe certain precautions necessary in thisspecial application. The principles explainedin this section are also important whenimaging biological structures in air, becausethese are mostly very soft and either containa lot of water or have a water layer on top.

In 1996, imaging of high-viscosity liq-uids like glycerin (Tamayo and Garcia, 1996)and dendrimers (Sheiko et al., 1996) wasdemonstrated. Soon thereafter, Herminghauset al. demonstrated the imaging of low-viscosity liquid surfaces with nanometer res-olution (Herminghaus et al., 1997). Now,even water structures could be imagedin investigations on wetting properties onthe nanometer scale (Pompe and Herming-haus, 2000). The basic mechanism of thisimaging technique may perhaps best beexplained in the model by Fery et al. (1999)since it provides useful insights in theimaging of liquid and liquid-like surfaces(Fig. 12.14).

For imaging liquid surfaces, the intermit-tent contact mode is used, meaning that aoscillating tip is scanning the surface. Thedamping of the oscillation provides the feed-back signal. The interaction between tip andliquid surface occurs via the intermittent for-mation of a very small liquid neck, which isillustrated in Fig. 12.14. During every oscil-lation cycle (at a frequency of approximately300 kHz), the intermittent formation and dis-ruption of the small liquid neck occur. At theturning point of the oscillation cycle the tipjust touches the surface. By moving awayfrom the surface, a nanometer-sized liquidneck is formed for only a short period of timein the oscillation cycle. During this time anattractive capillary force is acting on the tip.When the tip moves further away, the liq-uid neck becomes energetically unstable anddisruption occurs. The difference in surfaceenergy of the formed liquid neck is thoughtto be the amount of dissipated energy. Thusthe creation of the small liquid neck duringevery oscillation cycle leads to an energydissipation of the oscillating tip–cantileversystem. Hence, the cantilever oscillation isdamped and the damping can be detectedfrom the feedback system.

The force of the nanometer-sized liquidneck is very small and thus the feedbackmechanism has to be very sensitive to detect

tip

liquid(a) (b) (d)(c)

neck

Figure 12.14. Sketch of liquid neck formation and disruption. (A) The tip moves toward the surface. (B) At theturning point of oscillation, it just touches the surface. (C) By moving upward, a nanometer-sized liquid neck isformed. (d) At a certain distance of the tip from the surface, the liquid neck becomes energetically unstable anddisrupts from the tip.

Page 218: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

EXAMPLES AND SPECIAL PROBLEMS 213

the small damping of the cantilever oscilla-tion. From an analysis of the oscillation sig-nals, the power dissipation was determinedby 10−12 W. The model suggested that theliquid neck has a radius of only 4 nm andexhibits an average attractive force of only10−10 N to the tip. From this it is clear thatusually only 2–5% damping of the free oscil-lation amplitude is allowed for the feedback

circuit. When a stronger damping value isset in the feedback system, the tip actuallygoes into the liquid and is damped by viscousfriction. Strong oscillations of the feedbacksystem occur. Imaging of the liquid surfacewith nanometer resolution is no longer pos-sible. The result of these perturbations is thechange of the profile of a liquid droplet asshown in Fig. 12.15.

(a)

(b)

( )

())

()

(

Figure 12.15. (A) Section of an SFM topography image of a hexaethylene glycol droplet. (B) Another profile ofa droplet imaged with stronger damping conditions set in the feedback circuit. For the increased damping of thecantilever the tip has to crash into the liquid. The imaging with high resolution is no longer possible. Strongoscillations of the feedback circuit occur.

Page 219: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

214 IMAGING SOFT SURFACES BY SFM

The very small dissipation energy of thecantilever oscillation is the reason that onlysmall deviations from the optimal imagingconditions can disturb the whole imagingprocess. Therefore, the wetting propertiesof the tip and its geometry are crucialimaging parameters. The model for theimaging process (Fery et al., 1999) verifiedthis importance. It was shown that there isa dependence of the size of the liquid neckformed on the tip radius and on the assumedcontact angle of the measured liquid on thetip surface. For large tip radii and low contactangles, the liquid neck becomes quite large,and this leads to an overly strong force onthe tip, with the tip bent into the liquidsurface. Imaging of liquid surfaces wouldbe not possible under such conditions. Onthe other hand, very small tip radii and veryhigh contact angles lead to very small liquidnecks. However, a very small liquid neckcannot provide enough energy dissipation tothe tip oscillation. In this case, the dampingcould not be detected by the feedback systemand the tip would crash into the liquidsurface, making imaging impossible.

We therefore see that imaging of liquidsurface can only be successfully performedwith certain tip radii and liquids of par-ticular surface tension. In the experimentsconsidered here, droplets of salt solutions,hexaethylene glycol, and water were investi-gated. A typical image of such a micrometer-sized droplet is shown in Fig. 12.16.

Recently it has become possible toincrease the Q factor of the cantileveroscillation in the dynamic mode, as

was mentioned in the previous sectionconcerning modes for imaging soft surfaces.This method is thought to increase theopportunities for and stability of imagingliquid surfaces and liquid-like structures. Theimproved control of imaging in the contextof purely attractive forces and the highersensitivity should allow for better imaging inapplications at the very end of the sensitivityrange of feedback systems in conventionalconfigurations.

12.3.2. Swollen Polymer Surfaces

This section presents examples demonstrat-ing the influence of scan conditions on theimage of soft sample surfaces. AFM in liq-uids is the first choice for imaging soft sam-ples because low contact force is essential.Polymers are excellent materials on whichto practice AFM in liquids, since polymersshow a swelling behavior in water.

At first a blend of 10% poly(acrylic acid)(PAA) and 90% polyamide 6 (PA6) preparedas thin film (thickness 20 nm) on siliconwafer (Steinert, 2000) is examined. PAA isthe actual component that swells in water.Figure 12.17A shows a 3D contact modeimage of the surface scanned in air by acontact mode silicon cantilever; note thatthe surface is relatively flat and less struc-tured. The associated force–distance curveis shown in Fig. 12.17B. In the above-mentioned spectroscopy mode, the tip movesonly in the z direction and the deflectionof the cantilever as a function of �z ismeasured. During the approach (extending),

Figure 12.16. Image of a hexaethylene glycol droplet on a silicon wafer substrate. Due to the artificially patternedwettability of the substrate, the droplet exhibits a distinct curved form especially near the contact line, demonstratingthe high resolution of the imaging technique.

Page 220: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

EXAMPLES AND SPECIAL PROBLEMS 215

µm

2

4

6

8

200.

00 n

m

Figure 12.17. (A) Surface of a 10/90 PAA/PA6 blend, contact mode in air. (B) Associated force–distance curve(explanation in the text).

the contact with the surface is marked bythe beginning of the slope at the left side.During the retracting of the tip, adhesiveforces hold the tip down on the surfacebeyond the contact point until the springforce (F = k�z) equals the adhesion force;hence, the tip jumps out. This adhesion forceresults from the capillary forces between thefluid layers on the surface and on the tip,forces that are typical under ambient condi-tions. The contact force F can be calculatedfrom the distance between the crossing withthe setpoint and the jump-out �z (430 nm)

and the spring constant of the cantileverk (0.05 Nm−1): F = 21.5 nN. To eliminatethe influence of the capillary forces, exper-iments were done in water with differentcontact forces (Fig. 12.18 and 12.19). The“high” contact force (Fig. 12.18B), calcu-lated as the distance between the beginningof the deflection and the setpoint crossing(140 nm), is F = 7 nN. It should be notedthat no adhesive force is detected now dueto the absence of capillary forces. In thiscase the surface shows no significant changes(Fig. 12.18A). Figure 12.19 shows that as

Page 221: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

216 IMAGING SOFT SURFACES BY SFM

µm

2

4

6

8

200.

00 n

m

Figure 12.18. (A) Surface of a 10/90 PAA/PA6 blend, contact mode in water, “high” contact force. (B) Associatedforce–distance curve.

the force was further lowered, a significantchange in roughness and structure is seen(Fig. 12.19A), and the swollen PAA domainsare clearly detected. The contact force is F =2.5 nN (Fig. 12.19B). This example showsthat small changes in the scan conditions canlead to very different results, and thereforesoft surfaces should be scanned at the lowestpossible contact force.

The second example shows that evenforces at contact mode imaging in liquids canbe too high. The sample is a specially pre-pared hydrogel, poly-N -isopropylacrylamid

(PNIPAAm), spin-coated on a Teflon-likefilm on silicon wafer and stabilized by atreatment in Ar+-plasma (Nitschke, 2001).The thickness of the dry film was 10 nm;the thickness of the swollen film (water)was 30 nm. AFM was done in water.Figure 12.20 juxtaposes the results in contactmode (left) and in tapping mode (right). Bothimages of the swollen state show a net-likestructure, which in the case of contact modeis a slightly disturbed, such that the scanningtip relocates the highly flexible structure. Thetapping mode image reveals a finer network

Page 222: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

EXAMPLES AND SPECIAL PROBLEMS 217

µm

2

4

6

8

200.

00 n

m

Figure 12.19. (A) Surface of a 10/90 PAA/PA6 blend, contact mode in water, “low” contact force. (B) Associatedforce–distance curve.

Figure 12.20. Surface of a PNIPAAm film, scanned in water. (Left) Contact mode. (Right) Tapping mode.

Page 223: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

218 IMAGING SOFT SURFACES BY SFM

structure of about 30 nm. In contact mode,this structure was smeared out by the tip.In tapping mode, only the contact force wassmall enough not to disturb these structures.

12.4. CONCLUSIONS

This chapter presented an outline of thebasics of AFM and its application forimaging soft surfaces. Some technical detailsrelevant for investigations of sensitive, soft,and flexible surfaces were described. Thepurpose of the chapter is to show that AFMis a very powerful tool for the examinationof soft surfaces with very high resolutionand under ambient conditions. The softnessof biological structures, however, requireskeen awareness of certain precautions andtechnical details. Due to its adaptabilityto living culture conditions, AFM is anexcellent instrument for the imaging of livingtissues and for spectroscopy of biological,chemical, and physical characteristics of cellsand biomolecules.

REFERENCES

Anczykowski, B., Cleveland, J. P., Kruger, D.,Elings, V. B., and Fuchs, H. (1998). Analysisof the interaction mechanisms in dynamic modeSFM by means of experimental data and com-puter simulation. Appl. Phys. A66:885–889.

Binning, G., Quate, C., and Gerber, C. (1986).Atomic force microscope. Phys. Rev. Lett.56(9):930–933.

Digital Instruments (1993). NanoScope III Mul-timode Scanning Probe Microscope InstructionManual, Version 1.5.

Fery, A., Pompe, T., and Herminghaus, S. (1999).Nanometer resolution of liquid surface topog-raphy by scanning force microscopy. J. Adhes.Sci. Technol. 13(10):1071–1083.

Herminghaus, S., Fery, A., and Reim, D. (1997).Imaging of droplet of aqueous solutions by Tap-pingMode Scanning Force. Ultramicroscopy69(3):211–217.

Hills, M. M. (1995). Carbon dioxide jet spraycleaning molecular contaminants, Vacuum Sci.A13(1):30–34.

Humphris, A. D. L., Tamayo, J., and Miles, M. J.(2000). Active quality factor control inliquids for force spectroscopy. Langmuir16:7891–7894.

Luginbuhl, R., Szuchmacher, A., Garrison, M.D., Lhoest, J. B., Overney, R. M., and Rat-ner, B. D. (2000). Comprehensive surface anal-ysis of hydrophobically functionalized SFMtips. Ultramicroscopy 82:171–179.

Magonov, S. N., Elings, V., and Whangbo, M.-H. (1997). Phase imaging and stiffness intapping-mode atomic force microscopy. Surf.Sci. Lett. 375:L385–L391.

Magonov, S. N., Cleveland, J., Elings, V., Den-ley, D., and Whangbo, M.-H. (1997). Tapping-mode atomic force microscopy study ofthe near-surface composition of a styrene-butadiene-styrene triblock copolymer film.Surf. Sci. 389(1–3):201–211.

Nitschke, M. (2001). Institut fur Polymer-forschung Dresden, Germany, private commu-nication.

Ohnesorge, F., and Binnig, G. (1993). Trueatomic resolution by atomic force microscopythrough repulsive and attractive forces. Science260:1451–1456.

Pompe, T., and Herminghaus, S. (2000). Three-phase contact line energetics from nanoscaleliquid surface topographies, Phys. Rev. Lett.85(9):1930–1933.

Sheiko, S. S., Eckert, G., Ignateva, G., Muza-farov, A. M., Spickermann, J., Rader, H. J.,and Moller, M. (1996). Solid-like states of adendrimer liquid displayed by scanning force.Macromol. Rapid Commun. 17(5):283–297.

Spatz, J. P., Sheiko, S., Moller, M., Winkler, R.G., Reineker, P., and Marti, O. (1995). Nan-otechnology 6:40–44.

Steinert, V. (2000). Institut fur PolymerforschungDresden, Germany, private communication.

Tamayo, J., and R. Garcia. (1996). Deformation,contact time, and phase contrast in tappingmode scanning force microscopy. Langmuir12(18):4430–4435.

Page 224: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 219

Tsukruk, V. V. (1997). Scanning probe microscopyof polymer surfaces. Rubber Chem. Technol.70:430–467.

Winkler, R. G., Spatz, J. P., Sheiko, S., Moller, M.,Reineker, P., and Marti, O. (1996). Phys. Rev.B54:8908–8912.

Wong, S. S., Joselevich, E., Woolley, A. T., Che-ung, C. L., and Lieber, C. M. (1998). Covalentlyfunctionalized nanotube nanometresized probes inchemistry and biology. Nature 394:52–55.

Zhong, Q., Jennis, D., Kjoller, K., and Elings,V. B. (1993). Surf. Sci. Lett. 290:688.

Page 225: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

CHAPTER 13

HIGH-SPEED ATOMIC FORCE MICROSCOPYOF BIOMOLECULES IN MOTIONTILMAN E. SCHAFFERCenter for Nanotechnology and Institute of Physics, Westfalische Wilhelms-Universitat Munster,Gievenbecker Weg 11, 48149 Munster, Germany

13.1. INTRODUCTION

“Time is the Life of the Soul,” H. W. Longfel-low wrote in Hyperion (Longfellow, 1839).With regard to atomic force microscopy(AFM) in biology and medicine, “Time is theSoul of Life” may hold a truth of its own. Lifeembraces motion, development, and growth:the progression of events with time. Such aprogression cannot be captured in a single“snapshot” AFM image. The “soul” of life isinstead revealed in image sequences. This iswhy fast and time-resolved AFM imaging isof particular importance in the study of bio-logical systems.

Soon after its invention, AFM (Binniget al., 1986) was applied to the investigationof biomolecules in solution (Drake et al.,1989). Its capability of imaging with molec-ular and even submolecular resolution innumerous environmental conditions includ-ing biological buffer solutions distinguishesit from other high-resolution techniques thatare used to study biomolecules (e.g., x-raydiffraction, nuclear magnetic resonance, cryo-electron microscopy). As compared to thesecomplementary techniques, little sample pre-paration is required and the sample can beimaged in its native environment over and

over again, allowing for the recording ofimage sequences.

In an AFM, a sharp tip is mounted ontothe free end of a flexible cantilever. Thetip is brought into contact with a samplesurface and is scanned across it. Interac-tion forces between the tip and the sam-ple cause the tip and thus the cantilever todeflect. This deflection is detected by focus-ing an optical beam onto the cantilever anddirecting the beam reflected from it onto aposition-sensitive detector. This optical beamdeflection (“optical lever”) detection methodallows the remote and noninvasive detec-tion of tip deflections in biological (trans-parent) buffer solution with sub-angstromaccuracy.

A key improvement to the imaging of softsamples in liquid was made by the develop-ment of “tapping mode” in liquid (Hansmaet al., 1994; Putman et al., 1994). In tappingmode, the cantilever is externally driven suchthat the tip undergoes vertical sinusoidal oscil-lations. Traditionally, the external drive in liq-uid is of acoustic nature (Schaffer et al., 1996),but the cantilever can also be driven by mag-netic (Han et al., 1997; Revenko and Proksch,2000) or thermal (Marti et al., 1992) forces.Instead of being dragged over the sample as

Force Microscopy: Applications in Biology and Medicine, edited by Bhanu P. Jena and J.K. Heinrich Horber.Copyright 2006 John Wiley & Sons, Inc.

221

Page 226: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

222 HIGH-SPEED ATOMIC FORCE MICROSCOPY OF BIOMOLECULES IN MOTION

in contact mode, the tip now simply “taps”the surface once during each oscillation cycle.The oscillation amplitude is used as input to afeedback loop keeping the mean tip–sampleseparation constant. This mode exhibits lowshear forces and has enabled the destruction-free imaging of soft biological samples suchas single molecules on a routine basis (Fritzet al., 1995; Hansma, 2001).

While the spatial resolution that can beobtained on biomolecules might be higherwhen imaging in gaseous media, in vac-uum, or at cryogenic temperatures (Shao andZhang, 1996), dynamic processes involvingbiomolecules generally occur only in liq-uid environments. Therefore, this chapterfocuses on the imaging of dynamic processesin liquids. Although AFM imaging in liquidsis still considered somewhat of an “art” dueto its technical difficulty, much progress hasbeen made in the investigation of biologicalprocesses. Some examples of studies that areconcerned with time-resolved AFM imagingof biomolecular interactions are now given.

One of the first studies of dynamic pro-tein–DNA interactions was the observationof enzymatic degradation of DNA by DNaseI (Bezanilla et al., 1994), demonstrating thatthe AFM can image dynamic interactions ona molecular level. Endonucleolytic cleavageof DNA by the restriction enzyme EcoKIwas imaged in the presence of ATP (Elliset al., 1999). Transcription of double-strandedDNA by RNA polymerase (RNAP) wasdetected after adding nucleoside triphosphates(NTPs) to stalled DNA–RNAP complexesadsorbed onto mica, and transcription ratesof 0.5–2 bases/s were observed (Kasas et al.,1997; Guthold et al., 1999). Nonspecific pho-tolyase–DNA complexes were imaged, show-ing association, dissociation, and movementof photolyase along the DNA, suggestingthat photolyase uses a sliding mechanism toscan the DNA for damaged sites (van Noortet al., 1998). Dynamic interactions of thetumor-suppressor protein p53 with DNA wereobserved by time-lapse AFM imaging, reveal-ing dissociation/reassociation, sliding, and

possibly direct binding to the specific bindingsite, whereby two different modes of targetrecognition were detected (Jiao et al., 2001)(discussed in more detail in Section 13.2.2).

Protein–protein interactions were visual-ized by imaging the association and dis-sociation of the chaperonin GroEL–GroEScomplex, the lifetime of which was deter-mined as ≈ 5 s (Viani et al., 2000). Pro-tein–lipid interactions were visualized byimaging the degradation (hydrolysis) ofmembrane phospholipids by phospholipaseA2 with a lateral resolution of less than10 nm (Grandbois et al., 1998). The inter-actions of proteins with an inorganic crys-tal were investigated by adsorbing solubleproteins extracted from abalone nacre ontoa growing calcite crystal. Dependent on theircrystallographic orientation, the atomic stepedges changed in shape and growth speedas the proteins attached to the crystal sur-face (Walters et al., 1997a).

Enzyme activity was directly observed bymeasuring the height fluctuations of the pro-tein lysozyme as it was exposed to an oligo-glycoside substrate (Radmacher et al., 1994a).Dynamic movements of the arms of laminin-1 that might contribute to the diversity oflaminin functions were observed (Chen et al.,1998). Calcium-mediated structural changesof native nuclear pore complexes such as theopening and closing of individual nuclear bas-kets were observed, indicating that cytoplas-mic plugging and unplugging of the com-plex is insensitive to calcium in the absenceof ATP (Stoffler et al., 1999). The move-ments of single sodium-driven rotor proteinsfrom a bacterial ATP synthase, embedded ina lipid membrane, were monitored and theprincipal modes of the motion were distin-guished (Muller et al., 2003).

Conformational changes in supercoiledDNA induced by decreasing the ionic strengthof the imaging buffer were imaged (Nagamiet al., 2002). Bidirectional growth of poly-morphic amylin protein fibrils was moni-tored in vitro yielding an elongation rate of1.1 ± 0.5 nm/min (Goldsbury et al., 1999).

Page 227: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

TIME-RESOLVED IMAGING 223

The disassembly dynamics of microtubuleswas observed with an average rate of 1–10tubulin monomers per second. The large fluc-tuations in this rate might be caused either bymultiple pathways in the disassembly kineticsor by stalling due to defects in the microtubulelattice (Thomson et al., 2003). The motionof myosin V on mica in buffer solution wasvisualized (Ando et al., 2001). The activationprocess of human platelets could be followed(Fritz et al., 1993), and dynamic changes inthe stiffness of the cortex of adherent cul-tured cells were tracked during cell division(Matzke et al., 2001).

One of the main limitations common tothese investigations is the limited time res-olution of the imaging. In general, an AFMimage requires on the order of minutes to beacquired, and processes occurring faster thanthat cannot be investigated with conventionalAFM imaging. This chapter discusses someof the topics and issues associated with fastAFM imaging of biomolecules in motion.

13.2. TIME-RESOLVED IMAGING

13.2.1. Recording Image Sequences

Real-time biological processes can be inves-tigated by recording AFM image sequences.In principle, this amounts to no more thanscanning the same image area over and overagain. In practice, however, there are a num-ber of issues that become important whenrecording image sequences. The most obvi-ous issue is that the time needed to acquire animage sequence is proportional to the num-ber of images in the sequence. For sequenceswith a lot of images, the total duration ofimaging can amount to many minutes or evenhours. Drift effects therefore become espe-cially significant.

There are several kinds of drift issues thatneed to be considered. Mechanical drift canbe grouped into two categories: (a) lateraldrift and (b) vertical drift. (a) Lateral driftcan arise from two different effects. On theone hand, drift in the microscope (due to

thermal equilibration processes or due tocreep in the piezoelectric scanner) causesthe image area to slowly shift in time. Onthe other hand, the object of interest mightmove itself, driven for example by ther-mal diffusion or by conversion of energy.Both effects have the same consequence:moving the object of interest out of theimage area. Section 13.2.3 presents a methodfor (partially) compensating for lateral driftby aligning the images after the recording.(b) Vertical drift can be caused by micro-scope drift, too, which moves the cantileversupport relative to the sample or by driftin the cantilever deflection that moves thetip relative to the cantilever support. Driftin cantilever deflection might be caused byslow thermal bending of the cantilever due totemperature variations in the imaging envi-ronment, or by relaxation of the internalstress in the cantilever material. While verti-cal microscope drift is usually taken care ofby the feedback loop that keeps the cantileverdeflection or amplitude constant, the drift incantilever deflection causes the optical beamto move on the detector. This causes a prob-lem in both contact and tapping mode opera-tion: In contact mode, such a deflection isinterpreted as a change in imaging force.Such a change causes the feedback loop todecrease the tip–sample distance, resultingin increased imaging forces that can damagethe sample, or it causes the feedback loopto increase the tip–sample distance, whichcan result in a failure of accurate surfacetracking. In tapping mode, cantilever drift isless critical since the cantilever oscillationamplitude and not the cantilever deflectionis used as error signal in the feedback loop,and the (ac) amplitude is effected less by(dc) drift. But drift in cantilever deflection,which causes the optical beam to move onthe detector, results in a situation where thebeam is not centered on the detector anymore, causing additional detection noise inboth contact and tapping mode.

There are yet some other issues to con-sider when recording image sequences. For

Page 228: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

224 HIGH-SPEED ATOMIC FORCE MICROSCOPY OF BIOMOLECULES IN MOTION

example, the quality of the scanning tip candegrade over time, especially when the tip iscontinually being contaminated with organicdebris. And, last but not least, a fragile sam-ple might not “enjoy” being imaged over andover again.

13.2.2. Dynamic Interactions of p53with DNA

As an example of time-resolved imaging,dynamic interactions of p53 with DNA arenow discussed. p53 is a tumor-suppressorprotein that is involved in the regulation ofcell growth (Levine, 1997). Over 50% of allhuman cancers (e.g., skin, colon, and breastcancer) are related to nonfunctional p53. Thename, p53, stands for “p” as in protein andfor “53” as in 53 kD, the molecular weight ofthe protein. p53 has multiple functions, butmostly it serves as a transcription factor. Insolution, it forms tetramers that bind specifi-cally to the consensus binding site consistingof two decameric consensus units that areseparated by 0–21 base pairs. It also binds

nonspecifically to DNA with a lower affinity.Since most mutations that render the pro-tein dysfunctional occur in its DNA-bindingregion, it is important to study the inter-actions of p53 with DNA. The AFM is avaluable tool in this endeavor, since it canimage single molecules in solution at molec-ular resolution. Both p53 and DNA need tobe attached to an underlying imaging sur-face. Mica is usually chosen as the imagingsurface for high-resolution imaging, since itis atomically flat and can easily be cleaved.One of the difficulties of using mica as theimaging surface for DNA is that both micaand DNA are negatively charged for typicalbuffer conditions. A method that promotesbinding of DNA to mica is adding divalentcations to the solution that act as molecu-lar crosslinkers (Fig. 13.1). But care must betaken to add just the right concentration ofdivalent cations: Too little does not attachDNA tightly enough for stable imaging bythe AFM tip. Too much binds DNA downtoo tightly so that it might not be able tomove and interact with p53. It was found that

Figure 13.1. Schematic illustrating a difficulty when imaging dynamic interactions involving negatively chargedbiomolecules such as DNA. The negative charge of both DNA and the underlying mica surface usually preventsadsorption of DNA to the surface. Divalent cations such as magnesium are therefore commonly used as molecular“glue” between DNA and mica. With increasing concentration of divalent cations, the repulsive force between DNAand mica is reverted and becomes attractive, allowing DNA to adsorb to the mica surface. The difficulty is findingthe right concentration: Too little attaches DNA to the surface too weakly for AFM imaging, while too much bindsthe DNA too tightly to the surface so that it might not be able to move and participate in dynamic interactions.

Page 229: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

TIME-RESOLVED IMAGING 225

a divalent cation concentration of 20 mMMg2+ in the presence of Na-HEPES allowedboth DNA and p53 to loosely attach to micaso that (a) they still interact both specificallyand nonspecifically (however with a reducedaffinity) and (b) their interactions could beimaged by the AFM (Jiao et al., 2001). Sev-eral different types of p53–DNA interactionevents could be imaged, including dissocia-tion/reassociation and sliding (Fig. 13.2).

One of the issues that this study addressedis the particular target recognition modewith which p53 binds to DNA. The ques-tion is, How does a protein find its targetsite? In the simplest mode, a protein dif-fuses three-dimensionally in solution until itbinds to the target site directly from solu-tion (Fig. 13.3a). It was, however, shownfor a number of proteins other than p53

that such a binding mode can not accountfor the relatively high binding rates thatwere experimentally observed for those pro-teins. A second mode has therefore beenproposed that relies on a two-step process:Initially, the protein binds nonspecifically toDNA and then subsequently diffuses one-dimensionally along the DNA until it findsthe target site (Fig. 13.3b). Both modes oftarget recognition were detected in the inter-actions of p53 with DNA (Jiao et al., 2001).

13.2.3. Image Alignment

After an AFM image sequence of some pro-cess is recorded, it needs to be analyzed withthe goal of extracting information about theprocess. A fruitful way to display the imagesequence is to animate it, to “make a movie.”

Figure 13.2. Dynamic p53–DNA interactions observed by time-resolved tapping mode AFM imaging in solution.The buffer conditions were adjusted such that both p53 protein and DNA are weakly adsorbed to the micasurface. (a) A p53 protein molecule (arrow) is bound to a DNA fragment. The protein (b) dissociates from andthen (c) reassociates with the DNA fragment. (d) A downward movement of the DNA with respect to the proteinoccurs, constituting a “sliding” event whereby the protein changes its position on the DNA. Image size: 620 nm.Grayscale (height) range: 4 nm. Time units: minutes, seconds. [Reprinted from Jiao et al. (2001), with permissionfrom Elsevier.]

Page 230: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

226 HIGH-SPEED ATOMIC FORCE MICROSCOPY OF BIOMOLECULES IN MOTION

Figure 13.3. Schematic illustrating different protein–DNA interaction modes. (a) The protein diffuses three-dimen-sionally in solution until it comes close to the specific interaction target site to which it binds. The protein–DNAbinding rate in this mode is low because the protein is “unguided” in its search. (b) An alternative binding modeconsists of a two-step process: In the first step, the protein binds nonspecifically anywhere to the DNA. In thesecond step, it undergoes one-dimensional diffusion along the DNA, involving macroscopic dissociation/reassociation(“jumping”), microscopic dissociation/reassociation (“hopping”), intersegment transfer, and sliding, until it finds thetarget site to which it binds specifically. This interaction mode results in higher binding rates because the proteindiffuses in a reduced (one-dimensional) space.

One of the problems of animating an imagesequence is that the features in consecutiveimages are often not aligned with respect toeach other. For example, thermal drift causesthe imaging area to shift in position, caus-ing the features in the animation to “jump”around. The simplest solution to this prob-lem is to align the images “by hand”—thatis, to manually offset consecutive imagessuch that the features of choice overlap asbest as possible. This certainly is a possibil-ity, but becomes tedious when the sequenceconsists of hundreds of images. In the casethat the features in the image sequence onlychange slightly between images, it is pos-sible to align consecutive images automati-cally by using cross-correlation (van Noortet al., 1999). The cross-correlation of twoconsecutive images (Imagen−1 and Imagen)is defined as

Corr[Imagen−1, Imagen](p, q)

=∑i,j

Imagen−1(i + p, j + q)

Imagen(i, j) (13.1)

where the parentheses contain the pixelcoordinates of the respective image. Equation(13.1) can be expressed in terms of

discrete Fourier transforms, F, using thecorrelation theorem (yielding computationaladvantages):

Corr[Imagen−1, Imagen]

= F[F[Imagen−1]

(F[Imagen])∗] (13.2)

The asterisk denotes the complex conjugateand F denotes the inverse discrete Fouriertransform. To increase the accuracy of thecross-correlation alignment procedure, theimages are typically background-subtractedbefore applying Eq. (13.2). Apart from pro-ducing smooth movies, the cross-correlationalignment technique can be used to furtheranalyze the motion of the object of inter-est, such as the lateral diffusion of DNAfragments in buffer solution that are weaklyattached to a mica surface (Jiao et al., 2001).Figure 13.4a shows one of 30 images in theimage sequence. A typical cross-correlationbetween two consecutive images exhibitsa well-pronounced peak (Fig. 13.4b). Theposition of this peak can easily be deter-mined and directly gives the offset by whichthe two images need to be shifted withrespect to each other. Repeating this proce-dure for all consecutive image pairs yields

Page 231: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

TIME-RESOLVED IMAGING 227

Figure 13.4. Image alignment by cross-correlation of consecutive images. (a) One of the original images in theimage sequence: DNA fragments that are weakly attached to a mica surface. (b) Typical cross-correlation of twoconsecutive images. The cross-correlation shows a clearly defined peak that is used to determine the lateral offsetbetween the consecutive images and to subsequently align them. (c) Average of 30 aligned images. Some parts ofthe individual DNA fragments exhibit a high mobility and appear as fuzzy areas, whereas other parts are immobileand appear as bright areas. (d) Same as (c), but the images were binarized and further processed prior to averagingin order to reduce background noise and to more clearly reveal the DNA motion. Image size: 2.3 µm. Grayscale(height) range: 4 nm. [Parts a and c are reprinted from Jiao et al. (2001), with permission from Elsevier.]

a well-aligned image sequence, allowing forfurther analysis. For example, by averag-ing all 30 aligned images in the sequence(Fig. 13.4c), it is possible to pinpoint areasof high and low DNA mobility: The mobilesections appear fuzzy while the immobileones appear bright. It is interesting to notethat the DNA fragments seem to be subjectedto segmented motion that affect only parts ofthe molecule, probably caused by local vari-ations on the mica surface (van Noort et al.,1998; Jiao et al., 2001). Using binary thresh-olding and morphology techniques, the noisein the images can be reduced, and the motionof the DNA fragments can be seen evenclearer in the averaged image (Fig. 13.4d).

While the cross-correlation alignmentmethod generally works well for samples

that exhibit well-defined features on a flatbackground, there are some cases where thealignment algorithm is thrown off. This canhappen, for example, when the object movestoo much between images, or when imagedistortion or scan errors (e.g., erroneous scanlines) build up. Also, since this alignmentalgorithm considers all features in the imagewith equal weight, it might “lock on” toprominent but irrelevant image features.

13.2.4. AFM Images are ‘‘Movies’’by Themselves

When viewing an AFM image, one istempted to consider it as a momentary“snapshot” of the sample. Even a single AFMimage, however, is a movie by itself in the

Page 232: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

228 HIGH-SPEED ATOMIC FORCE MICROSCOPY OF BIOMOLECULES IN MOTION

sense that the scanning tip probes differentareas on the sample at different times. Inthe case of a typical rectangular raster scan,the pixel with pixel coordinates (p, q) isacquired at the time

t (p, q) = 1

finq

(p

2np

+ q

)(13.3)

where p and q are the (zero-based) pixelcoordinates in the fast and slow scan direc-tion, respectively, np and nq are the numberof pixels in the direction of p and q, respec-tively, and fi is the image (frame) scan rate.This assumes that the tip velocity is con-stant and that each scan line is scanned oncein the forward direction (“trace”) and oncein the backward direction (“retrace”). Thelast pixel in an image might be acquiredminutes after the first pixel was. An AFMimage should therefore rather be consideredas a sequence of “pixel snapshots” with apixel acquisition rate of fp = 2npnqfi . Thismechanism is contrary to that of an imageacquired by a CCD camera in optical widefield microscopy, for example. In such acamera, there is a shutter that exposes thewhole CCD chip to the light from the sam-ple at the same time. All pixels are thereforeacquired simultaneously. (The readout of theindividual pixels occurs in a sequential fash-ion, but this is solely a technical issue.)

There are a number of consequencesof this sequential nature of single AFMimages. Thermal drift in the microscopethat moves the scanning tip relative tothe sample might cause distortion in theimage, especially when the sample is scannedslowly. Furthermore, in the case where asample object moves or changes its shapeduring image acquisition, the object mightappear distorted even in the absence ofthermal drift. Motion of biomolecules is notuncommon: Certain proteins, for example,change their configuration as they hydrolyzeATP. In general, an object appears distortedwhen its motion occurs on a timescalethat is comparable to or faster than the

timescale with which the AFM image isbeing acquired. The basic properties ofsuch an imaging artifact can be understoodwith the help of a simple model (Kasaset al., 2000). An object that is modeledas an oscillating half-sphere, for example,appears with horizontal stripes in the image(Fig. 13.5). In the ideal case, the AFMwould acquire images fast enough so thatthe moving object can be assumed stationaryin each image resulting in a sequence ofundistorted images.

13.3. INCREASING THE IMAGINGSPEED

The time resolution in AFM investigations ofbiological processes is limited by the speedwith which image data can be acquired.For biological samples in liquid, a typicalimage rate corresponds to one image every1–10 min. Bottlenecks in the path to fasterimaging are manifold: the limited detec-tion bandwidth of the sensing cantilever,the limited actuation bandwidth of the scan-ning piezoelectric elements, and the limitedbandwidth of the data acquisition system, toname just a few. There are multiple ways bywhich the imaging speed can be increased.One possibility is to modify the AFM hard-ware configuration—for example, by pro-viding compatibility with small cantilevers(Section 13.4). It is possible, however, toscan faster with existing commercial AFMsby employing smart scanning software. Such“indirect” ways are discussed in this section.

13.3.1. Image Tracking

When imaging an object that exhibits lateralmobility, the object might move out of theimage area quickly. The object might alsobe lost when there is a large thermal sampledrift. In order to keep the object within theimage area during a long period of time, itis therefore necessary to scan an area muchlarger than the area that is initially occupiedby the object. If the motion of the object

Page 233: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

INCREASING THE IMAGING SPEED 229

Figure 13.5. Simulation of a moving protein in an AFM. The protein is modeled as a half-sphere that undergoesperiodic oscillations in its radius with an amplitude of 20% of the stationary radius and with a frequency of varyingmultiples, f , of the AFM image frame rate. (a) Stationary protein (f = 0). There are no imaging artifacts. (b) f = 3(i.e., the protein undergoes three cycles while the image is being acquired). The outlines of the protein appear curved.(c) f = 10. About 10 fairly defined horizontal stripes appear, each one representing a protein cycle. The proteinoscillation frequency can therefore be inferred by simply counting the horizontal stripes. (d) f = 27. This frequencyis higher than the “Nyquist frequency” that may be defined as half the number of pixels in vertical direction thatcover the protein (≈ 50/2 = 25). “Undersampling” therefore occurs, and it no longer is possible to uniquely infer theprotein oscillation frequency from the number of horizontal stripes in the image. The protein oscillation frequencyis so high that the protein appears as a fuzzy sphere. Moire patterns appear in the simulation as a consequence ofthe perfect periodicity of the protein oscillation. These simulations are based on the assumption that the AFM tipdoes not interfere with the moving protein.

could be accounted for, however, a smallerscan area could be selected which wouldincrease the image rate significantly.

Van Noort et al. (1999) devised a methodby which the sample object is tracked andthus kept in the center of the image at alltimes. They used this method to determinethe lateral diffusion coefficient of DNA frag-ments on a surface. The method is based on areal-time drift correction algorithm using thesame cross-correlation image alignment pro-cedure that was discussed in Section 13.2.3:After an image is acquired, it is comparedto the preceding image by cross-correlationby which the lateral shift of the object

is determined. The voltages that drive thelateral scanner are then offset on-line to shiftthe scan area such that the object is keptin the center of the image. A small scanarea therefore suffices to image the movingobject, resulting in higher image rates.

13.3.2. Reducing the Numberof Lateral Dimensions

AFM images usually are quadratic in shape.The features of interest in an image, how-ever, might be distributed over just a fewscan lines. It might therefore suffice to reducethe number of scan lines and scan a smaller

Page 234: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

230 HIGH-SPEED ATOMIC FORCE MICROSCOPY OF BIOMOLECULES IN MOTION

area with a higher image rate. For example,one might be interested in how the heightof single molecules that are distributed overa sample surface changes with time. Then itmight not be necessary to image a large sam-ple area with a lot of molecules. Instead, itmight be adequate to scan even just one lineon the sample over and over again, therebyeffectively reducing the number of lateraldimensions from two to one. A time seriesof cross sections is thus created off whichthe height of the molecules can be read.This method was utilized in investigationsof GroEL/GroES chaperonin proteins (Vianiet al., 2000). GroEL and GroES form a com-plex in order to help other proteins fold intotheir native state. By scanning in one lateraldimension only, it was possible to record pro-tein “tubes,” thereby resolving the associa-tion and dissociation of individual complexes(Fig. 13.6).

An imaging technique that especially ben-efits from a reduced number of lateral

dimensions is the so-called “force map-ping” technique, in which thousands offorce spectra are recorded and the imageis reconstructed from a quantitative analy-sis of each of the individual force spectra(Radmacher et al., 1994b; Baselt and Balde-schwieler, 1994; Jiao and Schaffer, 2004).The force mapping technique is capable ofmeasuring the locally resolved adhesion orelasticity of the sample, in addition to thetopography. By limiting the scan area to asingle repetitive line, it was possible to inves-tigate the process of furrow stiffening duringcell division (Matzke et al., 2001).

The reduction in the number of lateraldimensions can be pursued one step furtherby giving up the second lateral dimensionas well and keeping the tip stationary ata fixed lateral position. The tip can thenmonitor the variations in sample height atthat position with an even higher temporalresolution. The motion of a single protein,for example, can be monitored in real time

Figure 13.6. Example where the time resolution of the measurement was increased by reducing the number of lateraldimensions in the AFM image. (a) Single GroEL protein molecules adsorbed to a mica surface in buffer solution.The proteins appear as (bright) rings surrounding the (dark) central channel. Small cantilevers were used to provide ahigh imaging speed. Image size: 300 nm; grayscale range: 15 nm. (b) Motion along the slow (here: horizontal) scanaxis was disabled. The tip thus repeatedly scanned the same (vertical) line, over the same proteins. Those consecutive,one-dimensional vertical line scans were charted next to each other in horizontal direction, effectively generatingprotein “tubes.” Each protein “tube” corresponds to a single protein molecule whose height fluctuations can now bemonitored with a ≈ 250 × higher temporal resolution (≈ 100 ms) than what would be possible via regular imageshaving two spatial dimensions (≈ 25 s). By using protein “tubes,” it was possible to monitor the association anddissociation of the GroEL–GroES complex in the presence of ATP in real time. [Reprinted from Viani et al. (2000),by permission of Nature Publishing Group.]

Page 235: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

INCREASING THE IMAGING SPEED 231

by this approach (Radmacher et al., 1994a).One problem with this approach is that the tipeventually drifts away from its initial lateralposition. For typical globular proteins, forexample, a few nanometers of drift is enoughto move the tip off the protein. To increasethe duration with which the motion of a sin-gle protein can be monitored, a techniquedubbed “protein tracking” has been devel-oped (Thomson et al., 1996). In this tech-nique, lateral feedback is applied to the tip:In certain time intervals, the tip is movedin a single horizontal and subsequently ina single vertical scan line over the protein,whereby the new lateral position of the pro-tein is determined. Then the tip is offset tothis new position and therefore is located ontop of the protein again, where it resumesmonitoring the height variations of the pro-tein. This technique is particularly effectivesince it combines the advantages of reduc-ing the number of lateral dimensions and ofimage tracking.

13.3.3. Exceeding the Feedback Limit

In regular contact or tapping mode imaging,there is a feedback loop trying to keep theso-called error signal (tip deflection in thecase of contact mode and tip amplitude inthe case of tapping mode, subtracted by thesetpoint) zero, thereby keeping the scanningtip at a constant deflection/amplitude abovethe sample surface. If the sample surfaceis scanned at high speeds, the bandwidthof the feedback system might not be largeenough to provide perfect surface tracking.Instead, the error signal deviates further fromzero, reminiscent of varying imaging forces.If the sample is robust enough, however,such that it is not significantly deformed ordamaged by larger imaging forces, the errorsignal can be combined with the feedbacksignal (the voltage applied to the z-scanner)in order to reconstruct a better estimationof the true sample topography (Ookubo andYumotoa, 1999).

13.3.4. Video-Rate Imaging

While the methods that were discussed sofar offer some useful advantages, it seemsdesirable that eventually an AFM couldbe designed that is capable of imagingwith video rate. Video rate correspondsto 25 frames per second and 576 visiblehorizontal lines per frame for the PALvideo standard (29.97 frames per secondand 480 visible horizontal lines per framefor the NTSC video standard, respectively).The uncompressed video data bit rate ison the order of 200 Mbps for 24-bit color.Using digital data compression (MPEG), thedata rate can be reduced by a factor of20–50 down to 4–10 Mbps. In one of the sixrelatively new high-definition TV (HDTV)standards, there are 30 frames per secondand a frame consists of 1920 lines with1080 pixels (“pels”) each. The uncompressedbit rate, ≈1500 Mbps, is significantly higherthan that for the PAL/NTSC standards. Withcompression, however, the HDTV videosignal bit rate can be reduced to about20 Mbps.

An AFM that could image with video ratewould need to scan 25–30 images per sec-ond, each having on the order of 500 × 500pixels with 24-bit resolution each (depend-ing on the particular video standard). Thedata rate would be up to 200 Mbps (≈25Mbytes per second). High-speed data acqui-sition systems are currently being developedapproaching this goal (Fantner et al., 2005;Rost et al., 2005). With such a data rate, a 1-GB hard disk could store only about 40 s ofuncompressed image data. It therefore seemsclear that image compression will play a sig-nificant role in video-rate AFM imaging. Butwhile video-rate compression and data han-dling technology is already developed andcould easily be adapted, there are limitationsof current AFM hardware that need to beovercome before video-rate AFM imagingwill be possible. The next section introducessmall cantilevers as a key toward high-speedimaging of biological samples in liquid.

Page 236: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

232 HIGH-SPEED ATOMIC FORCE MICROSCOPY OF BIOMOLECULES IN MOTION

13.4. SMALL CANTILEVERS

13.4.1. Limits of ConventionalCantilevers

A typical conventional AFM cantilever thatis used for imaging in liquid is made of sil-icon or silicon nitride and is 80–200 µm inlength, 20–40 µm in width, and 0.4–1 µmin thickness. Resonant frequencies of thosecantilevers in air or vacuum are typically10–60 kHz, consistent with the predictionfrom continuum mechanics,

fres∼= 1.02

√E

ρ

T

L2(13.4)

where fres is the resonant frequency of thefirst (fundamental) transverse vibration modein vacuum and E, ρ, T , and L are theYoung’s modulus, the density, the thickness,and the length of the cantilever, respectively.Typical quality (Q) factors are 10–100.Upon immersion in aqueous solution, theresonant frequencies drop by a factor of 3–5to 2–30 kHz. This drop is caused by theadditional mass of the liquid in a boundarylayer around the cantilever that the cantileverneeds to displace during its oscillation. Thedrop in the Q factor is even more dramaticbecause typical Q factors in water are on theorder of one.

Typical imaging speeds in tapping modein liquid with conventional cantilevers are5 min per image consisting of 2 × 5122 pix-els (trace and retrace). This translates to apixel acquisition rate of about 2000 pix-els per second. In tapping mode in liq-uids, the cantilever is typically driven on orbelow its resonant frequency in the liquid.Driving above the resonant frequency—forexample, by utilizing higher-order vibra-tional modes—does not usually allow forstable imaging. Driving below the resonantfrequency, on the other hand, is easily possi-ble since the low Q factors cause the ampli-tude response function of the cantilever tobe approximately flat below the resonant fre-quency. In fact, multiple sharp resonance

peaks in the cantilever oscillation spectrum(“forest of peaks”) that stem from resonancesin the acoustic driving system (Schafferet al., 1996) usually influence the choice ofthe particular tapping frequency. For a typi-cal tapping frequency of 10 kHz, there areabout 5 “taps” per pixel; that is, the tipprobes the sample surface five times before“deciding” which value the pixel should get.Ideally, one tap per pixel should be aboutsufficient, because the Q factor of the can-tilever, a measure of how many oscilla-tions are needed for the cantilever ampli-tude to adjust to a new value, is on theorder of one. Nevertheless, these imagingspeeds are close to the limit of what isphysically possible with conventional can-tilevers.

13.4.2. Increasing the ResonantFrequency of Cantilevers

In order to image faster, cantilevers withhigher resonant frequencies are required.This can be achieved by varying the parame-ters in Eq. (13.4). The material parameters E

and ρ have only a weak (square-root depen-dency) effect on the resonant frequency. Themost direct way of increasing the resonantfrequency is therefore either increasing thethickness or decreasing the length of the can-tilever. An increase in thickness, however,strongly increases the spring constant of thecantilever:

k = E

4W

(T

L

)3

(13.5)

where W is the cantilever width. Largespring constants (k >≈ 1 N/m) are undesir-able for imaging soft samples, since theycause higher imaging forces that more eas-ily destroy the sample. The requirement thatthe spring constant be unchanged upon achange in cantilever dimensions results inthe requirement that T ∝ L (neglecting therather weak influence of the width), andconsequently fres ∝ L−1. Therefore, onlyby a reduction of the cantilever length

Page 237: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

SMALL CANTILEVERS 233

(together with a simultaneous reduction ofthe cantilever thickness) can one obtainhigh-resonant-frequency cantilevers suitablefor the imaging of soft biological samples.It should be noted that if all spatial dimen-sions are reduced simultaneously by the sameratio, the spring constant scales with length,and the resonant frequency scales with recip-rocal length.

Two prominent materials of which smallcantilevers have been fabricated is sil-icon nitride (Reiley et al., 1995; Walterset al., 1996; Viani et al., 1999a) and sili-con (Hosaka et al., 2000). Calculated springconstants [Eq. (13.5)] and resonant fre-quencies [Eq. (13.4)] are displayed forsilicon nitride cantilevers of various dimen-sions (Table 13.1). The resonant frequencyincreases by a factor of over 150 from18 kHz for the longest to 2.9 MHz forthe shortest cantilever. The spring con-stant increases by a factor of ≈ 6 only.It becomes apparent that the smaller thecantilever, the more difficult it is to keepits spring constant small. This is becausethere are several factors that set a lowerlimit to the cantilever thickness. One is oftechnical nature, because thin free-standingstructures generally impose a challenge inmicrofabrication processes. Another one isconnected to the optical detection mechanism

of cantilever deflections: The reflectivity of asilicon or silicon nitride cantilever becomessmall for small thicknesses, especially whenthe cantilever is submerged in a liquid.To increase the reflectivity, the cantilevercould be coated with a thin (10–30 nm)layer of a metal. But a thin silicon/siliconnitride–metal bilayer is highly susceptibleto temperature variations which cause arti-fact deflections of the cantilever (“bimetallicstrip effect”) that are undesirable for imag-ing applications. A solution to this problemis to coat the cantilever on both sides witha metal layer of equal thickness. In eithercase, the mass of the cantilever increasessignificantly, thereby decreasing the resonantfrequency. The bimetallic strip effect canbe minimized by coating the cantilever onan area close to the tip only, whereby cre-ating a “reflector pad” on which the lasercan be positioned (Viani et al., 1999a). Acoating process can be avoided by fabricat-ing the cantilever entirely out of metal inthe first place (Schaffer et al., 1997; Chandet al., 2000).

13.4.3. Properties of SmallCantilevers

Figure 13.7a shows an array of smallaluminum cantilevers. The cantilevers are

TABLE 13.1. Calculated Spring Constants and ResonantFrequencies for Silicon Nitride Cantilevers of VariousSizesa

Length(µm)

Width(µm)

Thickness(µm)

SpringConstant

(N/m)

ResonantFrequency

(kHz)

200 20 0.5 0.020 18.2100 10 0.5 0.078 72.9

20 5 0.1 0.039 36410 2 0.1 0.125 1458

5 2 0.05 0.125 2916

a The calculations were performed using Eqs. (13.4) and (13.5)with E = 250 GPa and ρ = 3100 kg/m3.

Page 238: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

234 HIGH-SPEED ATOMIC FORCE MICROSCOPY OF BIOMOLECULES IN MOTION

2.5 µm wide and 8.8–40 µm long. Theirresonant frequencies are 0.11–2.5 MHz inair (Fig. 13.7b), and they follow a curvethat is well-fitted by the predicted L−2

dependency [Eq. (13.4)]. In water, the res-onant frequencies drop by a factor of 2.5–4,depending on cantilever shape (Hodgeset al., 2001), where this factor is smallerfor higher frequencies (Fig. 13.7b). Still, thesmallest cantilever has a resonant frequencyof about 1 MHz in water. The theory

of Sader (1998) describes the vibrationalproperties of cantilevers in liquids andprovides excellent agreement with the mea-sured values.

One of the difficulties in fabricating smallcantilevers is designing a microfabricationprocess by which integrated tips are pro-duced. There have been successful attemptsto fabricate metal cantilevers with integratedsilicon tips (Chand et al., 2000). Anothermethod is microfabricating tipless cantilevers

Figure 13.7. (a) Scanning electron micrograph of an array of aluminum cantilevers, 8.8–40 µm in length and2.5 µm in width. (b) Resonant frequencies of these cantilevers. [Reprinted with modifications from Schaffer et al.(1997), by permission of the International Society of Optical Engineering.].

Page 239: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

SMALL CANTILEVERS 235

Figure 13.8. Scanning electron micrograph of a tip grown by electron beam deposition (EBD) onto a small cantilever.Such EBD tips have high aspect ratios and small tip radii and are well-suited for the imaging of biomolecules.

first and then manually growing tips onthem in an electron microscope by theelectron beam deposition (EBD) technique(Fig. 13.8). In this technique, the focusedelectron beam deposits material at a selectedlocation on the cantilever, thereby growinga tip (Akama et al., 1990), just like a stalag-mite in a stalactite cavern is grown. To enablelarge-scale production of EBD tips, an auto-mated technique based on pattern recognitionwas developed for the growth of EBD tipson whole wafers of cantilevers (Kindt et al.,2004b).

13.4.4. Thermal Cantilever Noise

Small cantilevers not only have high-speedproperties, but also exhibit a small thermalnoise level. Thermal noise is one of the lim-iting factors in many AFM measurements,and so it is important to utilize the low-noiseproperties of small cantilevers. In order tosimplify the discussion of why the thermalnoise of a cantilever decreases with can-tilever size, the remainder of this section isrestricted to applications of contact-mode-type measurements. For the purpose of thissection, the cantilever is modeled as a sim-ple harmonic oscillator. Such a model is

fairly accurate when the interactions withthe sample surface and with the surroundingmedium are weak. A simple harmonic oscil-lator responds to a periodic excitation force,F (f ), with an oscillation of amplitude

A(f ) = G(f )F (f ) (13.6)

G(f ) is the transfer function of the oscillatorthat gives the (positional) amplitude per unitexcitation force as a function of excitationfrequency:

G(f ) = 1/k√(1 − f 2

f 2res

)2

+(

f

fresQ

)2

(13.7)

Q is the quality factor of the simple harmonicoscillator. It is related to the coefficient ofviscous damping, γ, by

Q = k/(2πfresγ) (13.8)

For an excitation force at dc (f = 0),G(f ) = 1/k, and Eq. (13.6) transforms intoHooke’s law, A = F/k, relating the staticposition of the oscillator to the appliedstatic force. For an excitation force at

Page 240: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

236 HIGH-SPEED ATOMIC FORCE MICROSCOPY OF BIOMOLECULES IN MOTION

the resonant frequency, we have G(f ) =Q/k, and the amplitude response becomesA(fres) = QF(fres)/k, expressing the factthat a weakly (Q � 1) damped oscillatorreacts to forces especially well on resonance.

In thermal equilibrium, a simple harmonicoscillator undergoes Brownian motion. Thismotion can be modeled by assuming arandom thermal excitation force with aspectral force density of

F noise = √4kBT γ (13.9)

representing the so-called Nyquist relation,where kB is the Boltzmann constant andT is the absolute temperature. F noise hasunits of [N/

√Hz] and describes the amount

of thermal excitation force per unit of thesquare root of the bandwidth. This thermalexcitation force density is independent offrequency (“white” spectrum): F noise(f ) =F noise. Just like in Eq. (13.6), the transferfunction G(f ) relates the thermal excitationforce density to the thermal response ampli-tude density: Anoise(f ) = √

4kBT γG(f ). Atfrequencies sufficiently below the resonantfrequency, the transfer function is approxi-mately constant and the total RMS thermalnoise in a given measurement bandwidth,�f , is given by Anoise = √

4kBT γ�f /k. Thesignal-to-noise ratio (SNR) of a measurementof a force signal, F , sufficiently below theresonant frequency therefore becomes

SNR = Asignal

Anoise= F√

4kBT γ�f(13.10)

Since the measurement bandwidth is propor-tional to the measurement speed, it is instruc-tive to rewrite Eq. (13.10) as

SNR√

�f = F√4kBT γ

(13.11)

There are two important consequences ofthis result. First, there is a tradeoff betweenthe signal-to-noise ratio and the measurementbandwidth: Speeding up the measurement bya factor of 2 decreases the signal-to-noise

ratio by a factor of√

2. Second, there are twoways by which the product in Eq. (13.11)can be increased: reducing the temperatureor reducing the coefficient of viscous damp-ing. Since the former is not a viable optionfor most experiments with biomolecules insolution, reducing the coefficient of viscousdamping is the only practical way of gainingeither signal-to-noise ratio (when the speedis fixed) or speed (when the signal-to-noiseratio is fixed). The most direct way of reduc-ing the coefficient of viscous damping isto reduce the size of the cantilever (Vianiet al., 1999a). To demonstrate the advantageof small cantilevers, the amplitude noise den-sity is plotted for two simple harmonic oscil-lators in thermal equilibrium that model twodifferent cantilevers having the same springconstant but different coefficients of viscousdamping (Fig. 13.9). Figure 13.9a shows theamplitude noise density for a simple har-monic oscillator that models a large can-tilever, and Fig. 13.9b shows the amplitudenoise density for a simple harmonic oscilla-tor that models a small cantilever having a10 × higher resonant frequency and a 10 ×lower coefficient of viscous damping. In thegiven measurement bandwidth (0–10 kHz),the thermal RMS amplitude noise is lowerby a factor of

√10 for the small cantilever

than it is for the large cantilever.It should be noted that in Fig. 13.9 we

applied the simple harmonic oscillator modeldespite the fact that the fluid dynamics inthe surrounding medium is more compli-cated, especially when the cantilever experi-ences squeeze-film damping in proximity toa surface (Serry et al., 1995). Nevertheless,the simple harmonic oscillator model quali-tatively describes the vibrational behavior ofsuch cantilevers in a liquid medium and suf-ficed for the purposes of this section. A bettermodel for cantilever vibrations in a viscousmedium, however, does exist (Sader, 1998).

The results of this section give a some-what complementary reasoning with respectto the previous sections of why small can-tilevers perform better than large ones.

Page 241: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

SMALL CANTILEVERS 237

Figure 13.9. Calculated amplitude noise density for alarge and for a small cantilever in thermal equilibrium,based on a simple harmonic oscillator model. Comparedare the respective fundamental vibration modes, bothhaving the same spring constant (k = 0.1 N/m) andtemperature (T = 25◦C), but different coefficients ofviscous damping and resonant frequencies. (a) Largecantilever [γ = 1.59 × 10−6; fres = 10 kHz (Q = 1)].(b) Small cantilever [γ = 1.59 × 10−7; fres = 100 kHz(Q = 1)]. In a given measurement bandwidth (here:0–10 kHz, highlighted area), the thermal amplitudenoise of the small cantilever (≈ 0.51 ARMS) is smallerby a factor of

√10 than that of the large cantilever

(≈ 1.6 ARMS).

Finally, it should be noted that the reducedviscous drag coefficient of smaller can-tilevers also reduces artifact deflections dueto viscous drag effects during scanning. Suchartifact deflections are particularly criticalin force spectroscopy experiments (Rief andGrubmuller, 2002; Viani et al., 1999a, b;Janovjak et al., 2005).

13.4.5. Small Focused Spot Size

Current commercial microscopes that usethe optical beam deflection (“optical lever”)detection method have focused spot diame-ters on the order of 20–50 µm. With suchlarge diameters, not enough light is reflectedoff a cantilever onto the detector when usingcantilevers as small as 5 µm in length and2 µm in width. In order for all (or most) ofthe incident beam power to actually reachthe detector, the incident beam needs tobe focused to the size of the cantilever or

smaller, imposing new design criteria for theconstruction of AFMs for small cantilevers.A basic relationship between the 1/e2 diame-ter of the collimated incident Gaussian beam,d , and the 1/e2 diameter of the focused spot,w0, is given by

w0 = 4λf

πnd(13.12)

where λ is the wavelength (in vacuum) ofthe incident beam, f is the effective focallength of the lens system, and n is the indexof refraction of the medium (Fig. 13.10).For a focused spot diameter of 1–2 µm,an effective numerical aperture (with respectto the 1/e2 –beam diameter) of 0.4–0.2 isrequired when using red light with λ =670 nm. Standard commercial microscopeobjectives easily achieve such numericalapertures and can be used for generatingsmall spots, but their large physical sizemakes them not optimal for use in anAFM setup that should be mechanically asrigid and compact as possible. Using ray-tracing software, a compact lens system withsmall spherical aberrations was designedthat provides a nearly diffraction-limitedspot diameter of about 1.6 µm (Schafferet al., 1997).

There are a number of consequences whenrequiring a small focused spot diameter in anAFM for small cantilevers. A relatively largenumerical aperture, for example, is asso-ciated with a relatively large half-openingangle,

� = arctand

2f(13.13)

of the incident beam (Fig. 13.10). The inci-dent and the reflected beam therefore occupya large angular space. One way of dealingwith this issue is spatially overlapping thebeams and using polarizing optics to separatethem again (Schaffer et al., 1996). Anotherconsequence of having a small focused spotdiameter is that a small diameter causes asmall depth of focus. The depth of focusis the range in propagation direction of the

Page 242: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

238 HIGH-SPEED ATOMIC FORCE MICROSCOPY OF BIOMOLECULES IN MOTION

(a) (b)

Figure 13.10. Diffraction of a focused circular Gaussian beam. (a) Gaussian intensity distribution in the focal plane(z = 0). The focused spot diameter, w0, is defined as the diameter of the circular line in which the intensity is 1/e2

times the maximum intensity. (b) 1/e2 intensity profile along the propagation direction of the beam (z axis). Thebeam does not converge to a point in the focal (xy) plane but remains finite in size (w0 is marked by the two opposinghorizontal arrows). The Rayleigh range, 2zR, is the length in the z direction over which the beam diameter remainsbelow

√2w0 (marked by the two opposing vertical arrows). The larger the half-opening angle of the incident beam,

�, the smaller the focused spot diameter and the smaller the Rayleigh range.

beam in its focus over which the beamdiameter remains approximately collimated.The Rayleigh range, 2zR, serves as a mea-sure for the depth of focus and is definedas the propagation length in which the beamdiameter remains below

√2w0:

2zR = πnw20

2λ(13.14)

The Rayleigh range defines the “diame-ter” of the focused spot in axial direc-tion and depends quadratically on w0. Forw0 = 2 µm, n = 1.33, and λ = 670 nm, theRayleigh range is 2zR ≈ 12 µm. It thereforebecomes important to refocus the incidentbeam onto a newly mounted cantilever on aone-by-one basis, whereby an optical view-ing system with a CCD camera can be ofhelp. It should be mentioned that when usinga laser diode as light source, the incidentbeam is not circular as in Fig. 13.10, butelliptical in shape (unless optical elementsare used to artificially circularize the beam).The consequence of this is that the focusedspot is elliptical in shape, too. This does,

however, not usually cause a problem, aslong as the long axis of the focused spotis aligned along the cantilever length. So itusually is the small diameter of the focusedspot (along the cantilever width) that is criti-cal and relevant in Eqs. (13.12) and (13.14).A design of a compact AFM for smallcantilevers that produces a focused spot of1.6 µm in diameter is shown in Fig. 13.11.

When the focused spot diameter is a sig-nificant fraction of the cantilever length, thedeflected cantilever acts as a curved mirrorfor the incident beam and therefore distortsthe reflected beam. In this case, diffractiontheory needs to be used for a quantita-tive analysis of the detection sensitivity(Schaffer and Hansma, 1998; Schaffer,2002). This becomes especially importantwhen analyzing the motion of higher-ordercantilever vibration modes (Stark, 2004;Schaffer and Fuchs, 2005), when assessingthe amount of thermal noise in AFM mea-surements (Schaffer, 2005), or when usingthermal noise for a calibration of the can-tilever spring constant (Proksch et al., 2004).

Page 243: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

SMALL CANTILEVERS 239

Figure 13.11. Schematic of a head for small cantilevers. A collimated and polarized light source provides theincident beam that is focused onto the cantilever by a movable lens assembly. The reflected beam is separated fromthe incident beam by the polarizing beamsplitter–λ/4-plate assembly and is projected onto the photodiode detector.A top view mount provides optical access and aids in the focusing process. [Reprinted from Schaffer et al. (1997),by permission of the International Society of Optical Engineering.]

13.4.6. Fast, Low-Noise Detector

In Section 13.4.4, the result was derived thatthe thermal cantilever noise depends onlyon the coefficient of viscous damping. Inparticular, it is independent of the springconstant. For the detection of small can-tilever deflections, however, a smaller springconstant is of advantage, since it increasesthe magnitude of the transfer function [Eq.(13.7)], which therefore causes larger deflec-tions for a given force acting on the can-tilever. But it was shown above that thereis a lower limit to the thickness with whichsmall cantilevers can be fabricated, therebysetting a lower limit to the spring constant aswell. Also, for some applications it is desir-able to have a large spring constant (e.g.,for “position-clamp” type of experiments).For small cantilevers, low-noise detectiontherefore becomes particularly important,because only with low-noise detection can

the low-noise properties of small cantileversbe harvested.

For the widespread optical beam deflec-tion (“optical lever”) detection method,the significant noise sources are connectedwith the generation and the detection ofthe light beam: (a) time-correlated fluc-tuations/drift in the power of the beam(“intensity noise”), (b) time-correlated fluc-tuations/drift in the shape/direction of thebeam (“pointing noise”), and (c) time-uncor-related fluctuations in the photodetection pro-cess (shot noise). Other noise sources (darkcurrent noise, electronic Johnson noise) areusually not limiting, but electronic 1/f noisecan be a factor in dc measurements (con-tact mode, force spectroscopy). Intensity andpointing noise is caused by instabilities inthe laser cavity, resulting in mode-hoppingand mode competition. Pointing noise can betransformed into intensity noise by the use ofa single-mode optical fiber (Cleveland et al.,

Page 244: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

240 HIGH-SPEED ATOMIC FORCE MICROSCOPY OF BIOMOLECULES IN MOTION

1995), and intensity noise can be minimizedby the detection circuitry with the help of afast divider (although analog dividers intro-duce noise, too). Shot noise arises fromthe fact that photons are emitted at randompoints in time (resulting in Poisson statis-tics). The current produced by the electronsin the photodetection process, where pho-tons cause the separation of electron–holepairs, therefore exhibits random statisticalfluctuations in its magnitude [with a flat(“white”) frequency distribution]. These fluc-tuations increase with the square root of thebeam power and are a fundamental limit tothe detection sensitivity. They also impose afundamental limit on the scan speed, sincethe faster the measurement, the larger therequired bandwidth, and thus the higher thenoise level in the measurement.

Several methods have been devised toincrease the sensitivity of the photodetec-tion in an AFM. Since the beam deflec-tion detection method measures the lateralshift of a light beam, the particular size andshape of the beam is important. The sizeand shape of the focused spot should be tai-lored to the size and shape of the particularcantilever for optimum signal-to-noise ratio.One way of achieving this is the use of anadjustable aperture in the incident beam path.By using such an adjustable aperture, thesignal-to-noise ratio of a particular measure-ment of cantilever deflections was increasedby a factor of three (Schaffer and Hansma,1998).

The detection sensitivity can further beincreased by the use of an array detec-tor (Schaffer et al., 2000). Such an arraydetector consists of an array of photode-tector segments over which the reflectedbeam is distributed (Fig. 13.12). The indi-vidual signals from each segment are firstweighted by amplifying them with indi-vidual gain factors. Then all the weightedsignals from all the segments are added,producing the cantilever deflection signalthat is further used in the feedback ordata acquisition system. The interesting

Figure 13.12. Schematic of the array detector. Thereflected beam (highlighted area) is spread acrossan array of photodetector segments (1, 2, . . ., N ).The signal from each segment is amplified by anindividual weighting factor (g1, g2, . . . , gN ) that can beset dynamically. The weighted signals are then addedto form the cantilever deflection signal. Such an arraydetector can be programmed to dynamically adapt tothe experimental conditions. For each condition, a setof weighting factors can be found that maximizes thedetection sensitivity.

property of such a detector is that a setof gain factors can be found that opti-mizes the detection sensitivity for anygiven experimental condition. The arraydetector can be programmed to dynami-cally adapt to varying experimental con-ditions. Depending on the particular shapeof the spot on the detector, significantimprovements in the signal-to-noise ratioof measurements of cantilever deflectioncan be obtained. The more distorted andjagged the beam on the detector, the largerthe improvement. In a particular case ofa 12-µm-long cantilever, an increase inthe signal-to-noise ratio by a factor of5 was achieved (Fig. 13.13). Apart fromthis increased signal-to-noise ratio, an arraydetector also provides an increased measure-ment range, a property that is especiallyimportant for the recording of force-distancespectra (Schaffer, 2002).

Page 245: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

SMALL CANTILEVERS 241

Figure 13.13. Thermal noise amplitude density of a small cantilever (L = 12 µm), measured with a two-segmentdetector and an array detector. Both detectors faithfully identify the thermal resonance peak, but the detection noiselevel (off-resonance) is lower by a factor of 5 in case of the array detector. [Reprinted from Schaffer et al. (2000),by permission of the American Institute of Physics.]

13.4.7. Fast Scanner

In the AFM, piezoelectric scanners areroutinely used for the generation of thescan motion. Those scanners typically con-sist of ferroelectric lead zirconate titanate(PZT) ceramics that is made piezoelectricby “poling” [applying a large electricfield (>≈ 2000 V/mm) while the mate-rial is heated above the Curie temperature(TC ≈ 200–400◦C)]. A piezoelectric mate-rial expands or contracts upon applicationof a voltage due to the (inverse) longitudi-nal or transverse piezoelectric effect. Tubescanners are conventionally used since theycan be designed such that they independentlybend and change their length. By mountingone end of such a tube scanner to a rigid sur-face and attaching a sample to the other end,the sample can be scanned in three dimen-sions. A limit to the speed with which thesample can be moved is set by the resonantfrequency of the scanner. The fundamentallongitudinal resonant frequency of a pris-matic (constant cross section) elastic rod oflength L that is clamped at one end and freeat the other end is

f longres =

√E

ρ

1

4L(13.15)

where E and ρ are Young’s modulus andthe density of the rod, respectively. Fortypical PZT ceramics, ρ ≈ 7700 kg/m3 andE = E11 ≈ 65 GPa (Young’s modulus in thedirection of the rod elongation, which is usu-ally perpendicular to the polarization axisof the ceramics). A typical piezo tube thatmay be 5 cm in length therefore has a freeresonant frequency of about 14.5 kHz. Theextra mass, M , of the sample and mount-ing devices like magnets that are added tothe free end of the tube lowers the reso-nant frequency of the tube by a factor ofabout (1 + 3M/m)−1/2, where m is the massof the free tube (this uses the fact that theeffective mass of the tube is about one-third of its true mass). In the end, a typi-cal longitudinal resonant frequency of sucha scanner may be on the order of 10 kHz,and the resonant frequencies in the bend-ing modes may even be smaller. The mostdirect way to create fast scanners, just likein the case of cantilevers, is to make themsmaller [note that the resonant frequency inEq. (15) inversely scales with the length]. Ifthe size of the scanner is reduced by a fac-tor of 10, the resonant frequency increases tothe order of 100 kHz. Small multilayer PZTscanners (so-called “piezo stacks”) with freeresonant frequencies of 260 kHz have beenused to build a scanner with a mechanical

Page 246: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

242 HIGH-SPEED ATOMIC FORCE MICROSCOPY OF BIOMOLECULES IN MOTION

resonant frequency above 60 kHz that wasused to scan consecutive 100 × 100 pixelimages in a liquid environment at an imagerate of 12.5 Hz (Ando et al., 2001). Sys-tem resonances could be eliminated byan active damping method (Kodera et al.,2005). Using finite element analysis, it waspossible to significantly increase the resonantfrequency of a mechanical scanning struc-ture to 23.4 kHz (Kindt et al., 2004a). Animage rate of 80 Hz (128 × 128 pixels) wasdemonstrated on highly oriented pyrolyticgraphite (HOPG) with a scanning tunnel-ing microscope (STM) at atomic resolu-tion in vacuum (Rost et al., 2005). By usinga miniature scanner based on a mechani-cal microresonator, images of soft moltenpolymer surfaces in air could be acquiredat a rate of 70 Hz (Humphris et al., 2005).Even smaller scanners can be made by inte-grating a zinc oxide piezo actuator directlyon the cantilever (Sulchek et al., 2000a).Such self-actuated cantilevers offer a largeimaging bandwidth, but are currently notwell-suited for imaging delicate biologicalsamples due to their large spring constants.Recently, however, an insulated piezoelectriccantilever was used to produce tapping modeimages of E. coli bacteria in liquid with a tipspeed of 75.5 µm/s (Rogers et al., 2003).

13.4.8. Fast Feedback Loop

In tapping mode, the cantilever is oscillatedat the drive frequency. The signal from theoptical detector therefore is also oscillatory.The amplitude of the oscillations is obtainedby feeding the signal into an RMS-DCconverter. This amplitude accounts for theimaging force and is used as input to thesubsequent feedback loop that tries to keepthe amplitude constant (“constant amplitudemode”). A conventional RMS-DC converterworks by first rectifying the input signaland subsequently low-pass filtering it. Thelow-pass filter needs to be designed suchthat it completely separates the modulationsignal from the carrier signal (at the drive

frequency), typically requiring a low-passcutoff frequency substantially lower thanthe drive frequency. (Actually, the RMS-DCconverter shifts the carrier signal completelyto twice the drive frequency when thedeflection signal is ac-filtered.) In otherwords, a conventional RMS-DC convertertypically requires several carrier cycles forthe conversion. To circumvent this limitation,a novel RMS-DC converter was designedthat is based on peak detection, achievinghigher conversion rates (Ando et al., 2002).

In conventional AFMs, a proportional-integral-differential (PID) feedback con-troller is used to keep the error signal(deflection in contact mode, amplitude intapping mode) constant. A PID feedbackloop, however, is limited to a speed set by theresonant frequency of the (z-) scanner. Addi-tional resonances in the closed-loop systemwhich cause additional phase shifts reducethe maximum scan speed further. Mod-ern high-performance feedback controllersthat are based on H∞ theory account forthe presence of system resonances, result-ing in an increased scan rate (Schitter et al.,2001, 2004; Salapaka et al., 2005). Also,actively controlling the cantilever dynamicsvia Q control allows the scan rate to beincreased (Mertz et al., 1993; Sulchek et al.,2000b) or allows scanning in the attractivemode to be stabilized (Anczykowski et al.,1998).

13.4.9. Biological Applications

The following is a list of different appli-cations where small cantilevers have beenused so far. The first biomolecules imagedwith a small cantilever (L = 26 µm) wereIgG antibodies adsorbed to mica in liq-uid (Walters et al., 1996). The image ratewas 0.5 Hz for 128 × 128 pixels. A can-tilever with L = 10 µm was used to imagethe surface of cleaved and bleached nacre,a high-performance biomineralized materialfrom sea shells (Schaffer et al., 1997). Themotion of calcite crystal step edges (Walters

Page 247: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

CONCLUSION 243

Figure 13.14. High-speed image of DNA adsorbed tomica in buffer solution. (a) Height image of DNAplasmids, acquired in 5.6 s (256 × 256 pixels) with atapping frequency of 129 kHz. (b) Height image oflinear DNA fragments, acquired in 1.7 s (128 × 128pixels) with a tapping frequency of 191 kHz. [Reprintedfrom Viani et al. (1999), by permission of the AmericanInstitute of Physics.]

et al., 1997b) and the step growth closeto screw dislocations (Paloczi et al., 1998)were imaged, providing insight into thegrowth of sea shells that partially consist ofcalcite. Small cantilevers were used to pro-duce fast images of DNA (Fig. 13.14) andto perform fast force spectroscopy experi-ments (Viani et al., 1999a, b). The degra-dation of DNA by DNase I was imagedwith an image rate of 0.5 Hz (Hansma,2001). The association and dissociation of

the chaperonin GroEL–GroES complex wasobserved, and its lifetime (on a mica sur-face) was determined as ≈ 5 s (Viani et al.,2000). Myosin V was imaged in the pres-ence of ATP with an image rate of 12.5 Hz at100 × 100 pixels, revealing a relative motionbetween the head/neck region, the long tail,and the globular tail end (Ando et al., 2001).Caged compounds were used to confirm thatthis motion is likely to be caused by ATPhydrolysis (Ando et al., 2003).

13.5. CONCLUSION

While a photographic snapshot can providea beautiful image, it is not well-suitedfor reconstructing the timely successionof events because it preserves a muchreduced picture of reality. Image sequencesor movies, on the other hand, carry moreinformation about the succession of eventsand, in a sense, leave less room for sub-jective interpretation. In just the same way,AFM image sequences allow us to directlyaddress questions in biology and medicinethat single AFM images cannot. This chapterpresented an overview of some of the issuesthat are connected with high-speed imag-ing of biomolecules. It started by givingsome examples of time-resolved AFM imag-ing. Then, a number of indirect ways forincreasing the time resolution were pre-sented. Finally, small cantilevers were shownto be a key for increasing the scan speed. Itis expected that small cantilevers will con-tribute extensively in future investigations ofdynamic biological processes.

Acknowledgments

I would like to thank Paul Hansma, San-dor Kasas, Thomas Jovin, Yuekan Jiao, JohnSader, and Harald Fuchs for helpful dis-cussions and support. I am also grateful toJason Cleveland and Mario Viani of AsylumResearch for valuable technical and scien-tific support. This work was supported finan-cially by the Gemeinnutzige Hertie–Stiftung

Page 248: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

244 HIGH-SPEED ATOMIC FORCE MICROSCOPY OF BIOMOLECULES IN MOTION

in the Stifterverband fur die Deutsche Wis-senschaft. I am also grateful to Jason Cleve-land, Roger Proksch, and Mario Viani ofAsylum Research.

REFERENCES

Akama, Y., Nishimura, E., Sakai, A., and Mura-kami, H. (1990). New scanning tunnelingmicroscopy tip for measuring surface topogra-phy. J. Vac. Sci. Technol. A 8(1):429–433.

Anczykowski, B., Cleveland, J. P., Kruger, D.,Elings, V., and Fuchs, H. (1998). Analysis ofthe interaction mechanisms in dynamic modeSFM by means of experimental data and com-puter simulation. Appl. Phys. A 66:885–889.

Ando, T., Kodera, N., Takai, E., Maruyama, D.,Saito, K., and Toda, A. (2001). A high-speedAFM for studying biological macromolecules.Proc. Natl. Acad. Sci. USA 98:12468–12472.

Ando, T., Kodera, N., Maruyama, D., Takai, E.,Saito, K., and Toda, A. (2002). A high-speedatomic force microscope for studying biologicalmacromolecules in action. Jpn. J. Appl. Phys.41:4851–4856.

Ando, T., Kodera, N., Naito, Y., Kinoshita, T.,Furuta, K., and Toyoshima, Y. Y. (2003). Ahigh-speed atomic force microscope for study-ing biological macromolecules in action. Chem.Phys. Chem. 4:1196–1202.

Baselt, D. R., and Baldeschwieler, J. D. (1994).Imaging spectroscopy with the atomic forcemicroscope. J. Appl. Phys. 76:33–38.

Bezanilla, M., Drake, B., Nudler, E., Kashlev,M., Hansma, P. K., Hansma, H. G. (1994).Motion and enzymatic degradation of DNAin the atomic force microscope. Biophys. J.67:2454–2459.

Binnig, G., Quate, C. F., and Gerber, C. (1986).Atomic force microscope. Phys. Rev. Lett.56:930–933.

Chand, A., Viani, M. B., Schaffer, T. E., andHansma, P. K. (2000). Microfabricated smallmetal cantilevers with silicon tip for atomicforce microscopy. J. Microelectromech. Syst.9:112–116.

Chen, C., Clegg, D. O., and Hansma, H. G.(1998). Structures and dynamic motion oflaminin-1 as observed by atomic forcemicroscopy. Biochemistry 37:8262–8267.

Cleveland, J. P., Schaffer, T. E., and Hansma,P. K. (1995). Probing oscillatory hydrationpotentials using thermal–mechanical noise inan atomic force microscope. Phys. Rev. B.52:8692–8695.

Drake, B., Prater, C. B., Weisenhorn, A. L.,Gould, S. A. C., Albrecht, T. R., Quate, C. F.,Cannell, D. S., Hansma, H. G., and Hansma,P. K. (1989). Imaging crystals, polymers, andprocesses in water with the atomic force micro-scope. Science 243:1586–1589.

Ellis, D. J., Dryden, D. T. F., Berge, T., Edward-son, J. M., and Henderson, R. M. (1999).Direct observation of DNA translocation andcleavage by the EcoKI endonuclease usingatomic force microscopy. Nature Struct. Biol.6:15–17.

Fantner, G. E., Hegarty, P., Kindt, J. H., Schit-ter, G., Cidade, G. A. G., and Hansma, P. K.(2005). Data acquisition system for high speedatomic force microscopy. Rev. Sci. Instrum.76:026118.

Fritz, M., Radmacher, M., and Gaub, H. E. (1993).In vitro activation of human platelets trig-gered and probed by time-lapse atomic forcemicroscopy. Exp. Cell Res. 205:187–190.

Fritz, M., Radmacher, M., Cleveland, J. P.,Allersma, M. W., Steward, R. J., Gieselmann,R., Janmey, P., Schmidt, C. F., and Hansma,P. K. (1995). Imaging globular and filamentousproteins in physiological buffer solutions withtapping mode atomic force microscopy. Lang-muir 11:3529–3535.

Goldsbury, C., Kistler, J., Aebi, U., Arvinte, T.,and Cooper, G. J. S. (1999). Watching amy-loid fibrils grow by time-lapse atomic forcemicroscopy. J. Mol. Biol. 285:33–39.

Grandbois, M., Clausen-Schaumann, H., andGaub, H. E. (1998). Atomic force microscopeimaging of phospholipid bilayer degradation byphospholipase A2. Biophys. J. 74:2398–2404.

Guthold, M., Zhu, X., Rivetti, C., Yang, G.,Thomson, N. H., Kasas, S., Hansma, H. G.,Smith, B., Hansma, P. K., and Bustamante, C.(1999). Direct observation of one-dimensionaldiffusion and transcription by escherichia coliRNA polymerase. Biophys. J. 77:2284–2294.

Han, W., Lindsay, S. M., Dlakic, M., and Har-rington, R. E. (1997). Kinked DNA. Nature386:563.

Page 249: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 245

Hansma, P. K., Cleveland, J. P., Radmacher, M.,Walters, D. A., Hillner, P. E., Bezanilla, M.,Fritz, M., Vie, D., Hansma, H. G., Prater,C. B., Massie, J., Fukunaga, L., Gurley, J., andElings, V. (1994). Tapping mode in liquids.Appl. Phys. Lett. 64:1738–1740.

Hansma, H. G. (2001). Surface biology of DNAby atomic force microscopy. Annu. Rev. Chem.52:71–92.

Hodges, A. R., Bussmann, K. M., and Hoh, J. H.(2001). Improved atomic force microscope can-tilever performance by ion beam modification.Rev. Sci. Instrum. 72:3880–3883.

Hosaka, S., Etoh, K., Kikukawa, A., and Koy-anagi, H. (2000). Megahertz silicon atomicforce microscopy (AFM) cantilever and high-speed readout in AFM-based recording. J. Vac.Sci. Technol. B 18:94–99.

Humphris, A. D. L., Miles, M. J., and Hobbs,J. K. (2005). A mechanical microscope: High-speed atomic force microscopy. Appl. Phys.Lett. 86:034106.

Janovjak, H., Struckmeier, J., and Muller, D. J.(2005). Hydrodynamic effects in fast AFMsingle-molecule force measurements. Eur. Bio-phys. J. 34:91–96.

Jiao, Y., and Schaffer, T. E. (2004). Accurateheight and volume measurements on soft sam-ples with the atomic force microscope. Lang-muir 20:10038–10045.

Jiao, Y., Cherny, D. I., Heim, G., Jovin, T. M.,and Schaffer, T. E. (2001). Dynamic interac-tions of p53 with DNA in solution by time-lapse atomic force microscopy. J. Mol. Biol.314:233–243.

Kasas, S., Thomson, N. H., Smith, B. L.,Hansma, H. G., Zhu, X., Guthold, M., Bus-tamante, C., Kool, E. T., Kashlev, M., andHansma, P. K. (1997). Escherichia coli RNApolymerase activity observed using atomicforce microscopy. Biochemistry 36:461–468.

Kasas, S., Thomson, N. H., Schaffer, T. E.,Dietler, G., Catsicas, S., and Hansma, P. K.(2000). Simulation of an atomic force micro-scope imaging a moving protein. ProbeMicrosc. 2:37–44.

Kindt, J. H., Fantner, G. E., Cutroni, J. A., andHansma, P. K. (2004a). Rigid design of fastscanning probe microscopes using finite ele-ment analysis. Ultramicroscopy 100:259–265.

Kindt, J. H., Fantner, G. E., Thompson, J. B.,and Hansma, P. K. (2004b). Automated wafer-scale fabrication of electron beam depositedtips for atomic force microscopes using patternrecognition. Nanotechnology 15:1131–1134.

Kodera, N., Yamashita, H., and Ando, T. (2005).Active damping of the scanner for high-speedatomic force microscopy. Rev. Sci. Instrum.76(5):53708.

Levine, A. J. (1997). p53, the cellular gatekeeperfor growth and division. Cell 88:323–331.

Longfellow, H. W. (1839). Hyperion, 1st ed.,Book II, Chapter VI, Samuel Colman, Boston.

Marti, O., Ruf, A., Hipp, M., Bielefeldt, H.,Colchero, J., and Mlynek, J. (1992). Mechan-ical and thermal effects of laser irradiation onforce microscope cantilevers. Ultramicroscopy42:345–350.

Matzke, R., Jacobson, K., and Radmacher, M.(2001). Direct, high-resolution measurement offurrow stiffening during division of adherentcells. Nature Cell Biol. 3:607–610.

Mertz, J., Marti, O., and Mlynek, J. (1993). Reg-ulation of a microcantilever response by forcefeedback. Appl. Phys. Lett. 62:2344–2346.

Muller, D. J., Engel, A., Matthey, U., Meier, T.,Dimroth, P., and Suda, K. (2003). Observingmembrane protein diffusion at subnanometerresolution. J. Mol. Biol. 327:925–930.

Nagami, F., Zuccheri, G., Samori, B., andKuroda, R. (2002). Time-lapse imaging of con-formational changes in supercoiled DNA byscanning force microscopy. Anal. Biochem.300:170–176.

Ookubo, N., and Yumotoa, S. (1999). Rapidsurface topography using a tapping modeatomic force microscope. Appl. Phys. Lett.74:2149–2151.

Paloczi, G. T., Smith, B. L., Hansma, P. K., Wal-ters, D. A., and Wendman, M. A. (1998).Rapid imaging of calcite crystal growth usingatomic force microscopy with small cantilevers.Appl. Phys. Lett. 73:1658–1660.

Proksch, R., Schaffer, T. E., Cleveland, J. P.,Callahan, R. C., and Viani, M. B. (2004).Finite optical spot size and position correc-tions in thermal spring constant calibration.Nanotechnology 15:1344–1350.

Putman, C. A. J., van der Werf, K. O., de Grooth,B. G., and Van Hulst, N. F. (1994). Tapping

Page 250: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

246 HIGH-SPEED ATOMIC FORCE MICROSCOPY OF BIOMOLECULES IN MOTION

mode atomic force microscopy in liquid. Appl.Phys. Lett. 64:2454–2456.

Radmacher, M., Fritz, M., Hansma, H. G., andHansma, P. K. (1994a). Direct observation ofenzyme activity with the atomic force micro-scope. Science 265:1577–1579.

Radmacher, M., Cleveland, J. P., Fritz, M.,Hansma, H. G., and Hansma, P. K. (1994b).Mapping interaction forces with the atomicforce microscope. Biophys. J. 66:2159–2165.

Reiley, T. S., Fan, L. S., and Mamin, H. J.(1995). Micromechanical structures for datastorage. Microelectron. Eng. 27:495–498.

Revenko, I., and Proksch, R. (2000). Magneticand acoustic tapping mode microscopy ofliquid phase phospholipid bilayers and DNAmolecules. J. Appl. Phys. 87:526–533.

Rief, M., and Grubmuller, H. (2002). Force spec-troscopy of single biomolecules. Chem. Phys.Chem. 3:255–261.

Rogers, B., Sulchek, T., Murray, K., York, D.,Jones, M., Manning, L., Malekos, S.,Beneschott, B., Adams, J. D., Cavazos, H.,and Minne, S. C. (2003). High speed tappingmode atomic force microscopy in liquid usingan insulated piezoelectric cantilever. Rev. Sci.Instrum. 74:4683–4686.

Rost, M. J., Crama, L., Schakel, P., van Tol, E.,van Velzen-Williams, G. B. E. M., Overgauw,C. F., ter Horst, H., Dekker, H., Okhui-jsen, B., Seynen, M., Vijftigschild, A., Han, P.,Katan, A. J., Schoots, K., Schumm, R., vanLoo, W., Oosterkamp, T. H., and Frenken,J. W. M. (2005). Scanning probe microscopesgo video rate and beyond. Rev. Sci. Instrum.76:053710.

Sader, J. E. (1998). Frequency response of can-tilever beams immersed in viscous fluids withapplications to the atomic force microscope.J. Appl. Phys. 84:64–76.

Salapaka, S., De, T., and Sebastian, A. (2005).Sample-profile estimate for fast atomic forcemicroscopy. Appl. Phys. Lett. 87:53112.

Schaffer, T. E., Richter, M., and Viani, M. B.(2000). Array detector for the atomic forcemicroscope. Appl. Phys. Lett. 76:3644–3646.

Schaffer, T. E. (2002). Force spectroscopy with alarge dynamic range using small cantilevers andan array detector. J. Appl. Phys. 91:4739–4746.

Schaffer, T. E. (2005). Calculation of thermalnoise in an atomic force microscope with

a finite optical spot size. Nanotechnology16:664–670.

Schaffer, T. E., and Fuchs, H. (2005). Optimizeddetection of normal vibration modes of atomicforce microscope cantilevers with the opti-cal beam deflection method. J. Appl. Phys.97:083524.

Schaffer, T. E., and Hansma, P. K. (1998). Char-acterization and optimization of the detectionsensitivity of an atomic force microscope forsmall cantilevers. J. Appl. Phys. 84:4661–4666.

Schaffer, T. E., Cleveland, J. P., Ohnesorge, F.,Walters, D. A., and Hansma, P. K. (1996).Studies of vibrating atomic force micro-scope cantilevers in liquid. J. Appl. Phys.80:3622–3627.

Schaffer, T. E., Viani, M., Walters, D. A., Drake,B., Runge, E. K., Cleveland, J. P., Wendman,M. A., and Hansma, P. K. (1997). An atomic

force microscope for small cantilevers. Proc.SPIE 3009 (Micromachining and Imag-ing):48–52.

Schitter, G., Menold, P., Knapp, H. F., Allgower,F, and Stemmer, A. (2001). High performancefeedback for fast scanning atomic forcemicroscopes. Rev. Sci. Instrum. 72:3320–3327.

Schitter, G., Allgower, F., and Stemmer, A.(2004). A new control strategy for high-speedatomic force microscopy. Nanotechnology15:108–114.

Serry, F. M., Neuzil, P., Vilasuso, R., and Mac-lay, G. J. (1995). Air-damping of resonantAFM microcantilevers in the presence of anearby surface. Proceedings of the SecondInternational Symposium on Microstructuresand Microfabricated Systems, pp. 83–89, Elec-trochemical Society, Chicago.

Shao, Z., and Zhang, Y. (1996). Biological cryoatomic force microscopy: A brief review.Ultramicroscopy 66:141–152.

Stark, R. W. (2004). Optical lever detectionin higher eigenmode dynamic atomic forcemicroscopy. Rev. Sci. Instrum. 75(11):5053–5055.

Stoffler, D., Goldie, K. N., Feja, B., and Aebi, U.(1999). Calcium-mediated structural changes ofnative nuclear pore complexes monitored bytime-lapse atomic force microscopy. J. Mol.Biol. 287:741–752.

Sulchek, T., Hsieh, R., Adams, J. D., Minne,S. C., Quate, C. F., and Adderton, D. M.

Page 251: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 247

(2000a). High-speed atomic force microscopyin liquid. Rev. Sci. Instrum. 71:2097–2099.

Sulchek, T., Hsieh, R., Adams, J. D., Yarali-oglu, G. G., Minne, S. C., Quate, C. F., Cleve-land, J. P., Atalar, A., and Adderton, D. M.(2000b). High-speed tapping mode imag-ing with active Q-control for atomic forcemicroscopy. Appl. Phys. Lett. 76:1473–1475.

Thomson, N. H., Fritz, M., Radmacher, M., Cleve-land, J. P., Schmidt, C. F., and Hansma, P. K.(1996). Protein tracking and detection of proteinmotion using atomic force microscopy. Biophys.J. 70:2421–2431.

Thomson, N. H., Kasas, S., Riederer, B. M., Cat-sicas, S., Dietler, G., Kulik, A. J., and Forro, L.(2003). Large fluctuations in the disassemblyrate of microtubules revealed by atomic forcemicroscopy. Ultramicroscopy 97:239–247.

van Noort, S. J. T., van der Werf, K. O., Eker,A. P. M., Wyman, C., de Grooth, B. G., van

Hulst, N. F., and Greve, J. (1998). Direct visu-alization of dynamic protein–DNA interactionswith a dedicated atomic force microscope. Bio-phys. J. 74:2840–2849.

van Noort, S. J. T., van der Werf, K. O., deGrooth, B. G., and Greve, J. (1999). Highspeed atomic force microscopy of biomoleculesby image tracking. Biophys. J. 77:2295–2303.

Viani, M. B., Schaffer, T. E., Chand, A., Rief, M.,Gaub, H. E., and Hansma, P. K. (1999a). Smallcantilevers for force spectroscopy of singlemolecules. J. Appl. Phys. 86:2258–2262.

Viani, M. B., Schaffer, T. E., Paloczi, G. T., Pie-trasanta, L. I., Smith, B. L., Thompson, J. B.,Richter, M., Rief, M., Gaub, H. E., Plaxco,K. W., Cleland, A. N., Hansma, H. G., and

Hansma, P. K. (1999b). Fast imaging and fastforce spectroscopy of single biopolymers witha new atomic force microscope designedfor small cantilevers. Rev. Sci. Instrum. 70:4300–4303.

Viani, M. B., Pietrasanta, L. I., Thompson, J. B.,Chand, A., Gebeshuber, I. C., Kindt, J. H.,Richter, M., Hansma, H. G., and Hansma, P. K.(2000). Probing protein–protein interactions inreal time. Nature Struct. Biol. 7:644–647.

Walters, D. A., Cleveland, J. P., Hansma, P. K.,Wendman, M. A., Gurley, G., and Elings, V.(1996). Short cantilevers for atomic forcemicroscopy. Rev. Sci. Instrum. 67:3583–3589.

Walters, D. A., Smith, B. L., Belcher, A. M.,Paloczi, G. T., Stucky, G. D., Morse, D. E.,Hansma, P. K. (1997a). Modification of cal-cite crystal growth by abalone shell proteins:An atomic force microscope study. Biophys. J.72:1425–1433.

Walters, D. A., Viani, M., Paloczi, G. T., Schaf-fer, T. E., Cleveland, J. P., Wendman, M. A.,Elings, V., and Hansma, P. K. (1997b). Atomicforce microscopy using short cantilevers. Proc.SPIE 3009 (Micromachining and Imaging):43–47.

Page 252: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

CHAPTER 14

ATOMIC FORCE MICROSCOPYIN CYTOGENETICSS. THALHAMMERDepartment fur Geo-und Umweltwissenschaften, University of Munchen, Theresienstr.41, MunichGermany; GSF—National Center for Environment and Health, Institute of Radiation Protection,Ingolstadter Landstrasse 1, 85764 Neuherberg, Germany

W. M. HECKLDepartment fur Geo-und Umweltwissenschaften, University of Munchen, Theresienstr.41, MunichGermany

I must beg leave to propose a separatetechnical name “chromosome” for thosethings have been called by Boveri “chro-matic elements”, in which there occurs oneof the most important acts in karyokinesis,viz., the longitudinal splitting. They are soimportant that a special and shorter nameappears useful. If the term I propose ispractically applicable; it will become famil-iar, otherwise it will soon sink into obliv-ion.—Waldeyer (1890)

14.1. INTRODUCTION

The beginning of modern human cytoge-netics dates back to 1956. In this year,scientists from Indonesia, Sweden, and Eng-land showed the number of human chromo-somes to be 46 and not 48 as previouslyestimated (Tjio and Levan, 1956; Ford andHamerton, 1956). Chromosomes were a latediscovery, and were first described in 1842 byNageli (Nageli, 1842).

Advances in human cytogenetics havebeen largely dependent on technical factors,

such as the availability of suitable tissuecontaining an adequate number of cellswith the potential for cell division. In thelate 1950s, metaphase chromosomes wereobtained from skin fibroblasts. The use ofphytohemagglutinin (PHA) to stimulate celldivision in peripheral blood lymphocytes(Moorhead et al., 1960), along with hypo-tonic treatment for obtaining better metaphasespreads (Hsu, 1952), was a major break-through that increased the number of meta-phases available for analysis after 69–72 h.These metaphases had chromosomes of opti-mal quality that could clearly be classified intoseven groups, A to G. The X chromosome wasidentified within the C group, whereas the Ychromosome was placed in the G group. Theidentification of individual chromosomes wasan arduous task, because chromosomes withina group resembled one another morphologi-cally and specific staining techniques were notyet available.

In the 1960s, investigators started toidentify individual single chromosomes and

Force Microscopy: Applications in Biology and Medicine, edited by Bhanu P. Jena and J.K. Heinrich Horber.Copyright 2006 John Wiley & Sons, Inc.

249

Page 253: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

250 ATOMIC FORCE MICROSCOPY IN CYTOGENETICS

structural abnormal chromosomes usingautoradiographic techniques (German, 1964).However, neither chromosome morphologynor autoradiography provided unequivocalidentification of all chromosomes in thehuman genome. In this field the first advancescame from the Caspersson group. Theydescribed chromosome staining by quinacrinemustard (QM), a technique they termed Q-banding, since chromosomes were differen-tiated into bright and dark regions, termed“bands” (Caspersson et al., 1964). After that,more than a dozen new staining techniquesfor identifying individual chromosomes andabnormalities were developed. An overviewis listed by Verma and Babu (1989).

Fluorescence in situ hybridization (FISH)has become a popular technique over thepast years, often applied to gene mappingand molecular cytogenetics (Adionolfi andDavies, 1994). Biotin or digoxigenin-labeledprobes have been used most commonlyin conjunction with secondary reagents forprobe detection. Nucleoside triphosphateanalogs directly coupled to fluorophores thatcan be incorporated into DNA or RNA spec-imens by conventional enzymatic reactionsare now available from a variety of com-mercial available sources, as are chemicallysynthesized oligonucleotide labels with oneor more fluorochrome molecules.

In 1982 the scanning tunneling micro-scope (STM; Binnig et al., 1982) wasinvented. This instrument, which can imagesolid surfaces with atomic resolution, wasrevolutionary for microscopy and surfaceanalysis. In the STM a sharp conducting tipis brought into close proximity of the samplesurface and a current of electrons flows fromthe tip to sample, or vice versa dependingon the sign of the bias voltage. Scanning thetip in a grid pattern over the surface yieldsa topographic image of the sample surfaceunder investigation. The main limitation ofthe STM is the fact that only conductingor semiconducting samples can be imaged.To circumvent this problem of sample con-ductivity, Binnig, Quate and Gerber came

up with a new microscope, the atomic forcemicroscope (AFM; Binnig et al., 1986).

In the atomic force microscope a sharp tipis mounted on the free end of a cantilever.While scanning the tip over the sample sur-face, forces acting between tip and sampledeflect the cantilever. A two-dimensional mapof the local interactions between tip and sam-ple surface can be obtained, by measuringthese deflections from point to point witha beam-bounce detection method. With thistechnique, as well as utilizing various differ-ent scanning modes, a wide range of data (e.g.,the surface relevant structure or local measure-ments of the physical and chemical propertiesof the sample) can be recorded (Colton et al.,1998). A lateral resolution of 1 nm and verticalresolution of 0.1 nm can be achieved. Further-more, the AFM can operate under fluid. It isan essential step for biological applicationsto work and to image under physiologicalenvironment (Marti et al., 1987). Structuralanalysis at high resolution not only providesmore information about molecular complexesbut is also used for in vivo experiments usingbiological systems. Data can be recorded inreal time. Besides structural information ofbiological systems, three-dimensional data,micromechanical behaviors, dynamic pro-cesses, and molecular interactions can berecorded. Table 14.1 shows a short compar-ison of AFM to other microscopic techniquestogether with the necessary sample prepara-tion procedures.

14.2. IMAGING OF GENETICMATERIAL

Cytogenetics is basically a visual science.Established microscopic techniques, such aslight and electron microscopy, have beenwidely used for the study of chromosomes.After the invention of the atomic force micro-scope, it was applied in different fields ofgenetic applications. Double-stranded DNAon freshly cleaved mica in air was imaged byseveral groups (Thundat et al., 1992; Yang and

Page 254: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

IMAGING OF GENETIC MATERIAL 251

TABLE 14.1. Comparison of Different Microscopic Techniques to Image the Sample Topography—ForExample, Metaphase Chromosome.

Field Emission inLens Scanning

Conventional Scanning Electron Electron Atomic ForceOptical Microscopy Microscope Microscopy

Microscopy (SEM) (FEISEM) (AFM)

Microscopicenvironment

Ambient liquidvacuum

Vacuum Vacuum Ambient liquid vacuum

Focus depth Medium Small SmallResolution 100 nm n/a 5 nm n/a 0.1–1.0 nmx,y, z 0.01 nmMagnification 1 × –2 × 103× 10 × –106× 5 × 102 × –108×Necessary

samplepreparation

Low Freeze-drying,coating

Low Low

Necessarysampleproperties

Samples do not haveto be completelytransparent forvisible light

Samples should notcharge and haveto be vacuumcompatible

Samples should do nothave excessive changesin height compared totip geometry

Shao, 1993). Hansma et al. (1992 and 1993)successfully imaged plasmid-DNA fixed onmica in propanol. By adding new spreadingchemicals (e.g., quaternary ammonium salts),it is possible to reduce surface impurities, andthe surface density of the molecules could bereproducibly measured (Schaper et al., 1994).After the introduction of the tapping mode, itbecame possible to image DNA with lowershear forces during scanning, which resultedin being able to obtain much detailed imagesof the macromolecule (Delain et al., 1992). InFig. 14.1 we have imaged a double-strandedplasmid, pUC19, in AFM tapping mode in air.DNA in a highly condensed state in spermcells was imaged by Allen et al. (1993) inair and in liquids. Furthermore, Fritzscheand co-workers performed structural exper-iments on chromatin fibers (Fritzsche et al.,1997), as well as determined the volumeof metaphase chromosomes (Fritzsche andHenderson, 1996). Structural examinations onmetaphase chromosomes were also performedby de Grooth and Putman (de Grooth and Put-man, 1992; Rasch et al., 1993). Figure 14.2shows untreated human metaphase chromo-somes. Using chemically and enzymatically

untreated metaphase chromosomes, a GTG-like banding pattern—G-bands by trypsinusing Giemsa—could be observed in thetopographic images (Musio et al., 1994).Metaphase chromosomes imaged by AFMhave revealed structures similar to thosereported in light and electron microscopy.Depending on the preparation technique, sub-structural details can be recorded in metaphasechromosomes (Tamayo et al., 1999; Gobbiet al., 2000). After pepsin digestion of themetaphase chromosomes, a granular substruc-ture was detected using AFM in contactmode. In such cases, not only the cover-ing plasma layer but also the scaffold stabi-lizing proteins were digested. The recordeddetails represent a nucleosomal structure,which has been discussed by several investi-gators (Winfield et al., 1995; de Grooth andPutman, 1992; Fritzsche et al., 1994). Therecorded data are comparable to those gener-ated by scanning electron microscopy (Wan-ner et al., 1991). Metaphase chromosomesconsist of 30-nm fibers folded in a tandemarray of radial loops, which are packaged intoa fiber with an overall diameter between 200and 250 nm. High-resolution AFM images ofmetaphase chromosomes revealed structural

Page 255: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

252 ATOMIC FORCE MICROSCOPY IN CYTOGENETICS

Figure 14.1. AFM image of plasmid pUC19 (∼2.7 kbp) diluted in dH2O; deposited by spin stretching and imagedin tapping mode in air.

Figure 14.2. (a) Three-dimensional AFM image, recorded in contact mode, of untreated human chromosomes. Barrepresents 2 µm; (b) AFM enhancement of the p-arm of the recorded chromosome. Bar represents 1 µm.

features in the size range of 30–100 nm,which correspond to the loops of the 30-nmfiber (de Grooth and Putman, 1992). Other,(Winfield et al., 1995; McMaster et al., 1996)have reported features as small as 10–20 nm,which could correspond to individual nucleo-somes. While scanning in the contact mode,the tip of the AFM can be contaminatedwith chromosomal material. This material,adhering to the tip, can limit the use ofsuch tips for manipulation and microdissec-tion experiments.

In noncontact mode, the tip scans at a dis-tance of a few hundred angstroms over thechromosomal surface. The tip is not in con-tact with the sample surface and thereforedoes not get contaminated while scanning.

The advantages and disadvantages of thesetwo operation modes are combined in thetapping mode (Hansma, 1993), where thetip comes in contact with the chromoso-mal surface for a very short period. Insummary, these three operating modes canbe used for imaging chromosomal mate-rial (see Fig. 14.3), and to record sub-structures depending on sample preparation.Care is taken so that the topography ofthe metaphase chromosomes are preservedand not deformed (de Grooth and Putman,1992; Musio et al., 1994). Table 14.2 sum-marizes the methodical properties of con-tact, noncontact, and tapping mode forhigh-resolution imaging and manipulation ofmetaphase chromosomes.

Page 256: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

KARYOTYPING OF METAPHASE CHROMOSOMES 253

Figure 14.3. Comparison of different operating modes. (a) AFM image in contact mode. (b) Noncontact mode.(c) Tapping mode of a GTG-banded human metaphase chromosome 11. Bar represents 1 µm.

TABLE 14.2. Methodical Properties of the Different Operation Modes in AFM for High-ResolutionImaging, Manipulation, and Microdissection of Metaphase Chromosomes.

Operation Mode Contact Mode Noncontact Mode Tapping Mode

Tip loading force Low → high Low LowContact with sample

surfaceYes No Periodical

Manipulation ofsample

Yes No Yes

Contamination ofAFM tip

Yes No Yes

Microdissection Yes No No

Using specific in situ hybridization tech-niques, we can detect distinct areas ofhybridization in metaphase chromosomes.Biotinylated DNA probes were used for map-ping, and the specific sites were visualized bydetecting changes in topography induced by aperoxidase–diaminobenzidine reaction (Put-man et al., 1993; Rasch et al., 1993). Using thesame detection technique, Kalle et al. (1995)were able to identify specific signals afterRNA in situ hybridization. In studies on cerealchromosomes, a genome specific probe hasbeen used. Using AFM, we can detect changesin height of biotin–avidin–fluorescein isoth-iocyanate complexes formed as a conse-quence of fluorescence in situ hybridizationprocedures (McMaster et al., 1996).

The AFM has also been used to imagegenetic material in liquids. In this case, fixedmetaphase chromosomes swell depending onthe buffer used (Fig. 14.4). The viscoelasticproperties of rehydrated chromosomes and

their volume, have been determined inliquids using AFM (Frizsche, 1999). In com-parison to light or electron microscopy, theAFM is able to operate in liquids and to per-form local measurements on any point of thesample surface (Stark et al., 1998 and Jiao Y.et al., 2004; Hoshio et al., 2004). Due to thisattribute, data the biophysical properties ofthe metaphase chromosome can be obtainedusing the AFM.

14.3. KARYOTYPING OF METAPHASECHROMOSOMES

Chromosome banding techniques have facil-itated the precise identification of individualchromosomes. The GTG banding obtainedby digesting chromosomes with proteolytictrypsin, followed by Giemsa staining, is mostwidely used in routine chromosome analysis.However, the interpretation of the GTG

Page 257: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

254 ATOMIC FORCE MICROSCOPY IN CYTOGENETICS

0

600500400300200100

01 2 3 4 5

distance (µm)

heig

ht (

nm)

A B

560nm

Figure 14.4. (a) AFM image in contact mode of a rehydrated human metaphase chromosomes in PBS buffer. (b)A cross-sectional analysis from point A to B. The swelling in PBS buffer was 560 nm in comparison to TE bufferwith 900 nm (data not shown). Bar represents 1 µm.

bands is still unclear. A direct role of Giemsastain in producing the GTG bands was sug-gested by McKay (1973). Several authorsinferred that chromosomes contain a preex-isting structure, which is enhanced by GTG-banding. It is, however, still unclear howthis enhancement occurs (Comings, 1978;Ambros and Sumner, 1987). It is hypothe-sized that the differences between positiveand negative GTG bands may be induced bythe spatial organization of chromosomal pro-tein and DNA.

In atomic force microscopy, as opposed tolight microscopy, changes in color is unde-tectable. Differentiation can only occur bytopographical information of the metaphasechromosome. The resulting image is notonly the result of topographical changes inthe chromosome surface, but also a resultof the interaction of the tip with the vis-coelastic properties of the chromosome. InFig. 14.5A, a topographic AFM image of aGTG-banded chromosome 7 homologue isshown. The morphology of the chromosomeis preserved, and both the banding patternand the fibrous nature are detectable. Struc-tural protrusions along the chromosome, cor-responding to the dark bands in Fig. 14.5A,are also detectable. A linescan of the q-arm(point A to B in Fig. 14.5B) shows differ-ences in height between dark and light bandsof about 90 nm. The length of the ridges

is about 540 nm. The corresponding bandsare marked in the idiogram (Fig. 14.5C).It is known from chromosomes imagedby scanning electron microscopy that theGiemsa light and dark bands differ in height(Harrison et al., 1981). One must be awarethat the AFM image represents not onlythe topology of the sample surface but alsothe compressibility of the sample; thereforeheight is partially expressed as topography.Figure 14.6 shows a human 2n = 46, XXfemale metaphase spread. The light and darkbands are clearly detectable and all chro-mosomes are identifiable. The correspondingkaryotype is shown in Fig. 14.7.

Furthermore, it is possible to identifyfeatures equivalent to G-banding pattern inuntreated chromosomes and to use these forclassification (Musio et al., 1994; Tamayo J.2003). As in light microscopy, dark andlight bands can be correlated in GTG-bandedchromosomes and can be classified accord-ingly (Thalhammer et al., 2001). In earlierAFM studies, G-positive bands were deter-mined to be areas with a higher surfacerelief (Rasch et al., 1993). After more than20 years, the discussion about the band-ing mechanism is still unsolved. Also, notall related biochemical and physical reac-tions are understood. The comparison ofunstained and Giemsa-stained chromosomesby phase contrast microscopy (Yunis and

Page 258: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

KARYOTYPING OF METAPHASE CHROMOSOMES 255

a)

b)

c)

346 nm

259.5

173

86.5

00 1.12 2.24 3.36

Distance

Z D

ata

4.48 5.6 µm

Figure 14.5. (a) AFM image of a GTG-banded chromosome 7 homologue, topographic AFM image, grayscaleinverted. The bright and dark banding pattern is detectable (bar represents 2 µm). (b) Line measurement throughpoint A to B of the q-arm of chromosome 7. (c) Idiogram of the q-arm of chromosome 7.

Sanchez, 1993; McKay, 1973) set up thehypothesis that staining techniques amplifya preexsisting structure of incomplete struc-tural organization in chromatin. Electronmicroscopy studies support this hypothesis(Heneen and Caspersson, 1973; Bahr andLarsen, 1974). McMaster et al. (1994) sug-gested an influence of the stains to structureand morphology, based on their work onuntreated metaphase chromosomes. In scan-ning electron microscopy, light and dark

bands in the R and G banding pattern canbe differentiated by changes in height (Har-rison et al., 1981). This suggests that the highresolution of the AFM has allowed an intrin-sic banding pattern to be visualized whichotherwise would not be possible without theenhanced accumulation of stains for view-ing using light microscopy. The accuracyof chromosomal banding is strongly relatedto DNA organization with the associatedproteins.

Page 259: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

256 ATOMIC FORCE MICROSCOPY IN CYTOGENETICS

Figure 14.6. AFM image of a 2n = 46, XX female GTG-banded metaphase spread. Bar represents 10 µm.

Figure 14.7. AFM image of a 2n = 46, XX female karyotype.

Page 260: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

ANALYSIS OF THE 3D ORGANIZATION OF HUMAN METAPHASE CHROMOSOMES 257

Figure 14.8. AFM image of a 2n = 46, XY male CBG-banded metaphase spread. Bar represents 10 µm.Enlargement shows two chromosomes with highlighted centromeric region. Bar 2 represents µm.

14.3.1. CBG-Banding

The C-banding technique produces selec-tive staining of constitutive heterochromatin.These bands are mostly located at the cen-tromeric regions of chromosomes, and hencethey are known as C-bands. The originalmethod described by Arrighi and Hsu (1971)primarily involved treatment with an alkali(sodium hydroxide) to denature the chromo-somal DNA, with subsequent incubation in asalt solution. An alternate method, describedby Sumner (1972), utilizes a milder alkali,(barium hydroxide). However, both methodsproduce similar characteristic C-banding pat-terns (Fig. 14.8) observed using AFM in thecontact mode. In contrast to light microscopicimages, the stained centromeric regions areclearly detectable in the AFM micrographs.Similarly, C-banding for studying chromo-some rearrangements near centromeres andfor investigating polymorphisms have alsobeen performed using AFM by Tan et al.(2001) and Fukushi et al. (2005).

14.4. CORRELATIVEHIGH-RESOLUTIONMORPHOLOGICAL ANALYSISOF THE THREE-DIMENSIONALORGANIZATION OF HUMANMETAPHASE CHROMOSOMES

An understanding of the cell’s nuclear func-tions requires an accurate knowledge of thespatial organization of nuclear structures. Formany years, the study of human metaphasechromosomes has been carried out usinglight microscopy following staining proto-cols. Scanning electron microscopy (SEM)provides higher-resolution imaging com-pared to light microscopy, and it permits sur-face analysis of the chromosomal structure,which cannot be adequately obtained fromtransmission electron microscopy (TEM).Nevertheless, in order to obtain high-resolution SEM images, the use of a highelectron accelerating voltage (up to 30 kV)has been used (Sumner and Ross, 1989;

Page 261: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

258 ATOMIC FORCE MICROSCOPY IN CYTOGENETICS

Sanchez-Sweatman, 1993; Sumner, 1996).Under these experimental conditions, sputter-coating or conductive staining of the samplesis generally required (Sumner, 1994; Wannerand Formanek, 1995). However, since bothprocedures allow electron-charging disper-sion from the sample, sputtering or stain-ing obscures fine details and may producealterations to the sample (Hermann andMuller, 1992).

Today, only a few techniques are availablefor high-resolution imaging of chromosomalmaterial with reduced artifacts. These includethe field emission in lens scanning elec-tron microscope (FEISEM) and the atomicforce microscope (AFM). The FEISEM rep-resents a special kind of SEM, fitted witha cold cathode field emission electron gun(Nagatani et al., 1987; Pawley, 1997) thatcan operate at low accelerating voltage withreduced electron charging of the sample. Infact, the low-voltage and low-current elec-tron beam of the FEISEM, together witha liquid nitrogen anticontamination devicein correspondence to the specimen areaand an “in lens” assembly of the electron-optic column, allows high-resolution imagingof the biological sample without any con-ductive staining or metal coating. Hence,contamination of the specimen is greatly

reduced, compared to conventional SEMimaging (Nagatani et al., 1987). The samplelocation between the objective pole pieceslimits the dispersion of the secondary elec-trons collected by the magnetic field ofthe lens. These characteristics allow theobservation of uncoated biological sampleswith a higher resolution than with conven-tional SEM (Rizzoli et al., 1994; Rizzi et al.,1995; Lattanzi et al., 1998; Gobbi et al.,1999).

FEISEM and AFM microscopy can alsobe combined to observe the same metaphasechromosome prepared from human HL 60cells, by standard cytogenetic method aftercleaning the metaphase spreads (Rizzoliet al., 1994). The analysis of the samesamples can be facilitated by the use ofconductive glass (ITO glass) for the chromo-some map preparation. These two differenttechnical approaches show a high correla-tion of the respective morphological infor-mation, in both normal and treated samples.The high resolution potential of the FEISEM,together with the possibility of observinghydrated samples and/or nanomanipulatingthe specimen with the AFM, confirms mor-phological data and offers an enhanced infor-mation (Figs. 14.9 and 14.10) (Gobbi et al.,2000).

Figure 14.9. Atomic force microscopy images of human metaphase chromosomes. (a) A flat and short untreatedchromosome is well-identifiable on the ITO glass. (b) Present a more defined network structure. Bar represents1 µm.

Page 262: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

AFM NANOEXTRACTION 259

Figure 14.10. Field emission in-lens scanning electron microscopy analysis of protease K-treated samples. (a) thecentromeric region (arrows) and the chromatids are well-recognizable. A dark halo surrounds the entire chromosome(bar represents 1 µm). (b) Increasing the magnification, the chromosomal surface appears to be constituted of anetwork. Some fibrillar structures parallel to the axis of the chromatid are well-detectable (arrows). The halo aroundthe chromatid appears to be formed by a mix of fibers and homogeneous phase (asterisk) (Bar represents 100 nm).

14.5. AFM AS A NANOMANIPULATORAND DISSECTING TOOL

By combining high structural resolution withthe ability to change the image parametersat any place of the scan area, it is possi-ble to use the AFM as a manipulation tool.In 1991, Hoh et al. (1991) demonstrated theuse of AFM as a microdissection device.They performed microdissection on cellulartight junctions. Controlled nanomanipulationof biomolecules have also been performedon genetic material (Hansma et al., 1992)where 100- to 150-nm fragments were cut-out circular plasmid rings. Isolated DNAadsorbed on a mica surface have been dis-sected both in air (Henderson, 1992; Vesenkaet al., 1992; Geissler et al., 2000) and in liq-uids—for example, propanol (Hansma et al.,1992). These experiments demonstrate thefeasibility of microdissection in the nanome-ter range using the AFM. Combining AFMimaging and microdissection capability, theorganization of bovine sperm nuclei hasbeen investigated, showing small proteinand DNA containing subunits of 50–100 nmin diameter (Allen et al., 1993). Similarly,tobacco mosaic viruses have been dissectedand displaced on a graphite surface to recordmechanical properties of the virus (Falvoet al., 1997).

Microdissection of genetic material in adifferent condensation states (polytene chro-mosomes of Drosophila melanogaster) havebeen performed by Henderson and cowork-ers (Mosher et al., 1994; Jondle et al., 1995).Metaphase chromosomes have been microdis-sected, and the extracted material was used forbiochemical evaluation (Thalhammer et al.,1997; Xu and Ikai, 1998). As illustrated inFig. 14.11, chromosomes can be microdis-sected at selected regions, and the processcan be documented (Thalhammer et al., 1997;Stark et al., 1998; Iwabuchii, S. et al., 2002).In Figure 14.11c, an electron microscopicimage of an AFM tip following microdissec-tion is shown. The extracted genetic materialadhering to the tip can be amplified by poly-merase chain reaction and used as a probe forfluorescence in situ hybridization (Fig. 14.12)(Thalhammer et al., 1997). Importantly, theAFM can also operate in liquids. While per-forming microdissection in liquids on rehy-drated chromosomes, only uncontrolled dis-sections can be produced (Fig. 14.13) (Starket al., 1998) .

14.6. AFM NANOEXTRACTION

The procedure for AFM-based microdissec-tion is described in this section. All stepsshould be performed under sterile conditions

Page 263: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

260 ATOMIC FORCE MICROSCOPY IN CYTOGENETICS

a) c)b)

Figure 14.11. (a) AFM image after microdissection of band 2q12 of human chromosome 2. The arrow indicatesthe scan orientation. (b) Cross-sectional analysis along the cut site A to B. The cut width is 95 nm. (c) Electronmicroscopic image of the AFM tip after microdissection. The arrow indicates the extracted DNA, bar represents1 µm.

Figure 14.12. Fluorescence in situ hybridization (FISH) after microdissection of the centromeric region of a humanC group chromosome: (a) The hybridization signals are visible in the centromeric regions of chromosome 1, 5, 7,8, and 19. (b) AFM control image of the microdissected chromosome. Bar represents 1 µm.

Figure 14.13. Microdissection using atomic force microscopy (M-AFM) of a metaphase chromosome rehydrated inTE-Puffer. For dissection the loading force was increased to 1 µN, and a line scan at 1 µm/s was performed withoutz-modulation. A large part of the chromosome was pushed to the side in an uncontrolled manner.

Page 264: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

AFM NANOEXTRACTION 261

to avoid contamination. To identify the chro-mosomal region of interest and to minimizecontamination of the AFM tip during scan-ning the area of interest, GTG-bandedmetaphase chromosomes should be imagedin noncontact mode in ambient air. Thechromosome can also be identified usinga “preset” fluorescence in situ hybridiza-tion of chromosome-specific painting probes(Thalhammer et al., 1997) and fluores-cence microscopy. This “preset” hybridiza-tion also increases the amount of extracted

genetic material. For microdissection, thechromosome is placed at a 90◦ angle to thescan direction and the chromosomal area iszoomed into. For distance control, amplitudedetection is used and the damping level isset to 50% of the amplitude of free oscil-lation for imaging before extraction. Afteridentification of the extraction site, the scanis stopped and the feedback is turned off. Theloading force of the tip onto the sample isincreased. Figures 14.14 and 14.15 show theresults of AFM microdissection by applying

a) b)

Figure 14.14. Comparison of two different AFM dissection modes in air (z-scale 0–180 nm). (a) A series of cutshas been made without z-modulation. Each cut was performed by scanning one line scan at 1 µm/s at a certainloading force: #1, 2.8 µN; #2, 5.6 µN; #3, 8.5 µN; #4, 11.2 µN; #5, 14.0 µN; #6, 16.8 µN; #7, 19.6 µN; #8,22.4 µN; #9, 25.6 µN. (b) The human metaphase chromosome was imaged by AFM in ambient conditions after aseries of dissections made by AFM. For dissection z-modulation (∼5 nm) has been used. The oscillation amplitudeof the cantilever was smaller than 1% of the amplitude of free oscillation for all cuts.

250

200

150

100

50

00 1 2 3 4 5 6 7

AB

98

76 5 4 3 2

1

Substrate

Distance (µm)

Hei

ght (

nm)

(a) (b)

Figure 14.15. (a) Cross-sectional analysis as indicated in Fig. 14.13B. The positions of the different cuts are markedby the numbers. (b) Electron microscopic image of human metaphase chromosome after microdissection with verticalmodulation of the z-piezo. Loading forces are: # 1, 16.8 µN; #2, 19.6 µN; #3, 22.4 µN; #4, 25.6 µN; #5, > 27 µN.

Page 265: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

262 ATOMIC FORCE MICROSCOPY IN CYTOGENETICS

different loading forces to the specimen andby controlling the modulation of the z-piezo.

To extract DNA, a one-line scan with1 µm/s is performed at the site. During dis-section of the chromosome, lateral forcesplay an important role. The tip performs astick–slip movement, and the forces betweenthe cantilever tip and the chromosome arereduced while operating with z modulation.The shear forces of the tip are reduced duringdissection and reproducible cuts of 100 nmcan be made, depending on the geometry ofthe tip. During microdissection, not only theapex but also the flank of the tip is in contactwith the chromosome, and the loading areato the chromosome is increased. The chro-mosomal material is not dissected in a firststep but pushed like a snowplough. Parts ofthe chromosomal material adhere by van derWalls interaction and unspecific adsorptionto the tip. To increase the dissection effi-ciency, electron-beam-deposited tips (EBD)can be used. EBD tips with a rough surfacecan be used to increase the extraction effi-ciency (Fig. 14.16A), while EBD tips withpredetermined breaking points minimize therisk of contamination (Fig. 14.16B) (Thal-hammer et al., 2002). Thus, the modifiedAFM tips can be used like a mechanical“nanoscalpel” or a “nanoshovel”. After thetip is retracted from the sample surface, thecantilever is transferred into a reaction tube.A new cantilever is used to check the cut at

the nanoextraction site on the chromosome.The reaction tube contains a collection bufferto stabilize the extracted genetic material.Enzymatic digestion of the chromosome sta-bilizing and covering proteins is performedto increase primer binding and therefore theefficiency of the polymerase chain reaction(PCR). Unspecific amplification can also beperformed with PCR techniques using degen-erated primers (Thalhammer et al., 1997)or adaptor-linked PCR (Thalhammer et al.,2005). The generated genetic samples canfurther be used for cytogenetic studies—forexample, FISH (Thalhammer et al., 1997).

14.7. CONCLUSION

It is clear that based on the working princi-ples of the AFM, it may be used not only forhigh-resolution imaging of genetic material,but also as a nanosurgical tool. In additionto high structural three-dimensional analysisof genetic material, the AFM microdissectioncan be applied to isolate smaller cytogeneticsamples for biochemical analysis. These canbe used in combination with highly sensi-tive polymerase chain reactions and fluores-cence in situ hybridization, for either phys-ical mapping of the genome, evolutionarystudies, or diagnostic research. Furthermore,it will be interesting to implement a near-field optical microscope in order to identify a

(a) (b)200 nm

Figure 14.16. Scanning tunneling microscopic image of electron beam deposited AFM tips to increase the extractionefficiency after M-AFM. (a) Rough AFM tip, bar represents 200 nm; tip with syringe tip and predetermined breakingpoint, bar represents 1 µm.

Page 266: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 263

particular genomic region labeled with onlyfew dye molecules for subsequent nanodis-section using the AFM.

REFERENCES

Adinolfi, M., and Davies, A. F. (1994). Non-isotopic in situ hybridization. In: Applicationsto Clinical Diagnosis and Molecular Genetics,Medical Intelligence Unit series, R.G. Landes,Austin, Texas.

Allen, M. J., Lee, C., Lee, J. D., IV, Pogany,G. C., Balooch, M., Siekhaus, W. J., and Bal-horn, R. (1993). Atomic force microscopy ofmammalian sperm chromatin. Chromosoma102:623–630.

Ambros, P. F., and Sumner, A. T. (1987). Correla-tion of pachytene chromomeres and metaphasebands of human chromosomes and distinctiveproperties of telomeric regions. Cytogenet. CellGenet 44:223–228.

Arrighi, F. E., and Hsu, T. E. (1971). Localiza-tion of heterochromatin in human chromo-somes. Cytogenetics 10:81–86.

Bahr, G. F., and Larsen, P. M. (1974). Structuralbands in human chromosomes. Adv. Cell. Mol.Biol. 3:191–212.

Binnig, G., Rohrer, H., Gerber, C., and Weibl, E.(1982). Surface studies by scanning tunnelingmicrocopy. Phys. Rev. Lett. 49:57–61.

Binnig, G., Quate, C. F., and Gerber, C. (1986).Atomic force microscope. Phys. Rev. Lett.56:930–933.

Caspersson, T., Farber, S., Foley, G. E.,Kudynowski, J., Modest, E. J., Simonsson, E.,Wagh,U., and Zech, L. (1964). Chemical differentia-

tion along metaphase chromosomes. Exp. CellRes. 49:219–222.

Colton, R. J., Engel, A., Frommer, J. E., Gaub,H. E., Gewirth, A. A., Guckenberger, R., Rabe,J., Heckl, W. M., and Parkinson, B. (1998).

Procedures in Scanning Probe Microscopies.John Wiley & Sons, Chichester.

Comings, D. E. (1978). Mechanisms of chromo-somes banding and implications for chromo-some structure. Annu. Rev Genet. 12:25–46.

de Grooth, B. G., and Putman, C. A. J. (1992).High-resolution imaging of chromosome related

structures by atomic force microscopy. J.Microsc. 168:239–247.

Delain, E., Fourcade, A., Poulin, J. C., Barbin, A.,Coulaud, D., Lecam, E., and Paris, E. (1992).Comparative observations of biological spec-imens, especially DNA and filamentous actinmolecules in atomic force, tunneling and elec-tron microscopes. Microsc. Microanal. Micro-struct. 3:457–470.

Falvo, M. R., Washburn, S., Superfine, R.,Finch, M., Brooks, F. P., Chi, V., Taylor, R. M.(1997). Manipulation of individual viruses: Fric-tion and mechanical properties. Biophys. J.72:1396–1403.

Ford, C. E., and Hamerton, J. L. (1956). Thechromosomes of man. Nature 178:1020–1023.

Fritzsche, W. (1999). Salt-dependent chromo-some viscoelasticity characterized by scanningforce microscopy-based volume measurements.Microsc. Res. Tech. 44:357–362.

Fritzsche, W., and Henderson, E. (1996). Volumedetermination of human metaphase chromo-somes by scanning force microscopy. ScanningMicrosc. 10:1–7.

Fritzsche, W., Schaper, A., and Jovin, T. M.(1994). Probing chromatin with the scanningforce microscope. Chromosoma 103:231–236.

Fritzsche, W., Takac, L., and Henderson, E.(1997). Application of atomic force microscopyto visualization of DNA, chromatin andchromosomes. Crit. Rev. Eukaryot. Gene Expr.7:231–240.

Fukushi, D., and Ushiki, T. (2005). The structureof C-banded human metaphase chromosomesas observed by atomic force microscopy. ArchHistol Cytol. 68(1):81–7.

Geissler, B., Noll, F., and Hampp, N. (2000).Nanodissection and noncontact imaging ofplasmid DNA with an atomic force microscope.Scanning 22:7–11.

German, J. L. (1964). The pattern of DNA syn-thesis in the chromosomes of human bloodcells. J. Cell Biol. 20:37–55.

Gobbi, P., Falconi, M., Vitale, M., Galanzi, A.,Artico, M., Martelli, A. M., and Mazzotti, G.(1999). Scanning electron microscopic detec-tion of nuclear structures involved in DNAreplication. Arch. Histol. Cytol. 62:317–326.

Page 267: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

264 ATOMIC FORCE MICROSCOPY IN CYTOGENETICS

Gobbi, P., Thalhammer, S., Falconi, M., Stark, R.,Heckl, W. M., and Mazzotti, G. (2000). Cor-relative high resolution morphological anal-ysis of the three-dimensional organizationof human metaphasechromosomes. Scanning22:273–281.

Hansma, H. G., Vesenka, J., Siegerist, C., Kel-derman, G., Morrett, H., Sinsheimer, R. L., Elings, V., Bustamante, C., and Hansma, P. K.(1992). Reproducible imaging and dissectionof plasmid DNA under liquid with the atomicforce microscope. Science 256:1180–1183.

Hansma, H. G., Sinsheimer, R. L., Groppe, J., Bruice, T. C., Elings, V., Gurley, G., Bezanilla,M., Mastrangelo, I. A., Hough, P. V., and

Hansma, P. K. (1993). Recent advances inatomic force microscopy of DNA. Scanning15(5):296–299.

Harrison, C. J., Britch, M., Allen, T. D., and Har-ris, R. (1981). Scanning electron microscopy ofthe G-banded human karyotype. Exp. Cell Res.134:141–153.

Henderson, E. (1992). Imaging and nanodissec-tion of individual supercoiled plasmids byatomic force microscopy. Nucleic Acids Res.20:445–447.

Heneen, W. K., and Caspersson, T. (1973). Iden-tification of chromosomes of rye by distributionpatterns of DNA. Hereditas 74:259–272.

Hermann, R., and Muller, M. (1992). Towardshigh resolution SEM of biological objects.Arch. Histol. Cytol. 55:17–25.

Hoh, J. H., Lal, R., John, S. A., Revel, J. P.,and Arnsdorf, M. F. (1991). Atomic forcemicroscopy and dissection of gap junctions.Science 253:1405–1408.

Hoshi, O., Owen, R., Miles, M., and Ushiki, T.(2004). Imaging of human metaphase chromo-somes by atomic force microscopy in liquid.Cytogenet Genome Res. 107(1–2):28–31.

Hsu, T. C. (1952). Mammalian chromosomesin vitro. 1. The karyotype of man. J. Hered.43:167–172.

Iwabuchii, S., Mori, T., Ogawa, K., Sato, K.,Saito, M., Morita, Y., Ushiki, T., and Tamiya,E. (2002). Atomic force microscope-based dis-section of human metaphase chromosomes andhigh resolutional imaging by carbon nanotubetip. Arch Histol Cytol. Dec;65(5):473–9.

Jiao, Y., and Schaffer, T. E. (2004). Accurateheight and volume measurements on soft sam-ples with the atomic force microscope. Lang-muir. 9;20(23):10038–45.

Jondle, D. M., Ambrosio, L., Vesenka, J., andHenderson, E. (1995). Imaging and manipulat-ing chromosomes with the atomic force micro-scope. Chromosome Res. 3:239–244.

Kalle, W. H. J., Macville, M. V. E., van deCorput, M. P. C., de Grooth B. G., Tanke, H. J.,and Raap, A. K. (1995). Imaging of RNA in situhybridization by atomic force microscopy. J.Microsc. 182:192–199.

Lattanzi, G., Galanzi, A., Gobbi, P., Falconi, M.,Matteucci, A., Breschi, L., Vitale, M., and Maz-zotti, G. (1998). Ultrastructural aspects of theDNA polymerase a distribution during the cellcycle. J. Histochem. Cytochem. 46:1435–1442.

Marti, O., Drake, B., and Hansma, P. K. (1987).Atomic force microscopy of liquid-coveredsurfaces: Atomic resolution images. Appl PhysLett. 51:484–486.

McKay, R. D. (1973). The mechanism of G andC banding in mammalian metaphase chromo-some. Chromosoma 44:1–14.

McMaster, T. J., Hickish, T., Min, T., Cunning-ham, D., and Miles, M. J. (1994). Applicationof scanning force microscopy to chromosomeanalysis. Cancer Genet. Cellgenet. 76:93–95.

McMaster, T. J., Winfield, M. O., Karp, A., andMiles, M. J. (1996). Analysis of cereal chro-mosomes by atomic force microscopy. Genome39:439–444.

Moorhead, P. S., Nowell, P. C., Mellman, W. J.,Battips, D. M., and Hungerford, D. A. (1960).Chromosome preparations of leucocytes cul-tured from human peripheral blood. Exp. CellRes. 20:613–616.

Mosher, C., Jondle, D., Ambrosio, L., Vesenka, J.,and Henderson, E. (1994). Microdissection andmeasurement of polytene chromosomes usingthe atomic force microscope. Scanning Microsc.8(3):491–497.

Musio, A., Mariani, T., Frediani, C., Sbrana, I.,and Ascoli, C. (1994). Longitudinal patternssimilar to G-banding in intreated humanchromosomes: Evidence from atomic forcemicroscopy. Chromosoma 103:225–229.

Nageli, K. (1842). Zur Entwicklungsgeschichte derPollen . (Ilse Jahn, ed.) Spektrum AkademischerVerlag Gustav Fischer, Berlin, Zurich.

Page 268: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 265

Nagatani, T., Saito, S., Sato, M., and Yamada, M.(1987). Development of an ultra high resolutionscanning electron microscope by means ofa field emission source and in-lens system.Scanning Microsc. 1:901–909.

Pawley, J. (1997). The development of field-emission scanning electron microscopy forimaging biological surfaces. Scanning 19:324–336.

Putman, C. A. J., de Grooth, B. G., Wiegant, J.,Raap, A. K., van der Werf, K. O., van Hulst,N. F., and Greve, J. (1993). Detection of

in situ hybridization to human chromosomeswith the atomic force microscope. Cytometry14:356–361.

Rasch, P., Wiedemann, U., Wienberg, J., andHeckl, W. M. (1993). Analysis of bandedhuman-chromosomes and in situ hybridizationpatterns by scanning force microscopy. Proc.Natl. Acad. Sci. USA 90:2509–2511.

Rizzi, E., Falconi, M., Baratta, B., Manzoli, L.,Galanzi, A., Lattanzi, G., and Mazzotti, G.(1995). High-resolution FEISEM detection ofDNA centromeric probes in HeLa metaphasechromosomes. J. Histochem. Cytochem. 43:413–419.

Rizzoli, R., Rizzi, E., Falconi, M., Galanzi, A.,Baratta, B., Lattanzi, G., Vitale, M., Manzoli, L.,and Mazzotti, G. (1994). High resolution detec-tion of uncoated metaphase chromosome bymeans of field emission scanning electronmicroscopy. Chromosoma 103:393–400.

Schaper, A., Pascual, S. J. P., and Jovin, T. M.(1994). The scanning force microscopy of DNAin air and in n-propanol using new spreadingagents. FEBS Lett. 355:91–95.

Stark, R., Thalhammer, S., Wienberg, J., andHeckl, W. M. (1998). The AFM as a toolfor chromosomal dissection—the influence ofphysical parameters. Appl. Phys. A66:579–584.

Sumner, A. T. (1972). A simple technique fordemonstrating centromeric heterochromatin.Exp. Cell Res. 75:304–306.

Sumner, A. T. (1996). Problems in preparation ofchromosomes for scanning electron microscopyto reveal morphology and to permit immuno-cytochemistry of sensitive antigens.. ScanMicrosc. 10:165–176.

Sumner, A. T., and Ross, A. (1989). Factorsaffecting preparation of chromosomes for scan-ning electron microscopy using osmium impreg-nation. Scanning Microsc. (Suppl.) 3:87–97.

Sumner, A. T., Ross, A. R., and Graham, E.(1994). Preparation of chromosomes forscanning electron microscopy. Methods Mol.Biol. 29:41–50.

Tamayo, J., Miles, M., Thein, A., and Soothill, P.(1999). Selective cleaning of the cell debrisin human chromosome preparations studiedby scanning force microscopy. J. Struct. Biol.128:200–210.

Tamayo, J. (2003). Structure of human chromo-somes studied by atomic force microscopy. PartII. Relationship between structure and cytoge-netic bands. J Struct Biol. 141(3):189–97.

Tan, E., Iffet, F., Ergun, M. A., Ercan, I., andMenevse, A. (2001). C-banding visualized byatomic force microscopy. Scanning 23:32–35.

Thalhammer, S., Stark, R., Muller, S., Wienberg,J., and Heckl, W. M. (1997). The atomic force

microscope as a new microdissecting tool forthe generation of genetic probes. J. Struct. Biol.119(2):232–237.

Thalhammer, S., Kohler, U., Stark, R., and Heckl,W. M. (2001). GTG banding pattern on human

metaphase chromosomes revealed by high res-olution atomic-force microscopy. J. Microsc.202(3):464–467.

Thalhammer, S., and Heckl, W. M. (2005).Manipulation of genetic material. Nano Today5:40–49.

Thundat, T., Allison, D. P., Warmack, R. J., andFerrell, T. L. (1992). Imaging isolated strandsof DNA molecules by atomic force microscopy.Ultramicroscopy 4244:1101–1106.

Tjio, J. H., and Levan, A. (1956). The chromo-some number of man. Hereditas 42:1–6.

Verma, R. S., and Babu, A. (1989). Human Chro-mosomes: Manual of Basic Techniques, Perga-mon Press, New York.

Vesenka, J., Guthold, M., Tang, C. L., Keller, D.,Delaine, E., and Bustammante, C. (1992). Sub-strate preparation for reliable imaging of DNAmolecules with the scanning force microscope.Ultramicroscopy 4244:1243–1249.

Waldeyer, W. (1890). Karyokinesis and its rela-tion to the process of fertilization. Q. J.Microsc. Sci. 30:159–281.

Page 269: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

266 ATOMIC FORCE MICROSCOPY IN CYTOGENETICS

Wanner, G., and Formanek, H. (1995). Imagingof DNA in human and plant chromosomes byhigh-resolution scanning electron microscopy.Chromosome Res. 3:368–374.

Wanner, G., Formanek, H., Martin, R., and Her-rmann, R. G. (1991). High resolution scanningelectron microscopy of plant chromosomes.Chromosoma 100:103–109.

Winfield, M., McMaster, T. J., Karp, A., andMiles, M. J. (1995). Atomic force microscopyof plant chromosomes. Chromosome Res. 3:128–131.

Xu, X. M., and Ikai, A. (1998). Retrieval andamplification of single-copy genomic DNAfrom a nanometer region of chromosomes: Anew and potential application of atomic forcemicroscopy in genomic research. Biochem.Biophys. Res. Commun. 248:744–748.

Yang, J., and Shao, Z. (1993). Effect of probe forceon the resolution of atomic force microscopy ofDNA. Ultramicroscopy 50:157–170.

Yunis, J. J., and Sanchez, O. (1993). G-bandingand chromosome structure. Chromosoma 44:15–23.

Page 270: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

CHAPTER 15

ATOMIC FORCE MICROSCOPY IN THE STUDYOF MACROMOLECULAR INTERACTIONSIN HEMOSTASIS AND THROMBOSIS: UTILITYFOR INVESTIGATION OF THEANTIPHOSPHOLIPID SYNDROMEWILLIAM J. MONTIGNYMicroscopy Imaging Center, College of Medicine, University of Vermont, Bulington, Vermont

ANTHONY S. QUINNDepartment of Pathology and Microscopy Imaging Center, College of Medicine, University ofVermont, Burlington, Vermont

XIAO-XUAN WUDepartment of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine,Bronx, New York

EDWIN G. BOVILLDepartment of Pathology, College of Medicine, University of Vermont, Burlington, Vermont

JACOB H. RANDDepartment of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine,Bronx, New York

DOUGLAS J. TAATJESDepartment of Pathology and Microscopy Imaging Center, College of Medicine, University ofVermont, Burlington, Vermont

15.1. INTRODUCTION

This chapter demonstrates the unmatchedefficacy of atomic force microscopy (AFM)for the investigation of interactions occurringbetween biological cell-surface macromole-cules on artificial phospholipid membranes.Specifically, the delineation of a series ofcomplex interactions that take place among

factors believed to have seminal roles inmediating increased thrombosis in a clin-ically defined syndrome, the antiphospho-lipid syndrome (APS), clearly illustrates theextraordinary versatility and flexibility ofthis technique (Marchant et al., 2002; Randet al., 2003). The unique ability of theAFM to image objects of an unprecedentedrange of size and complexity (from single

Force Microscopy: Applications in Biology and Medicine, edited by Bhanu P. Jena and J.K. Heinrich Horber.Copyright 2006 John Wiley & Sons, Inc.

267

Page 271: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

268 STUDY OF MACROMOLECULAR INTERACTIONS IN HEMOSTASIS AND THROMBOSIS

small molecules, to complex multicompo-nent enzymes, to whole cells) in situ, in atemporal fashion and within ambient physio-logical buffers that preserve biological formand function, renders this technique invalu-able in studies such as this, that seek toexplore delicate and ephemeral biochemi-cal relationships (Bustamante and Rivetti,1996; Dunlap et al., 1997; Bustamante et al.,1999; Hansma et al., 1999). In concert withother techniques such as ellipsometry, theAFM provides a valuable method to linknanoscale cell-surface molecular changeswith the increased thrombosis and concomi-tant etiology of the antiphospholipid syn-drome (Rand et al., 2003).

15.2. ATOMIC FORCE MICROSCOPY

The development of the atomic force micro-scope has engendered a paradigm shift inthe way that the structure and functionof biological macromolecules and the com-plex relationships that are inherent amongthem are examined. The AFM (Binnig et al.,1986) was originally conceived and devel-oped as an outgrowth of scanning probemicroscopy, which includes near-field scan-ning optical microscopy (NSOM) (Betziget al., 1986; Durig et al., 1986), scanningtunneling microscopy (STM) (Binnig et al.,1982a, b), cryo AFM (Mou et al., 1993; Hanet al., 1995; Shao and Zhang, 1996), and mag-netic force AFM (MAC mode AFM) (Hanet al., 1996). While other atomic (angstrom)or molecular (nanometer) scale imaging tech-niques such as x-ray crystallography, nuclearmagnetic resonance, and transmission elec-tron microscopy are dependent on the detec-tion of transmission, diffraction, or deflec-tion of various wavelengths of light, elec-tron beams, and x-ray beams, the AFM relieson a technology that is based entirely ontactile sensing. In addition to its ability toconvert extremely subtle nuances of surfacedepth and texture into high-resolution three-dimensional images, many other parameters,such as biomechanical characteristics, force

measurements, and enzymatic processes, canbe explored by AFM. The tactile quality ofAFM, in which a nanoscale probe makes phys-ical contact with the sample, means that thistechnique can accurately quantify inter- andintramolecular force interactions as small asthe breaking of hydrogen bonds and as sub-tle as differences in antibody–antigen bind-ing energies (Stroh et al., 2004). The level ofsensitivity that can be achieved allows detec-tion and accurate measurement of pico-newton(pN) levels of force under ambient aqueousconditions that closely replicate in vivo envi-ronments. In fact, molecular interactions suchas DNA–protein binding, intramolecular stre-tching and unwinding, and receptor–ligandcoupling can be measured to tolerances of sev-eral pico-newtons (Bustamante et al., 2000;Marchant et al., 2002; Allemand et al., 2003).In imaging mode, because experiments canbe performed under physiologically relevantconditions with minimal sample processing,AFM can develop dramatic images of dynamicor enzymatic processes such as transcription,replication, DNA bending, DNA–protein, andDNA–lipid interactions in a temporal fash-ion, at submolecular resolution (Dunlap et al.,1997; Hansma et al., 1999; Montigny et al.,2001, 2003; Abdelhady et al., 2003).

The working principles for AFM, whilecomplex in execution, are simple in con-ception and are as follows (see Fig. 15.1):A nanoscale-dimension, oxide-sharpened sil-icon nitride probe is mounted at the end of asilicon nitride cantilever that, depending onvarious imaging modalities, can be of severaldifferent configurations, lengths, and springconstants. The nominal radius of curvature atthe apex of the probe has been estimated atabout 30 nm, although the ability to achieveresolution that may exceed 1 nm argues fora much sharper, higher aspect shape. Thismay in part be due to the presence of veryfine asperities formed during probe fabrica-tion that act as the primary scanning point(Bustamante and Rivetti, 1996; Taatjes et al.,1999). The back of the flexible cantilever iscoated with gold and is highly reflective. The

Page 272: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

ATOMIC FORCE MICROSCOPY 269

Feedback andx, y, z Scan Control

x, y, zPiezoscanner

Laser

Specimen oncleaved mica substrate

Cantileverwith probe

Topographic3-D image

Photodiode

Figure 15.1. Schematic representation of the principles of atomic force microscopy.

cantilever-probe unit is attached to a blockaffixed to a supporting structure that is in turnmounted on the piezo tube. The design andconfiguration of this support is dependent onthe mode of imaging. In air, macromoleculesare deposited (with or without fixation) onsimple metal or mica discs, and the cantilevermount can be quite simple. Imaging in ambi-ent room atmosphere (i.e., “contact modein air”) is the simplest mode of AFM andwas the first method developed. However, aplethora of uncontrollable variables, whichare treated in more detail below, renderthis method problematic for imaging mostbiological samples. The existence of strongadhesive forces (such as capillary attraction),engendered by the nucleation of fluid lay-ers between tip and sample, requires the useof extremely high tip–sample force levelsthat often produce severe sample damage(Bustamante and Rivetti, 1996; Morris et al.,1999). The use of buffered fluid environ-ments for AFM imaging, while more dif-ficult, is far more amenable for preservingdelicate biological molecules or for captur-ing dynamic biochemical processes. It haslargely replaced air imaging for most bio-logical applications (Bustamante et al., 1997;Kindt et al., 2002).

For imaging in fluid environments, a Plexi-glas fluid cell with a watertight gasket is used.The sample chamber used under these condi-tions can be as simple as a water meniscuscreated by a drop of buffer applied directlyto a freshly cleaved mica disc epoxied to aslide; more complex arrangements have beendevised that are sealed with O-rings to pre-vent evaporation and consequent changes inthe ionic strength of imaging buffers. The mostsophisticated units permit the introduction ofbuffer through external ports while simulta-neously scanning, and they may utilize anautomated pump delivery system for continu-ous replenishment and/or instantaneous mod-ification of imaging buffer solutions in situ.Also, finely calibrated temperature control canbe implemented, allowing a level of approx-imation and modulation of the physiologicalenvironment that is incomparable at such high(submolecular) levels of resolution (Kindtet al., 2002).

The probe support assembly (i.e., thefluid cell in fluid mode) with its attachedcantilever/probe unit is mounted at the endof a tube constructed of piezoelectric ceramic(lead titanium zirconate/PTZ) material. Elec-trodes are attached to the column in sucha fashion as to create a bias voltage across

Page 273: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

270 STUDY OF MACROMOLECULAR INTERACTIONS IN HEMOSTASIS AND THROMBOSIS

the piezoelectric tube at sets of horizontallyopposed points located 90◦ apart and at thetop and bottom of the tube. The piezoelectriceffect refers to the ability of these materials togenerate voltage upon compression and defor-mation; the reverse piezoelectric effect causesthe piezoelectric tube to deform in atomicallyaccurate increments upon administration of abias voltage, allowing precise movement inthe x, y, and z directions of the scanning plane(Taylor, 1993; Bustamante and Rivetti, 1996;Morris et al., 1999). During operation, sam-ples are affixed to freshly cleaved muscovitemica, which has an atomically flat surfacewith an RMS surface roughness of 0.06 nm.This surface provides a low-relief nonreac-tive substrate that easily exceeds the surfacetolerances needed to image the smallest bio-logical molecules, and it can even be uti-lized for atomic-scale imaging (Hansma andLaney, 1996; Kindt et al., 2002). The probeis scanned across the surface in raster fashionin the x and y axis in response to the elec-trical voltage applied to the electrodes. As theprobe encounters anomalies such as molecularshapes that perturb the uniform mica surfaceduring the scan, the cantilever is deflected andthe degree of deflection is detected by useof an optical lever effect, created by focus-ing a laser on the back of the highly reflec-tive cantilever surface. The laser is reflectedup to a four-quadrant photodiode, which con-verts laser movement on its surface into ameasurable electrical voltage change, whichis in turn translated via computer softwareinto sensitive height and amplitude informa-tion. Changes in the laser spot position on thevertical axis of the photodiode indicate heightdifferences, while horizontal changes indicatetwisting or torque-like movements of the can-tilever which furnish information about sur-face characteristics such as charge, malleabil-ity, and adhesion. In addition, a feedback loopcan be used to maintain preset force and can-tilever deflection levels; z-axis movements toraise or lower the piezo in response to surfaceheight changes are monitored, and these dataprovide an alternative to direct recording of

changes in photodiode voltage to generate dataabout the scanned surface. The optical levereffect vastly amplifies the sensitivity of detec-tion, with very small low-angle laser deflec-tions at the probe–surface interface trans-lated into comparatively large movements onthe photodiode surface (Bustamante and Riv-etti, 1996; Morris et al., 1999). The theoret-ical detection limits of cantilever displace-ment are infinitesimally small, on the order of4 × 10−4 A, with a signal-to-noise ratio of 1.However, this has been achieved only whenextremely short, stiff cantilevers are utilizedin rigidly controlled settings that are differ-ent from those found in biochemical AFMexperiments (Meyer and Amer, 1988). Underactual experimental conditions, where ran-dom thermal excitation of the cantilever andinstrumental noise are unavoidable, the can-tilever displacement detection sensitivity stillfalls well within atomic scale. That is, a 0.01-nm displacement can generate a detectablevoltage change (Rugar and Hansma, 1990;Bustamante and Rivetti, 1996; Morris et al.,1999). The lower limit of resolution for bio-logical specimens is dependent on a myriadof factors, and ranges from 10−10 to 10−9 m(angstrom–nanometer range). All cantileverdeflection and piezo movement informationfrom a scan is converted into a digital pointmap of the surface, and these height andamplitude data are used to generate a three-dimensional topographical surface renderingwith precise images of sample shape at sub-molecular resolution.

The AFM can be operated in several dif-ferent modes (tapping, contact, fluid imag-ing, etc.) that have varying degrees ofutility depending on experimental condi-tions and sample types. The deciding fac-tors in choosing a particular imaging modeare concerned with attaining balance betweenoften competing parameters that include(a) achieving reproducible, high-resolutionimages, (b) preserving delicate and com-plex structures, (c) simulating physiologicalconditions, and (d) achieving binding con-ditions that permit surface equilibration and

Page 274: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

ATOMIC FORCE MICROSCOPY 271

movement. Contact mode in air is relativelystraightforward and simple, with moleculesbound tightly to the mica surface, providingconsistent reproducible images. However,forces generated by the probe in this modeare large enough to do severe damage tobiological samples, especially due to lateralshearing, and molecules may be swept com-pletely off the mica substrate. Under normalambient environmental conditions in air, cap-illary forces and hard-core repulsions domi-nate the interaction landscape between theprobe tip and the sample (Bustamante andKeller, 1994; Bustamante and Rivetti, 1996).

A shell of hydration several water mole-cules thick forms on all objects, particularlyon sharp tips with small radii of curvature,which act as nucleation points for the deposi-tion of water molecules. Capillary interactions(Laplace’s force) are relatively enormous(on the order of hundreds of nano-newtons),dwarfing other forces, and necessitating theuse of extremely high piezo forces to over-come the attractive interaction (Israelachvili,1985; Morris et al., 1999). In some cases, theprobe may actually become pinned to the sur-face due to this attractive force (Rugar andHansma, 1990). Forces in contact mode canbe minimized by stringently controlling atmo-spheric humidity levels to less than 35%. Atthis level, forces in contact mode can be lim-ited to 1–10 nN, but this still puts a high lowerlimit on the tip–sample force, often causingstructural damage to delicate samples or mul-tisubunit assemblies. In some cases this forceis still high enough to sweep samples off of themica surface (Bustamante and Rivetti, 1996).

The AFM tapping mode seeks to ame-liorate some of these concerns, and it is suc-cessful in most respects. In this mode the can-tilever is excited (by various electrical, mag-netic, piezo, or acoustic stimuli) at close toits resonance frequency. This induces high-frequency oscillation of the cantilever, caus-ing the tip to make a series of extremely briefcontacts on the sample surface, which is farless damaging and invasive, and generates

lower surface force (Hansma et al., 1993; Put-man et al., 1994). However, due to the attrac-tive capillary forces that still exist betweentip and sample, a relatively high lower limitis still set on the force needed to keep thetip in oscillation and not bound to the sam-ple surface (Bustamante et al., 1994). Con-sequently, tapping force levels, while lowerthan those found in contact mode, are stillhigh enough to cause damage to “soft” biolog-ical macromolecules or assemblies, especiallydue to shearing forces. Also, at the force lev-els needed to prevent probe capture, probeoscillation and amplitude are much more dif-ficult to control, leading to difficulties inachieving consistent high-resolution images.Finally, both contact and tapping modes (inair) may induce artifacts, severely alter struc-tural relationships and molecular interactions,and completely abrogate dynamic enzymaticprocesses, due to sample dehydration and des-iccation (Engel et al., 1999).

Use of the tapping mode, in concert witha buffered liquid environment, eliminatesnearly all of the above problems while intro-ducing several other advantages that makethis mode of operation far more amenable forthe analysis of most biological samples. Mostimportantly, imaging in liquid abrogates theshell of hydration at the interface of samplesurface and atmosphere. This nearly elimi-nates capillary interactions, which dwarf allother types of atomic interactions, causing theproblems noted above. Now the force inter-action landscape is dominated by van derWaals forces, electrostatic attractive forces,and hard core repulsions, all of which arerelatively small (0.1–1 nN), allowing theforce that is applied during imaging to bemuch smaller (Bustamante and Rivetti, 1996).Even these lower levels of force are notideal for use with biological macromolecules,and so tapping in conjunction with a liq-uid environment, while difficult, was devel-oped to provide the optimum combinationof lowest force levels, high-quality resolu-tion, and physiological environment (Hansmaet al., 1993; Putman et al., 1994; Kindt et al.,

Page 275: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

272 STUDY OF MACROMOLECULAR INTERACTIONS IN HEMOSTASIS AND THROMBOSIS

2002). Since the lower limits of the forcewhich must be applied are greatly reduced(because there is no possibility for capil-lary entrapment of the probe by the surface)to 0.01–0.1 nN, the amplitude of oscilla-tion is concomitantly reduced by a signifi-cant degree, allowing far greater control andfar less damage in tip–sample interactions.The use of fluids in the imaging environmentalso eliminates the artifacts and alterationsintroduced by sample dehydration, and itallows for the design and maintenance of spe-cific highly controlled conditions that closelyapproximate the biochemistry and physiol-ogy of the cellular milieu. Imaging undersuch conditions allows for extremely sensi-tive modulation of surface binding energiesby varying the ionic concentration (primar-ily through divalent cations) of both deposi-tion and imaging solutions (Hansma and Hoh,1994; Hansma and Laney, 1996; Thompsonet al., 1996; Morris et al., 1999). More sophis-ticated and subtle details can be controlled,such as the ability to allow for movement andequilibration of molecules as they transitionfrom a three-dimensional solution structureto a two-dimensional surface shape (Busta-mante and Rivetti, 1996; Guthold et al., 1999).This ensures that lowest-energy conforma-tions approximating the solution structure ofthe molecule or molecular assembly are accu-rately preserved, without artificial conforma-tional constraints that might have major con-sequences in studying inter- and intramolecu-lar relationships (Rivetti et al., 1996, 1998).Such an ambient, physiologically bufferedenvironment allows for enzymatic or dynamicprocesses including transcription, DNA loop-ing and bending, and protein-induced con-formational changes to be captured in situ,in dynamic temporal scale, essentially “live”(Guthold et al., 1999; Houchens et al., 2000).There is no other imaging technology that cando all of these things within the specific con-straints that apply to the analysis of biologi-cal systems, while achieving consistently highresolution over an enormous range of scale,complexity, and conditions.

Severalconcernsaboutpotential limitationsduring the initial forays into this techniquehave proved to be of little significance, oreven to be advantageous. The first concern wasthe perception that, due to the unique mannerin which samples are prepared prior to AFMimaging (by binding to atomically flat micasurfaces), conditions would be less physio-logic than those found in conventional “solu-tion/test tube” environments. However, asnoted elsewhere, cellular biochemistry oftentakes place when molecules are localized onsurfaces of one type or another (inner and outersurfaces of membranes, ribosomes, extracel-lular matrix, cytoskeleton, interacting macro-molecular surfaces, etc); therefore, AFM influid tapping mode generates conditions thatare certainly as valid as the proverbial test tube,as a simulacrum of the biological environ-ment (Hansma, 2001; Kindt et al., 2002). Thesecond caveat was the initial disappointmentamong scientists when it appeared that achiev-ing atomic (i.e., angstrom-level) resolution (a“holy grail” of structural biochemistry) mightnot be technically feasible for the AFM whenapplied to biological macromolecules underaqueous physiological conditions. While it hassince been demonstrated that such resolutionis indeed possible, albeit only under severaltypes of highly constrained conditions (Engelet al., 1999; Muller et al., 1999; Engel andMuller, 2000; Stahlberg et al., 2001; Horberand Miles, 2003), it is far more important tonote the incomparable levels of resolution thatare possible for a variety of substrates andconditions thatcannotbe imagedunderanycir-cumstances by such “atomic level” techniquesasx-raycrystallography, nuclear magnetic res-onance, and cryo-EM. These include the abil-ity to achieve nanoscale molecular-level res-olution of objects too large, complex, and/ordelicate to be analyzed by the aforementionedatomic-scale techniques. This size/complexityrange, described as the “mesoscopic” realm(Bustamante and Rivetti, 1996), includesmacromolecules between 10 nm and 200 nmin size, such as multicomponent polypep-tide assemblages, multisubunit enzymes, and

Page 276: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

AFM AND LIPIDS 273

dynamic protein–protein or protein–DNAcomplexes, whose analysis has proven highlyintractable to other techniques. The ability toachieve very high levels of resolution withvery low signal-to-noise ratios, without resort-ing to damaging fixation or staining tech-niques, in ambient physiological buffers andin real time, allows (a) the preservation andanalysis of essential structural, functional,and enzymatic characteristics and (b) delicateinter- and intramolecular relationships thatwere previously unavailable to other forms ofanalysis.

15.3. AFM AND LIPIDS

AFM has been used with much success toimage and analyze the structure and confor-mation of lipid films, surfaces, and bilayershaving a broad range of different chemi-cal compositions. Because such work can bedone in fluid tapping mode utilizing ambi-ent physiological buffers, the experimentalconditions can replicate the cellular environ-ment to a far greater degree than any othertechnique having comparable resolution. Anyanalysis of lipid bilayers that is not con-ducted in a buffered liquid environment willhave serious issues of relevance from a bio-logical standpoint. The interactions of thehydrophobic and hydrophilic components ofthe phospholipid bilayers that form biologi-cal membranes, and aspects of their two- andthree-dimensional structure, are created andconstrained by their relationships with intra-and extracellular fluid interfaces. Conven-tional techniques such as transmission elec-tron microscopy must resort to dehydration,staining, or freeze-fracture techniques thatcan profoundly affect the most basic char-acteristics of lipid bilayers or membranes.These treatments thus remove the aqueouspartner in the powerful and delicate interplayof the hydrophobic interactions that main-tain the integrity of lipid layers. In addition,dehydration may also introduce imaging arti-facts that are difficult to interpret. In con-trast, the AFM is particularly well-suited for

the examination of lipid surfaces, which canbe imaged at very high resolution. Usingfluid tapping mode, the resolution that canbe achieved under certain experimental con-ditions is unmatched, often approximatingatomic-scale (angstrom) levels. In thesecases, protein components within lipid layersmay manifest as “two-dimensional crystals”(Fig. 15.2) (Reviakine et al., 1998; Mulleret al., 1999, 2000; Stahlberg et al., 2001;Rand et al., 2003), which form denselypacked or regularly repeated arrays that areamenable to high-resolution imaging, fol-lowed by Fourier transform image process-ing. Defects that involve the absence ofseveral atoms can sometimes be visualized(Engel and Muller, 2000). By varying thecomposition of the lipids through the intro-duction of different ratios and types of phos-pholipids, and by subsequent addition ofother membrane components such as pro-teins, it is possible to construct an infinitevariety of imaging substrates with a highdegree of physiological fidelity to biologicalmembranes. These lipid surfaces provide fordynamic imaging and force experiments withthe AFM that cannot be achieved with otherexperimental methods.

Mica surfaces prepared with planar phos-pholipid bilayers must be continually sub-merged in buffers in fluid chambersduring incubations and imaging to mini-mize oxidation, which can result in structuraldefects. Fortuitously, lipid bilayer prepara-tions with void defects on atomically flatmica provide a means of measuring layerthicknesses (vertical displacement) via sec-tional analysis. Such baseline measurementsare valuable for future structural analyses andvolumetric measurements of membrane pro-teins and subsequent complexes (i.e., two-dimensional crystals and antibody–cofactortoroids) that form upon their interaction withthe lipid planar surface and other componentsin, or subsequently added to, a fluid envi-ronment. Visualizing dynamic protein–lipidmolecular interactions in an artificial envi-ronment under physiological conditions has

Page 277: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

274 STUDY OF MACROMOLECULAR INTERACTIONS IN HEMOSTASIS AND THROMBOSIS

Figure 15.2. An example of the temporal formation of a two-dimensional crystalline lattice of the anticoagulantprotein AnxA5 on an artificial planar lipid surface composed of 30% phosphatidylserine and 70% phosphatidylcholine(PSPC). (A) The planar lipid membrane (only) showing several defects. (B) Bovine serum albumin is present in thedefects. Between images B and C, AnxA5 had been added. Images C and D display a pebbled and raised surfaceappearance indicative of the anionic phospholipid-containing planar bilayer conformationally changing in responseto AnxA5 and physiological 1.25 mM CaCl2 being present. Within an hour the AnxA5 forms nucleation sitesand coalesces into a two-dimensional crystalline lattice over the planar lipid membrane as shown in the followingtemporal sequence of images E–L. Two-dimensional AnxA5 crystal growth displays junctions termed furrows (blackarrows), which are suspected to represent weak points in the AnxA5 lattice. The images depicted in A–L are original10.0-µm × 10.0-µm topographical amplitude scans with no off-line processing. Vertical scan lines in the left sideof the images result from the “ringing” effect, which is not uncommon at a scan rate of 3.05 kH or higher.

greatly improved the present understandingof morphological changes in lipid bilay-ers during peptide–membrane interactions(Janshoff and Steinem, 2001).

15.4. AFM AND HAEMOSTATICPROTEINS

Fibrinogen is a large multifunctional glyco-protein with a molecular weight of 340 kDa.

Page 278: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

β2GPI COFACTOR 275

In the blood plasma, fibrinogen partici-pates in cardiovascular events through itsconversion by thrombin to the insoluble fib-rin complex, as well as via its binding toactivated platelets leading to cross-linkingand platelet aggregation. We have previouslyimaged purified human fibrinogen bound topoly-L-lysine coated mica by fluid tapping-mode AFM (Taatjes et al., 1997). Usingboth conventional silicon nitride pyramidaltips and electron-beam-deposited carbon tips,we observed that human fibrinogen in fluidappeared as a bi- or trinodular, slightlycurved linear molecule. These fibrinogenmolecules averaged 65.8 nm in length andpresented as a “dumbbell” shape. Thisappearance was in good agreement with priorimages generated of fixed, dehydrated fib-rinogen by transmission electron microscopy(Hall and Slayter, 1959). Similar AFM imag-ing of fibrinogen has been reported byMarchant and co-workers (Marchant et al.,1997; Sit and Marchant, 1999).

15.5. THE ANTIPHOSPHOLIPIDSYNDROME (APS)

APS is an enigmatic autoimmune disorderthat is defined by the presence of circu-lating antiphospholipid (aPL) antibodies inpatients who also have evidence of throm-bosis, embolism, or recurrent spontaneouspregnancy losses (for reviews see Randet al., 1997a; Macik et al., 2001; Warkentinet al., 2003; Rand, 2003). The epitopes rec-ognized by aPL antibodies are believed tobe phospholipid-binding cofactor proteins,of which the major one is ß2-glycoproteinI (ß2GPI). The mechanism for the syn-drome has been particularly elusive sincethe antibodies generated by patients withthe disorder frequently have anticoagulantproperties in vitro. The anticoagulant effectsof the antibodies—known as the “lupus anti-coagulant” (LA) phenomenon—are due totheir avid binding to cofactor–phospholipidcomplexes, thereby reducing the availabil-ity of phospholipid for coagulation reactions.

Remarkably, the positive LA tests are asso-ciated with thrombosis, rather than bleeding,in vivo.

15.6. MECHANISMFOR THE ANTICOAGULANT EFFECTSOF aPL ANTIBODIES

Phospholipids play a critical role in the fol-lowing four enzymatic reactions that lead tothe generation of fibrin, the end product ofblood coagulation, from fibrinogen: (1 and2) the activations of factors IX and X by thetissue factor-VIIa complex, (3) the activationof factor X by the factor IXa–VIII complex,and (4) the activation of factor II by the fac-tor Xa–V complex. aPL antibody-binding todomains of ß2GPI other than its carboxyter-minal phospholipid binding site increases theavidity of the ß2GPI dimers and pentamersfor the phospholipid, thereby reducing theiravailability for coagulation reactions.

15.7. β2GPI COFACTOR

Human β2GPI, also known as apolipoproteinH, is a 42- to 70-kDa membrane-adhesionplasma glycoprotein that was discoveredin 1990 to be a cofactor for anticardi-olipin antibodies from patients with APS(McNeil et al., 1989; Galli et al., 1990; Mat-suura et al., 1992). Under native condi-tions and with no detergent and reducingagents present, β2GPI shows a molecularmass of approximately 320 kDa and is dis-sociable by boiling in 6 M urea (Gushikenet al., 2003). β2GPI in a dissociated, puri-fied form varies slightly in molecular weight(kDa) as indicated by SDS-PAGE, depend-ing on source and preparation(s) (Arvieuxet al., 1996; Mori et al., 1996; Galazka et al.,1998; Harper et al., 1998; Horbach et al.,1998; Ma et al., 2000; Willems et al., 2000;Gushiken et al., 2003). Therefore, purifiedβ2GPI will be presented as a monomer, dimeror an oligomer of a few to several sub-units. In vivo, it has been suggested that

Page 279: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

276 STUDY OF MACROMOLECULAR INTERACTIONS IN HEMOSTASIS AND THROMBOSIS

Figure 15.3. Sequential time-captured three-dimensional amplitude images of dynamic visualization of complexformation between the cofactor β2GPI and human aPLmAb IS2 on lipid (PSPC). (A) PSPC alone on freshly cleavedmica. (B) Following addition of β2GPI, globular structures representing β2GPI multimers form (white arrows).(C–H) Temporal sequence showing the effect of the addition of the monoclonal antibody IS2 to the cofactormultimers present on the planar lipid layer. A conformational rearrangement to form toroid-shaped structuresoccurs. The 1.75-µm × 1.75-µm images were electronically zoomed from original 10-µm × 10-µm images. Thethree-dimensional amplitude images are displayed at 70◦ pitch and 0◦ rotation and have been subjected to fastFourier transformation with the 2d spectrum off-line module of the NanoScope IIIa software.

β2GPI may circulate as a multimer andmay bind in an aggregate form, which maypresent a much needed higher density formfor high-affinity antibody binding (Harperet al., 1998). Harper et al. (1998) presenteddata indicating that β2GPI binding to mem-branes with physiological anionic content isrelatively weak compared to that of plasmacoagulation proteins. Increasing the lipidvesicle content of phosphatidylserine (PS)from 5% to 20% resulted in a twofoldincrease in binding affinity. Comparatively,prothrombin and factor X presented an

approximate 10-fold increase over the samePS concentration range. Anti β2GPI andanti-prothrombin antibodies have been sug-gested as probable risk factors for arte-rial and venous thrombosis [for a recentreview see Galli et al(2003)]. During thelast decade, work from various laboratoriesconvincingly showed that aPL antibodies donot recognize anionic phospholipids with-out a plasma protein cofactor (Fig. 15.3). Inother words, aPL antibodies require anotherplasma protein such as β2GPI, prothrombin,protein C, factor Va, and so on, to bind

Page 280: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

DISRUPTION OF THE AnxA5 ANTICOAGULANT SHIELD 277

to suitable anionic (not necessarily phos-pholipid) surfaces, and the degree of suchbinding is influenced by the binding affini-ties of such phospholipid-binding proteins(Walker, 1981; Krishnaswamy and Mann,1988; Cutsforth et al., 1989; Tait et al., 1989;Harper et al., 1998). These antibodies mayresult in a multiplicity of effects, and thepotential roles of these various aPL antibodyspecificities in the pathophysiology of APShas been previously reviewed (Rand, 2003).These have included mechanisms affect-ing platelet function, prostaglandin synthesis,cell signaling, apoptosis, and many more.Since phospholipids play so many key bio-logic roles, it is not surprising that antibodiesthat bind to them, either directly or via cofac-tor proteins, can, in experimental conditions,perturb virtually any of those activities. Theproblem is determining which of these effectsare relevant to the mechanisms of throm-bosis in the disease state. This difficulty ishighlighted by the lupus anticoagulant phe-nomenon, which has no in vivo consequencewith respect to a bleeding tendency.

Among the questions that remain tobe elucidated are: Do antibodies recogniz-ing epitopes on various plasma proteinsaffect or influence the binding capabilitiesof aPL antibodies? What differences mayexist between artificial phospholipid bilayersand cellular membranes? How might variouselectrolytes and physiological constituentsinfluence binding capabilities? What compo-nents interact with the membrane first? Doesbinding of components happen in the circu-lating environment initially and subsequentlybind to anionic surfaces, and what are thebiologic effects of these interactions?

15.8. ANNEXIN A5

Annexin A5 (AnxA5) (previously known asannexin-V) is a particularly attractive can-didate for studying aPL antibody–cofactorinteractions because it is itself a phospholipid-binding protein with potent anticoagulantproperties. The anticoagulant properties of

the protein are a consequence of its highaffinity for anionic phospholipid (Funakoshiet al., 1987; Tait et al., 1988). AnxA5 formstwo-dimensional crystals on phospholipid sur-faces (Fig. 15.2) (Mosser et al., 1991; Vogeset al., 1994; Reviakine et al., 1998; Randet al., 2003) that shield the phospholipids fromavailability for phospholipid-dependent coag-ulation reactions (Andree et al., 1992). AnxA5is highly expressed by human endothelialcells (Flaherty et al., 1990) and trophoblasts(Kirkun et al., 1994) and is present on the sur-faces of these cells (Kirkun et al., 1994; Randet al., 1997b). AnxA5 is required for the main-tenance of placental integrity in mice (Wanget al., 1999). Infusion of anti-AnxA5 IgG anti-bodies into pregnant animals decreased theavailability of AnxA5 to bind to the tro-phoblast surfaces and caused placental throm-bosis, necrosis, and fetal loss.

15.9. DISRUPTION OF THE AnxA5ANTICOAGULANT SHIELD

The first evidence to suggest that aPL mayaffect AnxA5 was the demonstration, byimmunohistochemistry and ELISA, that theprotein is markedly reduced on apical mem-branes of placental villi exposed to aPL anti-bodies (Rand et al., 1994, 1997b). The anti-bodies also reduced the quantity of AnxA5on cultured trophoblasts and endothelial cellsand accelerated the coagulation of plasmaexposed to these cells (Rand et al., 1997b). Itwas then established via ellipsometry, bindingassays, and immunoassays that aPL reducedthe binding of AnxA5 on reconstituted mem-branes—that is, artificial phospholipid bilay-ers, coated microtiter plates, and phospholipidsuspensions (Rand et al., 1998; Hanly andSmith, 2000).

Based upon the above work, we pro-posed the “Disruption of the AnxA5 Anti-coagulant Shield” hypothesis for thrombo-sis in APS. This hypothesis also offers aplausible explanation for the 50-year-old LAparadox (Rand et al., 1998); a model forthe concept is depicted in Fig. 15.4. Within

Page 281: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

278 STUDY OF MACROMOLECULAR INTERACTIONS IN HEMOSTASIS AND THROMBOSIS

this construct, aPL antibodies (mediated bycofactors) can increase the availability ofphospholipid by causing defects in AnxA5crystallization. Remarkably, the combinationof two competing phospholipid-binding moi-eties—aPL antibodies and AnxA5—does noteliminate the availability of phospholipids forcoagulation reactions. Rather, the presence ofthe antibodies causes defects in the AnxA5

crystalline array, thereby increasing the expo-sure of unshielded phospholipids, which, inturn, promote coagulation reactions. Firm evi-dence for the concept that aPL antibodies canactually disrupt the AnxA5 anticoagulant wasprovided by AFM imaging (Rand et al., 2003)(see below).

The above studies were followed by aninitial translational study and showed thatdisruption of AnxA5 binding and reduction

A

C

B

D

FIBRIN FORMATION

FIBRIN FORMATION FIBRIN FORMATION

IIX

X, IXTF-VIIa IXa-VIIIa

Phospholipid Bilayer

Phospholipid Bilayer Phospholipid Bilayer

Phospholipid Bilayer

(−) Annexin-V (+) Annexin-V

Xa-Va

IIXa-Va

IIXa-Va

(+) aPL (+) Annexin-V

(+) aPLß2GPIß2GPI

Figure 15.4. Model for the mechanisms of the “lupus anticoagulant effect” and for a “lupus procoagulant effect.”(A) Anionic phospholipids serve as potent cofactors for the assembly of three different coagulation complexes—thetissue factor (TF)–VIIa complex, the IXa–VIIIa complex, and the Xa–Va complex—and thereby accelerate bloodcoagulation. (B) AnxA5, in the absence of aPL antibodies, is a potent anticoagulant that forms a crystal latticeover the anionic phospholipid surface, shielding it from availability for assembly of the phospholipid-dependentcoagulation complexes. (C) In the absence of AnxA5, aPL antibody–ß2GPI complexes can prolong the coagulationtimes, compared to control antibodies. This occurs via antibody recognition of domains I or II on the ß2GPI, whichresults in dimers and pentamers of antibody–ß2GPI complexes having high affinity for phospholipid via domain V.These high-affinity complexes serve to reduce the access of coagulation factors to anionic phospholipids. This couldresult in a “lupus anticoagulant” effect in conditions where there are limiting quantities of anionic phospholipids. (D)In the presence of AnxA5, antiphospholipid antibodies, either directly or via interaction with protein–phospholipidcofactors, disrupt the ability of AnxA5 to form two-dimensional crystals on the phospholipid surface. This results in anet increase in the amount of anionic phospholipid available for promoting coagulation reactions. (From Biochimicaet Biophysica Acta 1498:169–173, 2000.)

Page 282: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

CONTRIBUTIONS OF AFM TO THE STUDY OF THE ANTIPHOSPHOLIPID SYNDROME 279

of its anticoagulant effect could also beobserved in plasmas from patients withaPL antibodies (Rand, 2003). The aPL-mediated effects on AnxA5 were confirmedby other investigators using phospholipid-coated microtiter plates (Hanly and Smith,2000) and with platelets (Tomer, 2002).

15.10. CONTRIBUTIONS OF AFMTO THE STUDY OF THEANTIPHOSPHOLIPID SYNDROME

To investigate the seemingly contradictoryeffects of aPL antibodies and annexin A5,

we reconstituted an in vitro model of theAnxA5 two-dimensional crystal on a phos-pholipid bilayer, to be imaged by tappingmode AFM in fluid. Mica disks were incu-bated with a 30% phosphatidylserine:70%phosphatidylcholine phospholipid mixture. Aplanar lipid layer formed after 60 min atroom temperature. To this planar lipid layer,purified human AnxA5 was added, and two-dimensional crystals formed on the lipidlayer over the course of an hour (Fig. 15.2).This two-dimensional AnxA5 crystal struc-ture was observed to grow from nucleationsites until the separately forming crystalsmet at furrow sites, as previously described

Figure 15.5. Effect of human monoclonal antibodies on previously formed AnxA5 crystal structure. (A, C) AnxA5surface in the presence of monoclonal antibodies CL15 and CL1, respectively, prior to the addition of β2GPI cofactor.Note the two-dimensional lattice structure of the AnxA5. After the addition of the required β2GPI cofactor, CL15grossly disrupts the AnxA5 crystal lattice (B), forming large furrows where the AnxA5 has been displaced by theantibody. Note that in C, the addition of monoclonal antibody CL1 to the AnxA5 crystal lattice resulted in thedeposition of some globules (arrows), but the crystal structure remains intact. When the β2GPI cofactor is added,the globules enlarge (arrows in D) and the AnxA5 crystal structure is minimally disrupted (asterisk). Images B–Dare height images with off-line zero-flatten and low-pass filtering applied; image in A is an amplitude image withno processing. (From American Journal of Pathology 163:1193–1200, 2003.)

Page 283: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

280 STUDY OF MACROMOLECULAR INTERACTIONS IN HEMOSTASIS AND THROMBOSIS

(Reviakine et al., 1998). The thickness ofthe formed two-dimensional AnxA5 crystalwas measured to be 2.44 ± 0.13 nm on topof the planar lipid layer (Rand et al., 2003).After confirming that AnxA5 formed a crys-tal lattice on top of the planar lipid layerattached to mica, we sought to investigatethe effect of several human antiphospholipidantibodies on the integrity of this crystallayer. The addition of ß2GPI cofactor alonehad no effect on the crystalline lattice struc-ture (Fig. 15.5). However, the addition ofa monoclonal antiphospholipid antibody inconjunction with the cofactor led to dis-ruption of the crystal lattice. The sever-ity and overall appearance of the disruptionwas determined by the antibody applied. For

instance, incubation with the antibody desig-nated CL15 led to a gross disruption of thecrystal lattice (Fig. 15.5), while incubationwith the antibody designated IS3 resultedin the appearance of circular pits and somevacancy defects (Fig. 15.6). This mechanismcan also be well-visualized by analyzing aseries of height images during the course ofan experiment. This is depicted in Fig. 15.7,where a series of height images, togetherwith height measurements, are shown. As theAnxA5 crystal forms over the planar lipidlayer, the height increases from 4.503 nmto 6.949 nm. Following addition of an aPLantibody, the height measurement decreasesto 4.372 nm, representing the removal ofthe AnxA5 crystal layer from the underlying

Figure 15.6. Effect of monoclonal antibody IS3 on preformed AnxA5 crystal two-dimensional lattice. When IS3and β2GPI were added to the AnxA5 crystal lattice formed on a planar lipid layer, circular pits (arrows in A and B)appeared, indicating disruptions in the crystal lattice. A representative pit is shown at higher magnification in the insetto A and B. At higher resolution (C,D), multiple vacancy defects (small, round dark holes) in the crystalline latticeare apparent. The amplitude images (A,C) were processed with a 1 × convolution filter, and the height images (B,D)were processed by a zero-order flatten filter. Original magnifications: 10-µm × 10-µm scan (A,B); 500 nm × 500 nm(C,D). (From American Journal of Pathology 163:1193–1200, 2003.)

Page 284: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

CONTRIBUTIONS OF AFM TO THE STUDY OF THE ANTIPHOSPHOLIPID SYNDROME 281

Figure 15.7. Temporal height mode images of the dynamic visualization of the structural alteration of AnxA5crystalline lattice on a planar lipid layer by cofactor β2GPI and human aPLmAb IS4. (A) An isolated lipid (PSPC)patch on freshly cleaved mica. (B) The first of three steps of AnxA5 formation; the “pebbled” granulated appearancerepresents the binding of monomeric AnxA5 molecules to the planar lipid surface. Membrane trimers begin formingduring the second step in the process (C). The third step (D) of two-dimensional crystalline arrays of trimersrepresents complete coalescence with no visible furrows. Note the increase in mass thickness of the indicated lipidpatch in A–D, as the AnxA5 crystal forms on top of the lipid layer. (E) Following the addition of β2GPI cofactorand incubation for 1 h, no significant change in vertical measurements is apparent. (F) Finally, following subsequentaddition of aPLmAb IS4, a dramatic decrease in lipid/AnxA5 patch thickness is revealed, representing removal ofthe AnxA5 crystal by the aPL/cofactor complex. While comparing the images in A–F, note the differences in surfaceimage contrast; surface is a light gray (A) and changes to a “pebbled” white (B), to all white (D), and returns toa grayish tone (F). The contrast is a representation of the vertical height: The more toward the white the imagetends, the greater the height (as demonstrated by the linear height measurements represented below each image). The2.5-µm × 2.5-µm images were electronically zoomed from 10-µm × 10-µm scanned originals. No other processingof images was performed. The corresponding cross-sectional profile plots (below each image) were acquired utilizingthe off-line Sectional Analysis module of the NanoScope IIIa software.

Page 285: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

282 STUDY OF MACROMOLECULAR INTERACTIONS IN HEMOSTASIS AND THROMBOSIS

planar lipid layer. This change in height canalso be visualized as a change in gray levelon the images themselves (Fig. 15.7). Addi-tionally, the incubation of a planar lipid layerwith aPL antibodies and cofactor preventedthe formation of a crystal lattice upon thesubsequent addition of AnxA5.

These studies represent the first morpho-logical evidence that aPL antibodies dis-rupt the AnxA5 anticoagulant shield. Thismay therefore represent a pathophysiologi-cal mechanism for thrombosis and sponta-neous pregnancy loss in the aPL syndrome,as depicted in the model in Fig. 15.4.

15.11. SUMMARY

AFM offers a novel approach for investigat-ing macromolecular interactions in a tempo-ral fashion at high resolution. In the case ofthe enigmatic disorder known as the APS,AFM has provided novel insights into themechanism by which aPL antibodies mayprovoke a thrombotic condition. Althoughnot intended as a diagnostic, clinical tool, theAFM should continue to provide the meansby which valuable mechanistic informationunderlying pathological conditions may begarnered.

ACKNOWLEDGMENT

This work was supported in part by a grantfrom the National Heart, Lung, and BloodInstitute to JHR (R01 HL061331).

REFERENCES

Abdelhady, H., Allen, S., Davies, M., Roberts, C.,Tendler, S. and Williams, P. (2003). Direct real-time molecular scale visualization of the degra-dation of condensed DNA complexes exposed toDNase I. Nucleic Acids Res. 31:4001–4005.

Allemand J.-F., Bensimon, D. and Croquette, V.(2003). Stretching DNA and RNA to probe

their interactions with proteins. Curr. Opin.Struct. Biol. 13:266–274.

Andree, H. A. M., Stuart, M. C., Hermens, W. T.,Reutelingsperger, C. P., Hemker, H. C., Fred-erik, P. M., and Willems, G. M. (1992). Clus-tering of lipid-bound annexin V may explainits anticoagulant effect. J. Biol. Chem. 267:17907–17912.

Arvieux, J., Darnige, L., Hachulla, E., Roussel, B.,Bensa, J. C., and Colomb, M. G. (1996). Speciesspecificity of anti-beta 2 glycoprotein I autoan-tibodies and its relevance to anticardiolipinantibody quantitation. Thromb. Haemost. 75:725–730.

Betzig, E., Lewis, A., Harootuniam, A., Isaac-son, M., and Kratschmer E. (1986). Near-fieldscanning optical microscopy (NSOM): Devel-opment and biophysical applications. Biophys.J. 49:269–279.

Binnig, G., Rohrer, H., Gerber, C. and Weibel, E.(1982a). Tunneling through a controllable vac-uum gap. Phys. Rev. Lett. 40:178–180.

Binnig, G., Rohrer, H., Gerber, C., and Weibel, E.(1982b). Surface studies by scanning tunnelingmicroscopy. Phys. Rev. Lett. 49:57–61.

Binnig, G., Quate, C. F., and Gerber, C. (1986).Atomic force microscope. Phys. Rev. Lett.56:930–933.

Bustamante, C., and Keller, D. (1994). Scanningforce microscopy in biology. Phys. Today48:32–38.

Bustamante, C. and Rivetti, C. (1996). Visualizingprotein–nucleic acid interactions on a largescale with the scanning force microscope. Annu.Rev. Biophys. Biomol. Struct. 25:395–429.

Bustamante, C., Erie, D. A., and Keller, D. (1994).Biochemical and structural applications of scan-ning force microscopy. Curr. Opin. Struct. Biol.4:750–760.

Bustamante, C., Rivetti, C., and Keller, D. (1997).Scanning force microscopy under aqueous solu-tions. Curr. Opin. Struct. Biol. 7:709–716.

Bustamante, C., Guthold, M., Zhu, X., andYang, G. (1999). Facilitated target location onDNA by individual Escherichia coli RNApolymerase molecules observed with the scan-ning force microscope operating in liquid.J. Biol. Chem. 274:16665–16668.

Bustamante, C., Smith, S., Liphardt, J., andSmith D. (2000). Single molecule studies of

Page 286: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 283

DNA mechanics. Curr. Opin. Struct. Biol.10:279–285.

Cutsforth, G. A., Whitaker, R. N., Hermans, J.,and Lentz, B. R. (1989). A new model todescribe extrinsic protein binding to phos-pholipid membranes of varying composition:Application to human coagulation proteins.Biochemistry 28:7453–7461.

Dunlap, D., Maggi, A., Soria, M., and Monaco, L.(1997). Nanoscopic structure of DNA con-densed for gene delivery. Nucleic Acids Res.25:3095–3101.

Durig, U., Pohl, D. W., and Rohner, F. (1986).Near-field optical-scanning microscopy. J ApplPhys 59:3318–3327.

Engel, A., and Muller, D. J. (2000). Observ-ing single biomolecules at work with theatomic force microscope. Nature Struct. Biol.7:715–718.

Engel, A., Lyubchenko, Y., and Muller, D. (1999).Atomic force microscopy: A powerful tool toobserve biomolecules at work. Trends Cell Biol.9:77–80.

Flaherty, M. J., West, S., Heimark, R. L.,Fujikawa, K., and Tait, J. F. (1990). Placentalanticoagulant protein-I: Measurement in extra-cellular fluids and cells of the hemostatic sys-tem. J. Lab. Clin. Med. 115:174–181.

Funakoshi, T., Heimark, R. L., Hendrickson,L. E., McMullen, B. A., and Fujikawa, K.(1987). Human placental anticoagulant pro-tein: Isolation and characterization. Biochem-istry 26:5572–5578.

Galazka, M., Keil, L. B., Kohles, J. D., Li, J.,Kelty, S. P., Petersheim, M., and DeBari, V. A.(1998). A stable, multi-subunit complex ofbeta2glycoprotein I. Thromb. Res. 90:131–137.

Galli, M., Comfurius, P., Maassen, C., Hemker,H. C., de Baets, M. H., van Breda-Vriesman,P. J., Barbui, T., Zwaal, R. F., and Bevers,E. M. (1990). Anticardiolipin antibodies (ACA)directed not to cardiolipin but to a plasma pro-tein cofactor. Lancet 335:1544–1547.

Galli, M., Luciani, D., Bertolini, G., and Bar-bui, T. (2003). Anti-beta 2-glycoprotein I,antiprothrombin antibodies, and the risk ofthrombosis in the antiphospholipid syndrome.Blood 102:2717–2723.

Gushiken, F. C., Le, A., Arnett, F. C., and Thi-agarajan, P. (2003). Polymorphisms of beta2-glycoprotein I: Phospholipid binding andmultimeric structure. Thromb Res 108:175–180.

Guthold, M., Xingshu, Z., Rivetti, C., Yang, G.,Thompson, N., Kasas, S., Hansma, H., Smith,B., Hansma, P., and Bustamante, C. (1999).Direct observation of one-dimensional diffusionand transcription by E. coli RNA polymerase.Biophys. J. 77:2284–2294.

Hall, C. E., and Slayter, H. S. (1959). The fib-rinogen molecule: Its size, shape and modeof polymerization. J. Biophys. Biochem. Cytol.5:11–16.

Han, W., Mou, J., Sheng, J., and Shao, Z. (1995).Cryo-atomic force microscopy: A new app-roach for biological imaging at high resolution.Biochemistry 34:8215–8220.

Han, W., Lindsay, S. M., and Jing, T. (1996). Amagnetically driven oscillating probe micro-scope for operation in liquids. Appl. Phys. Lett.69:1–3.

Hanly, J. G., and Smith, S. A. (2000). Anti-beta2-glycoprotein I (GPI) autoantibodies, annexin Vbinding and the anti-phospholipid syndrome.Clin. Exp. Immunol. 1120:537–543.

Hansma, H. (2001). Surface biology of DNAby atomic force microscopy. Annu. Rev. Phys.Chem. 52:71–92.

Hansma, H., and Ho, J. (1994). Biomolecularimaging with the atomic force microscope. Annu.Rev. Biophys. Biomol. Struct. 23:115–139.

Hansma, H. G., and Laney, D. E. (1996). DNAbinding to mica correlates with cationic radius:Assay by atomic force microscopy. Biophys. J.70:1933–1939.

Hansma, H., Bezanilla, M., Zenhausen F., Adrian,M., and Sinsheimer, R. (1993). Atomic force

microscopy of DNA in aqueous solutions.Nucleic. Acids. Res. 21:505–512.

Hansma, H., Golan, R., Hsieh, W., Daubendiek, S.,and Kool, E. (1999). Polymerase activities andRNA structures in the atomic force microscope.J. Struct. Biol. 127:240–247.

Harper, M. F., Hayes, P. M., Lentz, B. R., andRoubey, R. A. (1998). Characterization of beta-2-glycoprotein I binding to phospholipid mem-branes. Thromb. Haemost. 80:610–614.

Horbach, D. A., van Oort, E., Tempelman, M. J.,Derksen, R. H., and de Groot, P. G. (1998). Theprevalence of a non-phospholipid-binding form

Page 287: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

284 STUDY OF MACROMOLECULAR INTERACTIONS IN HEMOSTASIS AND THROMBOSIS

of beta2-glycoprotein I in human plasma—consequences for the development of anti-beta2-glycoprotein I antibodies. Thromb. Haemost.80:791–797.

Horber, J., and Miles, M. (2003). Scanning probeevolution in biology. Science 302:1002–1005.

Houchens, C. R., Montigny, W., Zeltser, L., Dai-ley, L., Gilbert, J. M., and Heintz, N. H. (2000).The dhfr oribeta-binding protein RIP60 contains15 zinc fingers: DNA binding and looping by thecentral three fingers and an associated proline-rich region. Nucleic Acids Res. 28:570–581.

Israelachvili, J. (1985). Intermolecular and Sur-face Forces, Academic Press, New York.

Janshoff, A., and Steinem, C. (2001). Scanningforce microscopy of artificial membranes.ChemBioChem 2:798–808.

Kindt, J., Sitko, J., Pietrasanta, L., Oroudjev, E.,Becker, N., Viani, M. and Hansma, H. (2002).Methods for biological probe microscopy inaqueous fluids. In: Atomic Force Microscopyin Cell Biology: Methods in Cell Biology,Vol. 68 (B. P. Jena and J. K. Horber, eds.),pp. 214–229, Academic Press, New York.

Kirkun, G., Lockwood, C. J., Wu,, X. X., Zhou,X. D., Guller, S., Calandri, C., Guha, A.,Nemerson, Y., and Rand, J. H. (1994). Theexpression of the placental anticoagulant pro-tein, annexin V, by villous trophoblasts:Immunolocalization and in vitro regulation.Placenta 15:601–612.

Krishnaswamy, S., and Mann, K. G. (1988). Thebinding of factor Va to phospholipid vesicles.J. Biol. Chem. 263:5714–5723.

Ma, K., Simantov, R., Zhang, J. C., Silverstein, R.,Hajjar, K. A., and McCrae, K. R. (2000). Highaffinity binding of beta 2-glycoprotein I tohuman endothelial cells is mediated by annexinII. J. Biol. Chem. 275:15541–15548.

Macik, B. G., Rand, J. H., and Konkle, B. A.(2001). Thrombophilia: What’s a practitionerto do? Am. Soc. Hematol. Educ. Program322–338.

Marchant, R. E., Barb, M. D., Shainoff, J. R.,Eppell, S. J., Wilson, D. L., and Siedlecki,C. A. (1997). Three dimensional structureof human fibrinogen under aqueous condi-tions visualized by atomic force microscopy.Thromb. Haemost. 77:1048–1051.

Marchant, R. E., Kang, I., Sit, P. S., Zhyou, Y.,Todd, B. A., Eppell, S. J., and Lee, I. (2002).

Molecular views and measurements of hemo-static processes using atomic force microscopy.Curr. Protein Pept. Sci. 3:249–274.

Matsuura, E., Igarashi, Y., Fujimoto, M.,Ichikawa,K., Suzuki, T., Sumida T, Yasuda, T., and

Koike, T. (1992) Heterogeneity of anticardi-olipin antibodies defined by the anticardiolipincofactor. J. Immunol. 148:3885–3891.

McNeil, H. P., Chesterman, C. N., and Krilis,S. A. (1989). Anticardiolipin antibodies andlupus anticoagulants comprise separate anti-body subgroups with different phospholipidbinding characteristics. Br. J. Haematol. 73:506–513.

Meyer, G., and Amer, N. (1988). Novel approachto atomic force microscopy. Appl. Phys. Lett.53:1045–1047.

Montigny, W. J., Houchens, C. R., Illenye, S.,Gilbert, J., Coonrod, E., Chang, Y. C., andHeintz, N. H. (2001). Condensation by DNAlooping facilitates transfer of large DNAmolecules into mammalian cells. Nucleic AcidsRes. 29:1982–1988.

Montigny, W. J., Phelps, S. F., Illenye, S., andHeintz, N. H. (2003). Parameters influencinghigh-efficiency transfection of bacterial arti-ficial chromosomes into cultured mammaliancells. Biotechniques 35:796–807.

Mori, T., Takeya, H., Nishioka, J., Gabazza, E. C.,and Suzuki, K. (1996). Beta 2-glycoprotein Imodulates the anticoagulant activity of activatedprotein C on the phospholipid surface. Thromb.Haemost. 75:49–55.

Morris V., Kirby, A., and Gunning, A. (1999).Atomic Force Microscopy for Biologists, Impe-rial College Press, London.

Mosser, G., Ravavat, C., Freyssinet, J. M., andBrisson, A. (1991). Sub-domain structure oflipid-bound annexin-V resolved by electronimage analysis. J. Mol. Biol. 217:241–245.

Mou, J., Yang, J. and Shao, Z. (1993) An opti-cal detection low temperature atomic forcemicroscope at ambient pressure for biologicalresearch. Rev. Sci. Instrum. 64:1483–1488.

Muller, D. J., Fotiadis, D., Scheuring, S., Muller,S. A., and Engel, A. (1999). Electrostaticallybalanced subnanometer imaging of biologicalspecimens by atomic force microscope. Bio-phys. J. 76:1101–1111.

Muller, D. J., Heymann, J. B., Oesterhelt, F.,Moller, C., Gaub, H., Buldt, G., and Engel, A.

Page 288: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

REFERENCES 285

(2000). Atomic force microscopy of nativepurple membrane. Biochim. Biophys. Acta1460:27–38.

Putman, C., Van der Werf, K., De Grooth, B., VanHulst, N. and Greve, J. (1994). Tapping modeatomic force microscopy in liquid. Appl. Phys.Lett. 64:2454–2456.

Rand, J. H. (2003). The antiphospholipid syn-drome. Annu. Rev. Med. 54:409–424.

Rand, J. H., Wu, X. X., Guller, S., Gil, J.,Guha, A, Scher, J., and Lockwood, C. J. (1994).Reduction of annexin-V (placental anticoag-ulant protein-I) on placental villi of womenwith antiphospholipid antibodies and recurrentspontaneous abortion. Am. J. Obstet. Gynecol.171:1566–1572.

Rand, J. H., Wu, X. X., Andree, H. A., Lock-wood, C. J., Guller, S., Scher, J., and Harpel,P. C. (1997a). Pregnancy loss in the anti-phospholipid-antibody syndrome—a possiblethrombogenic mechanism. N. Engl. J. Med.337:154–160.

Rand, J. H., Wu, X. X., Guller, S., Scher, J.,Andree, H. A. M., and Lockwood, C. J.(1997b). Antiphospholipid immunoglobulin Gantibodies reduce annexin-V levels on syncy-tiotrophoblast apical membranes and in culturemedia of placental villi. Am. J. Obstet. Gynecol.177:918–923.

Rand, J. H., Wu, X. X., Andree, H. A. M.,Alexander Ross, J. B., Rosinova, E., Gascon-Lema, M. G., Calandri, C., and Harpel, P. C.(1998). Antiphospholipid antibodies accelerateplasma coagulation by inhibiting annexin-Vbinding to phospholipids: A “lupus procoagu-lant” phenomenon. Blood 92:1652–1660.

Rand, J. H., Wu, X.-X., Quinn, A. S., Chen, P. P.,McCrae, K. R., Bovill, E. G., and Taatjes, D. J.(2003). Human monoclonal antiphospholipidantibodies disrupt the annexin A5 anticoag-ulant crystal shield on phospholipid bilayers.Evidence from atomic force microscopy andfunctional assay. Am. J. Pathol. 163:1193–1200.

Reviakine, I., Bergsma-Schutter, W., and Bris-son, A. (1998). Growth of protein 2-D crys-tals on supported planar lipid bilayers imagedin situ by AFM. J. Struct. Biol. 121:356–361.

Rivetti, C., Guthold, M., and Bustamante, C.(1996). Scanning force microscopy of DNA

deposited onto mica: Equilibration versus ki-netic trapping studied by statistical polymerchain analysis. J. Mol. Biol. 264:919–932.

Rivetti, C., Walker, C., and Bustamante, C. (1998).Polymer chain statistics and conformationalanalysis of DNA molecules with bends orsections of different flexibility. J. Mol. Biol.280:41–59.

Rugar, D., and Hansma, P. K. (1990). Atomicforce microscopy. Phys. Today 43:23–30.

Shao, Z., and Zhang, Z. (1996). Biological cryoatomic force microscopy: A brief review.Ultramicroscopy 66:141–152.

Sit, P. S., and Marchant, R. E. (1999). Surface-dependent conformations of human fibrino-gen observed by atomic force microscopyunder aqueous conditions. Thromb. Haemost.82:1053–1060.

Stahlberg, H., Fotiadis, D., Scheuring, S., Remigy,H., Braun, T., Mitsuoka, K., Fujiyoshi, Y., andEngel, A. (2001). Two-dimensional crystals: Apowerful approach to assess structure, functionand dynamics of membrane proteins. FEBS Lett.504:166–172.

Stroh, C., Wang, H., Bash, R., Ashcroft, B., Nel-son, J., Gruber, H., Lohr, D., Lindsay, S. andHinterdorfer, P. (2004). Single-molecule recog-nition imaging microscopy. Proc. Natl. Acad.Sci. 34:12503–12507.

Taatjes, D. J., Quinn, A. S., Jenny, R. J., Hale, P.,Bovill, E. G., and McDonagh, J. (1997). Ter-tiary structure of the hepatic cell protein fib-rinogen in fluid revealed by atomic forcemicroscopy. Cell Biol. Int. 21:715–726.

Taatjes, D. J., Quinn, A. S., Lewis, M. R., andBovill, E. G. (1999). Quality assessment ofatomic force microscopy probes by scanningelectron microscopy: Correlation of tip struc-ture with rendered images. Microsc. Res. Tech.44:312–326.

Tait, J. F., Gibson, D., and Fujikawa, K. (1989).Phospholipid binding properties of human pla-cental anticoagulant protein-I, a member ofthe lipocortin family. J. Biol. Chem. 264:7944–7949.

Tait, J. F., Sakata, M., McMullen, B. A., Maio,C. H., Funakoshi, T., Hendickson, L. E., andFujikawa, K. (1988). Placental anticoagulantproteins: Isolation and comparative characteri-zation of four members of the lipocortin family.Biochemistry 27:6268–6276.

Page 289: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

286 STUDY OF MACROMOLECULAR INTERACTIONS IN HEMOSTASIS AND THROMBOSIS

Taylor, M. (1993). Dynamics of piezoelectric tubescanners for scanning probe microscopy. Rev.Sci. Instrum. 64:154–158.

Thompson, N., Kasas, S., Smith, B., Hansma, H.and Hansma, P. (1996). Reversible bindingof DNA to mica for imaging. Langmuir12:5905–5908.

Tomer, A. (2002). Antiphospholipid antibodysyndrome: Rapid, sensitive, and specific flowcytometric assay for determination of anti-platelet phospholipid autoantibodies. J. Lab.Clin. Med. 139:147–154.

Voges, D., Berendes, R., Burger, A., Demange, P.,Baumeister, W., and Huber, R. (1994). Three-dimensional structure of membrane-boundannexin V. A correlative electron microscopy–X-ray crystallography study. J. Mol. Biol. 238:199–213.

Walker, F. J. (1981). Regulation of activated pro-tein C by protein S. The role of phospho-lipid in factor Va inactivation. J. Biol. Chem.256:11128–11131.

Wang, X., Campos, B., Kaetzel, M. A., and Ded-man, J. R. (1999). Annexin V is critical in themaintenance of murine placental integrity. Am.J. Obstet. Gynecol. 180:1008–1016.

Warkentin, T. E., Aird, W. C., and Rand, J. H.(2003). Platelet–endothelial interactions: Sep-sis, HIT, and antiphospholipid syndrome. He-matology (Am. Soc. Hematol. Educ. Program)497–519.

Willems, G. M., Janssen, M. P., Comfurius, P.,Galli, M., Zwaal, R. F., and Bevers, E. M.(2000). Competition of annexin V and anti-cardiolipin antibodies for binding to phos-phatidylserine containing membranes. Biochem-istry 39:1982–1989.

Page 290: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

INDEX

Actin regulation:atomic force microscopic imaging, inverted

optical microscopic assembly, 140–141cell secretion and, 8–17

Adenylate cyclase, signal transduction, 55–56Adhesion mapping:

microbial cell surface imaging, atomic forcemicroscopy analysis, 81–82

sonicated unilamellar vesicle bilayers,192–197

Adsorption strategies:microbial cell surface imaging, atomic force

microscopy analysis, 71polymer-based nanodrug delivery, 120–121

Alkanethiol self-assembled monolayers,microbial cell surface imaging:

atomic force microscopy analysis, 71probe surface chemistry, 80–81

Ambisome, nanodrug delivery with, 131–132Annexin A5 (AnxA5), antiphospholipid

syndrome, 277–279atomic force microscopy, 279–282

Antibiotics, microbial cell surface effects, atomicforce microscopy analysis, 78–79

Anticoagulant effects, antiphospholipidsyndrome, 275

annexin A5, 277–279Antigen presenting cell (APC), avidity

modulation, basic principles, 169–171Antiphospholipid syndrome (APS), atomic force

microscopic imaging:annexin A5 shield, 277–279aPL antibodies, anticoagulation effects, 275clinical applications, 279–282β2GPI cofactor, 275–277hemostatic proteins, 274–275lipid analysis, 273–274theoretical background, 267–272

Apolipoprotein (APL) antibodies:anticoagulant effects, 275antiphospholipid syndrome, annexin A5,

278–279

Aquaporins:microbial cell surface imaging, atomic force

microscopy analysis, 74–75immobilization strategies, 71

secretory vesicle content expulsion, basicprinciples, 37–38, 40

Array detectors, atomic force microscopycantilevers, 240–241

Atomic-based membrane fusion processes,calcium participation in, 27–34

Atomic force microscopy (AFM):antiphospholipid syndrome:

annexin A5 shield, 277–279aPL antibodies, anticoagulation effects, 275clinical applications, 279–282β2GPI cofactor, 275–277hemostatic proteins, 274–275lipid analysis, 273–274theoretical background, 267–272

biomolecular motion:basic principles, 221–223biological applications, 242–243cantilever properties:

limits, 232resonant frequency, 232–233small cantilevers, 233–235thermal noise, 235–237

fast, low-noise detector, 239–241fast feedback loop, 242fast scanner, 241–242focused spot size, 237–239imaging speed, 228–231

feedback limit, 231lateral dimension reduction, 229–230tracking mechanisms, 228–229video-rate imaging, 231

time-resolved imaging, 223–228alignment, 225–227moving images, 227–228

Force Microscopy: Applications in Biology and Medicine, edited by Bhanu P. Jena and J.K. Heinrich Horber.Copyright 2006 John Wiley & Sons, Inc.

287

Page 291: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

288 INDEX

Atomic force microscopy (AFM) (continued )p53-DNA dynamic interactions, 224–225recording sequences, 223–224

cytogenetics applications:C-banding technique, 257genetic material imaging techniques,

250–253metaphase chromosome karyotyping,

253–257metaphase chromosome three-dimensional

analysis, 257–259nanoextraction, 259–262nanomanipulation and dissection, 259theoretical background, 249–250

electrophysiological techniques, 137–150inner ear tissue sections, 144–150inverted optical microscope, 137–141

growth hormone fusion pore structure anddynamics, 59–60

growth hormone secretory vesicles, 61–63historical evolution of, 2–3leukocyte adhesion molecules, 160

avidity modulation:adhesive force measurements, 172–173basic principles, 169–171cells and reagents, 171clinical applications, 176–178elasticity measurements, 173integrin lateral redistribution, 173–175ionomycin/thapsigargin stimulation,

175–176protein immobilization, 1723A9 cell crosslinking, 171–172

integrin activation, 162–166single-molecule unbinding, 160–166

lipid bilayers, thickness and micromechanicalproperties, 181–197

supported thickness, 182–183lipid membrane preparation, porosome

reconstitution, 4microbial cell surface property analysis,

69–90bacillus S-layers, 73basic principles, 69–70cell surface layer imaging, 73–75elasticity, 85–89

layers stretching, 87single cell stretching, 87–89single macromolecules, 86–87

external agent effects, 78–79future applications, 89–90hexagonally packed intermediate layer, 73imaging techniques, 70–79

immobilization strategies, 71selection criteria, 71–72substrate requirements, 70–71

microbial biofilms, 75–76nanostructures, 76–78

physical properties measurement, 79–89force-distance curves, 80force measurements, 80–81functionalized probes, 80–81

physicochemical properties and molecularinteractions, 81–85

adhesion mapping, 81–82cell probe technique, 85surface charges and electrostatic

interactions, 83–85surface energy and solvation interactions,

82–83physiological changes, 78porin crystals, 74–75purple membranes, 73–74

pancreatic acinar cell isolation, 3–4pancreatic plasma membrane isolation, 4plant cell wall imaging, 96–97

cellulose, 99–100lignins, 103–108pectins, 97–99

porosome imaging, 4–5structural analysis, 6–17

secretory vesicle content expulsion, vesicleswelling in pancreatic acinar cells, 38–44

SNARE-induced membrane fusion, bilayerinteraction and conducting channelformation, 26–27

soft tissue imaging, swollen polymer surfaces,214–218

sonicated unilamellar vesicle:bilayer thickness and morphology, 187–191micromechanical properties, 191–197

synaptosome/synaptosomal/synaptic vesicleisolation, 4

Avidity modulation, leukocyte adhesionmolecules:

adhesive force measurements, 172–173basic principles, 169–171cells and reagents, 171clinical applications, 176–178elasticity measurements, 173integrin lateral redistribution, 173–175ionomycin/thapsigargin stimulation, 175–176protein immobilization, 1723A9 cell crosslinking, 171–172

Bacillus S-layers, microbial cell surface imaging,atomic force microscopy analysis, 73

immobilization strategies, 71Bell single-molecule unbinding model, leukocyte

adhesion molecules, 160–166integrin activation, 165–166

Bending modulus, sonicated unilamellar vesiclebilayer micromechanics, 192–197

“Bimetallic strip effect,” atomic forcemicroscopy cantilevers, 233

Page 292: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

INDEX 289

Biocompatibility, polymer-based nanodrugdelivery, 131–132

Biocorrosion studies, microbial cell surfaceimaging, atomic force microscopy analysis,76

Biofilms, microbial cell surface imaging, atomicforce microscopy analysis, 75–76

Biomembrane force probe (BFP), leukocyteadhesion molecules, 160

Biomolecular motion, atomic force microscopy(AFM):

basic principles, 221–223biological applications, 242–243cantilever properties:

limits, 232resonant frequency, 232–233small cantilevers, 233–235thermal noise, 235–237

fast, low-noise detector, 239–241fast feedback loop, 242fast scanner, 241–242focused spot size, 237–239imaging speed, 228–231

feedback limit, 231lateral dimension reduction, 229–230tracking mechanisms, 228–229video-rate imaging, 231

time-resolved imaging, 223–228alignment, 225–227moving images, 227–228p53-DNA dynamic interactions, 224–225recording sequences, 223–224

Bis(sulfosuccinimidyl) suberate (BS3), aviditymodulation:

integrin lateral redistribution, 174–175LFA-1 receptor cross-linking, 176–178

Boltzmann constant:atomic force microscopy, thermal cantilever

noise, 236–237leukocyte adhesion molecules, single-molecule

unbinding theory, 160–166photonic force microscopy, 152–157

Brownian motion, atomic force microscopy,thermal cantilever noise, 236–237

Buffered fluid environment, atomic forcemicroscopy, antiphospholipid syndrome,269–272

Calcium ions:leukocyte adhesion:

avidity modulation, 170–172ionomycin/thapsigargin stimulation,

175–176signal transduction mechanisms, 55–56SNARE-induced membrane fusion:

atomic levels for, 27–35cell-based mechanisms, 27

Cancer pathophysiology, nanodrug deliverysystems, 115–116

liposome structure, 117–118Cantilever structures:

atomic force microscopy:antiphospholipid syndrome, 268–272biological applications, 242–243conventional sizes, 232fast, low-noise detector, 239–241fast feedback loop, 242fast scanner, 241–242focused spot size, 237–239inverted optical microscope schematic,

137–141resonant frequency, 232–233small cantilever properties, 233–235thermal noise, 235–237

leukocyte adhesion molecules, aviditymodulation, 172–173

microbial cell surfaces, atomic forcemicroscopy analysis, imaging modes,71–72

photonic force microscopy, 151–157scanning probe microscopy, soft surface

imaging, 203, 210–211supported lipid bilayer thickness

measurements, 184–186Capacitance measurements, SNARE-induced

membrane fusion, atomic-level calciumparticipation in, 30–34

C-banding technique, metaphase chromosomes,257

Cell-based fusion machinery, SNARE andcalcium for, 27

Cell clustering, leukocyte adhesion, 177–178“Cell probe” technique, microbial cell surface

imaging, atomic force microscopy analysis,85

Cell secretion:growth hormone secretory vesicles, 61–63molecular mechanisms for, 17, 19–20porosome functions, 1–3

AFM imaging of, 6–17secretory vesicle content expulsion, molecular

mechanisms:basic principles, 37–38swelling process in cellular secretion,

38–44swelling process in neuron secretion,

44–46Cell spreading, leukocyte adhesion, 177–178Cellulolytic microorganisms, cellulose digestion,

atomic force microscopic imaging,99–100

Cellulose:atomic force microscopic imaging, 99–100plant cell walls, basic properties, 96

Page 293: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

290 INDEX

Charge-coupled device (CCD) camera:atomic force microscopy cantilevers, focused

spot dimensions, 238–239time-resolved imaging, 227–228

Chloride channels, porosome structural analysis,11–17

Cholesterol incorporation, sonicated unilamellarvesicle bilayer micromechanics, 195–197

Conductance measurements, SNARE-inducedmembrane fusion, atomic-level calciumparticipation in, 30–34

Constant-deflection imaging, microbial cellsurfaces, atomic force microscopy analysis,71–72

Constant force mode, scanning probemicroscopy, soft surface imaging, 203

Constant-height imaging:antiphospholipid syndrome, 281–282microbial cell surfaces, atomic force

microscopy analysis, 71–72scanning probe microscopy, soft surface

imaging, 203–205sonicated unilamellar vesicle bilayers,

thickness and morphology, 187–191Contact mode imaging:

cytogenetics research, 251–253microbial cell surfaces, atomic force

microscopy analysis, 71–72scanning probe microscopy:

liquid droplets, 212–214soft surface imaging, 203–205swollen polymer surfaces, 214–218

sonicated unilamellar vesicle bilayers,thickness and morphology, 188–191

Corticotropin-releasing hormone (CRH),growth-hormone release and synthesis, 55

Covalent immobilization, microbial cell surfaceimaging, atomic force microscopy analysis,71

Critical micelle concentration (CMC), supportedlipid bilayer thickness measurements,184–186

Cross-correlation alignment:real-time correction algorithm, 229time-resolved imaging, 225–227

Cross-linking mechanisms, leukocyte adhesion:avidity modulation, 171–172bis(sulfosuccinimidyl) suberate (BS3),

176–178Cyclic AMP (cAMP), signal transduction, 55–56Cyclooxygenase-2 (COX-2), nanodrug delivery

systems, dendrimer-ibuprofen nanodevices,127

Cytochalasin B, cell secretion and, 8–17Cytogenetics, atomic force microscopy:

C-banding technique, 257genetic material imaging techniques, 250–253metaphase chromosome karyotyping, 253–257

metaphase chromosome three-dimensionalanalysis, 257–259

nanoextraction, 259–262nanomanipulation and dissection, 259theoretical background, 249–250

Cytotoxic T-lymphocyte response (CTL),nanodrug delivery systems, 129–130

Daunoxome, nanodrug delivery with, 131–132De-adhesion, leukocyte adhesion,

ionomycin/thapsigargin stimulation,175–176

Degradation mechanisms, polymer-basednanodrug delivery, 120–121

Dendrimers, nanodrug delivery systems,122–130

chemistry, 124drug conjugation, 126–129free dendrimer interactions, 124–126ibuprofen nanodevices, 126–127methyl prednisolone nanodevices, 127–129

“Depression technique,” microbial cell surfaceelasticity, atomic force microscopy imaging,85–89

Differential interference contrast (DIC) lightmicroscopy, inner ear tissue studies,144–150

Diffusion mechanisms:membrane viscosity, thermal fluctuation

measurements, 155–157polymer-based nanodrug delivery, 120–121

Dioleoyl phosphatidyl ethanolamine (DOPE),nanodrug delivery systems, 118–120

Discrete Fourier transform (DFT), time-resolvedimaging, alignment techniques, 226–227

DNA:atomic force microscopic imaging, 243

cytogenetics research, 250–253metaphase chromosome karyotyping,

255–257enzymatic degradation, atomic force

microscopic imaging, 222–223p53 protein-DNA interaction, time-resolved

imaging, 224–226Drift phenomena:

atomic force microscopy, real-time correctionalgorithm, 229

biomolecular motion, time-resolved imaging,223–228

time-resolved imaging:alignment techniques, 226–227thermal drift, 227–228

Dynamic force spectroscopy, leukocyte adhesionmolecules:

integrin activation, 162–166selectin/sLeX complexes, 162single-molecule unbinding theory, 161–166

Page 294: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

INDEX 291

Dynamic mode imaging. See Tapping modeatomic force microscopy (TMAFM)

Egg yolk phosphatidylcholine (EggPC):sonicated unilamellar vesicle bilayers:

micromechanical measurements, 192–197thickness and morphology, 187–191

supported lipid bilayer thickness, 182–186Elasticity:

atomic force microscopy imaging, inner eartissue studies, 147–150

leukocyte adhesion, avidity modulation, 173microbial cell surfaces, atomic force

microscopy imaging, 85–89layers stretching, 87single cell stretching, 87–89single macromolecules, 86–87

photon force microscopy, single molecules,154–157

sonicated unilamellar vesicle bilayermicromechanics, 192–197

Electron beam deposition (EBD), atomic forcemicroscopy cantilevers, 233

nanoextraction, 262Electron microscopy:

metaphase chromosome karyotyping, 255–257microbial cell surface imaging, nanostructures,

77–79plant cell wall imaging, 97

Electrophysiology:atomic force microscopy and, 137–150

inner ear tissue studies, 144–150inverted optical microscope schematic,

137–141patch-clamp integration into, 141–144

microbial cell surface imaging, atomic forcemicroscopy analysis, 83–85

porosome structure and function, 17scanning probe microscopy, soft surface

imaging, 211secretory vesicle content expulsion:

basic principles, 37–38vesicle swelling in pancreatic acinar cells,

41–44SNARE-induced membrane fusion,

atomic-level calcium participation in,30–34

Electrostatic interactions, microbial cell surfaceimaging, atomic force microscopy analysis,83–85

Encapsulation efficiency, polymer-basednanodrug delivery, 120–121

Endocrine pancreas, porosome structure andfunction and, 8–17

Endocytosis, nanodrug delivery systems,114–116

Enhanced permeability and retention (EPR),nanodrug delivery systems, liposomestructure, 117–118

Enzyme activity, atomic force microscopicimaging, 222–223

ESEM imaging, lignin formation, 103–108Exocrine pancreas, porosome structure and

function and, 8–17Exocytosis, atomic force microscopic imaging,

inverted optical microscopic assembly,140–141

External agents, microbial cell surface effects,atomic force microscopy analysis, 78–79

Extracellular polymeric substances (EPS),microbial cell surface imaging, atomic forcemicroscopy, 76

Extravasation, leukocyte adhesion molecules,159

Feedback limits, atomic force microscopy, 231fast feedback loop, 242

Fibrinogen, atomic force microscopic imaging:anticoagulant effects, 275antiphospholipid syndrome, 274–275

Field emission in lens scanning electronmicroscopy (FEISEM), metaphasechromosomes, 258–259

Fluorescein isothiocyanate (FITC), nanodrugdelivery systems:

cellular interactions, 125–126dendrimer chemistry, 124methyl prednisolone-dendrimer nanodevices,

128–129Fluorescence in situ hydridization (FISH):

cytogenetics research, 250nanoextraction, 261–262

Fluorescence lifetime imaging (FLIM), plant cellwalls, 108–109

Fluorimetric fusion assays, SNARE-inducedmembrane fusion, atomic-level calciumparticipation in, 30–34

Focused spot dimensions, atomic forcemicroscopy cantilevers, 237–239

Folate receptor ligand, nanodrug deliverysystems, 119–120

Force-distance curves:atomic force microscopy, low-noise detection,

240–241microbial cell surface imaging:

atomic force microscopy analysis, 80–81elasticity measurements, 88layered surface applications, 87

future applications, 89–90soft tissue imaging, swollen polymer surfaces,

214–218

Page 295: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

292 INDEX

Force-distance curves (continued )sonicated unilamellar vesicle bilayers,

192–197supported lipid bilayer thickness, 183–186

Force measurements:atomic force microscopic imaging:

inner ear tissue studies, 148–150lateral dimension reduction, 230–231lipid bilayers and vesicles, 181–182

leukocyte adhesion molecules:avidity modulation, 172–173integrin lateral redistribution, 174–175single-molecule unbinding theory, 161–166

microbial cell surface imaging, atomic forcemicroscopy analysis, 80–81

sonicated unilamellar vesicle bilayers,191–197

Fourier Transform infrared (FTIR) imaging,lignin structures, 108

Freely jointed chain (FJC), microbial cell surfaceelasticity, 87–88

Friction force microscopy (FFM), soft surfaceimaging, 205

Fusion pores. See Porosomes“Fusogenic liposomes,” nanodrug delivery

systems, 118–120cycotoxic T-lymphocyte response, 129–130

Ghrelin:growth-hormone release and synthesis,

53–54signal transduction mechanisms, 55–56

Glass substrates, microbial cell surface imaging,atomic force microscopy analysis, 70–71

Glucosides, lignin synthesis, 100–108Gonadotropin-releasing hormone (GnRH),

growth-hormone release and synthesis, 54β2GPI cofactor, antiphospholipid syndrome,

atomic force microscopy, 275–277GroEL/GroES chaperonin proteins, atomic force

microscopy, 230–231Growth-hormone (GH), cell fusion pores,

pituitary gland:atomic force microscopy analysis, 59–60hypothalamic hormone control, 50–55

ghrelin, 53–54growth-hormone-releasing factor, 50–52peptide regulation, 54–55somatostatin, 52–53

immunocytochemical somatotrophdistribution, 57–58

overview, 49post-secretion secretory vesicle amounts,

60–63signal transduction mechanisms, 55–56in vivo neuroendocrine regulation, 49–50

Growth-hormone-releasing hormone (GHRH):

growth hormone release and synthesis, 50–52growth hormone secretory vesicles, secretion

effects, 62–63signal transduction mechanisms, 55–56

GTG banding, atomic force microscopicimaging:

genetics research, 250–253metaphase chromosome karyotyping,

253–257nanoextraction, 261–263

Guanosine triphosphate (GTP), secretory vesiclecontent expulsion:

basic principles, 37–38vesicle swelling in neurons, 44–46vesicle swelling in pancreatic acinar cells,

41–44

Harmonic oscillation, atomic force microscopy,thermal cantilever noise, 235–237

Hemicelluloses, plant cell walls, basic properties,96

Hemostatic proteins, atomic force microscopicimaging, antiphospholipid syndrome,274–275

Herzian model, sonicated unilamellar vesiclebilayers, force mechanics, 191–197

Hexagonally packed intermediate layers,microbial cell surface imaging, atomic forcemicroscopy analysis, 73

immobilization strategies, 71layered surface applications, 87

High-definition television, atomic forcemicroscopy, 231

Highly-oriented pyrolytic graphite, scanningtunneling microscopy, 242

High-pass frequency filter, plant cell walls,atomic force microscopic imaging, 96–97

Hooke’s law:force-distance curves, microbial cell surface

imaging, 80–81scanning probe microscopy, soft surface

imaging, 211Hydration mechanisms, SNARE-induced

membrane fusion, atomic-level calciumparticipation in, 30–34

Hydrogels, scanning probe microscopy, swollenpolymer surfaces, 216–218

Hydrophilic interactions, plant cell wall imaging,lignin structures, 106–108

Hydrophobic interactions, plant cell wallimaging, lignin structures, 106–108

Hydrophobic substrates, microbial cell surfaceimaging, atomic force microscopy analysis,70–71

Hypothalamic deafferentiation, in vivogrowth-hormone secretion, 49–51

Page 296: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

INDEX 293

Hypothalamic hormone control, growth-hormonerelease and synthesis, 50–55

ghrelin, 53–54growth-hormone-releasing factor, 50–52peptide regulation, 54–55somatostatin, 52–53

Ibuprofen, nanodrug delivery systems:chemistry, 124dendrimer-ibuprofen nanodevices, 126–127

Image alignment techniques, time-resolvedimaging, 225–227

Image tracking techniques, atomic forcemicroscopy, biomolecular motion, 228–229

Imaging speed, atomic force microscopy:biomolecular motion, 228–231

feedback limit, 231lateral dimension reduction, 229–230tracking mechanisms, 228–229video-rate imaging, 231

cantilever resonant frequency, 232–233Immobilization strategies:

leukocyte adhesion, avidity modulation, 172microbial cell surface imaging, atomic force

microscopy analysis, 71Immuno-atomic force microscopy

(ImmunoAFM):fixed cells, 5live cells, 5secretory product imaging and, 10–17

Immunocytochemistry, growth hormone cells, 57Immunogold localization, live pancreatic acinar

cells, 5Immunoprecipitation:

pancreatic plasma membrane, 5–6porosome structural analysis, 11–17

Inner ear tissue, atomic force microscopy studieson, 144–150

Integrins, leukocyte adhesion molecules:activation analysis, 162–166basic properties, 159–160lateral redistribution, 173–175

Intercellular adhesion molecule-1 (ICAM-1):avidity modulation:

basic principles, 169–171cells and reagents, 171force measurements, 172–173integrin lateral redistribution, 173–1753A9 cell line adhesion, 176–178

ionomycin/thapsigargin stimulation, 175–176Intermolecular forces:

leukocyte adhesion molecules:basic principles, 159–160integrin activation, 162–166selectin/sLeX complexes, 162single-molecule unbinding, 160–166

plant cell wall imaging, lignin structures,107–108

Inverted optical microscope:atomic force microscopy on, 137–141photonic force microscopy and, 152–157

In vivo studies:growth-hormone secretion, neuroendocrine

regulation, 49–50lignin synthesis, scanning probe microscopic

imaging, 100–110photon force microscopy, 154–157

Ion channel formation:atomic force microscopy, patch-clamp

integration, 141–144secretory vesicle content expulsion, basic

principles, 37–38SNARE-induced membrane fusion, 25–27

Ionomycin, leukocyte adhesion and, 175–176

Jump-in point, supported lipid bilayer thickness,183–186

Karyotyping, atomic force microscopic imaging,metaphase chromosomes, 253–257

Kinesin structures, photonic force microscopy,single molecules, 153–157

Laemmli sample preparation, pancreatic plasmamembrane, 5–6

Langmuir-Blodgett films, plant cell wallimaging, lignin structures, 108

Laser instrumentation, photonic forcemicroscopy, 152–155

Lateral dimensions, atomic force microscopy,229–230

Lateral drift, biomolecular motion, time-resolvedimaging, 223–228

Lateral force microscopy (LFM), soft surfaceimaging, 205–206

LCAO-MO calculations, lignin formation,101–108

Leptin, growth-hormone release and synthesis,54

Leukocyte adhesion molecules:avidity modulation studies:

adhesive force measurements, 172–173basic principles, 169–171cells and reagents, 171clinical applications, 176–178elasticity measurements, 173integrin lateral redistribution, 173–175ionomycin/thapsigargin stimulation,

175–176

Page 297: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

294 INDEX

Leukocyte adhesion molecules (continued )protein immobilization, 1723A9 cell crosslinking, 171–172

intermolecular forces:basic principles, 159–160integrin activation, 162–166selectin/sLeX complexes, 162single-molecule unbinding, 160–166

Leukocyte function-associated antigen-1(LFA-1):

avidity modulation:basic principles, 169–171ionomycin/thapsigargin stimulation,

175–176results and discussion, 176–178

leukocyte adhesion molecules:basic properties, 159–160integrin activation, 162–166lateral redistribution, 173–175

Life mode, scanning probe microscopy, softsurface imaging, 210

Ligand-decorated liposomes, nanodrug deliverysystems, 119–120

Light scattering analysis, SNARE-inducedmembrane fusion, atomic-level calciumparticipation in, 30–34

Lignin:plant cell walls, basic properties, 96scanning probe microscopic imaging, 100–108

Lipid bilayers:atomic force microscopic imaging,

antiphospholipid syndrome, 273–274secretory vesicle content expulsion, vesicle

swelling in pancreatic acinar cells, 41–44SNARE-induced membrane fusion, 25–27

atomic-level calcium participation in, 27–34thickness and micromechanical properties:

atomic force microscopic imaging, 181–197supported thickness, 182–183

Lipid membrane:porosome reconstitution on mica, 4porosome structure and function and, 12–17“raft” structures, viscosity measurements,

156–157secretory vesicle content expulsion, vesicle

swelling in pancreatic acinar cells, 42–44SNARE-induced membrane fusion, bilayer

interaction and conducting channelformation, 26–27

Liposomes, nanodrug delivery systems:conventional structures, 116–118modified liposomes, 118–120

Liquid droplets, scanning probe microscopy, softsurface imaging, 212–214

Lithium chloride, microbial cell surface effects,atomic force microscopy analysis, 79

Loading force measurements, sonicatedunilamellar vesicle bilayers,micromechanical measurements, 192–197

Low-noise detection, atomic force microscopycantilevers, 239–241

L-plastin, leukocyte adhesion, aviditymodulation, 170–171

Lupus anticoagulant (LA) phenomenon,antiphospholipid syndrome, 275

annexin A5, 278–279

MacMARCKS substrate, leukocyte adhesion,avidity modulation, 170–171

Macromolecules:atomic force microscopic imaging, hemostasis

and thrombosis, 267–283microbial cell surface elasticity, atomic force

microscopy imaging, 86–87pectin “brush structure,” atomic force

microscopic imaging, 97–98Macropinocytosis, nanodrug delivery systems,

114–116Madin-Darby canine kidney (MDCK) cells,

nanodrug delivery systems, dendrimerstructures, 124

Major histocompatibility complex (MHC)molecules, nanodrug delivery systems,129–130

Mechanical drift, biomolecular motion,time-resolved imaging, 223–228

Membrane fusion:atomic force microscopic imaging, inverted

optical microscopic assembly, 140–141porosome functions, 1–3SNARE-induced, molecular mechanisms,

25–34bilayer interaction circular array,

conducting channel formation, 25–27calcium ion participation, 27–34cell fusion machinery with, 27

Membrane viscosity, photonic force microscopy,thermal fluctuation measurements, 155–157

Metaphase chromosomes, atomic forcemicroscopic imaging:

correlative three-dimensional morphologicalanalysis, 257–259

genetic material, 250–253GTG-banding patterns, 251–253historical background, 249–250karyotyping of, 253–257

Methyl prednisolone nanodevices, nanodrugdelivery systems, dendrimer conjugates,127–129

Mica substrates:atomic force microscopic imaging,

antiphospholipid syndrome, 273–274

Page 298: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

INDEX 295

microbial cell surface imaging, atomic forcemicroscopy analysis, 70–71

Micelles, polymer-based nanodrug delivery,121–122

Microbial cell surfaces, atomic force microscopyanalysis, 69–90

bacillus S-layers, 73basic principles, 69–70cell surface layer imaging, 73–75elasticity, 85–89

layers stretching, 87single cell stretching, 87–89single macromolecules, 86–87

external agent effects, 78–79future applications, 89–90hexagonally packed intermediate layer, 73imaging techniques, 70–79

immobilization strategies, 71selection criteria, 71–72substrate requirements, 70–71

microbial biofilms, 75–76nanostructures, 76–78physical properties measurement, 79–89

force-distance curves, 80force measurements, 80–81functionalized probes, 80–81

physicochemical properties and molecularinteractions, 81–85

adhesion mapping, 81–82cell probe technique, 85surface charges and electrostatic

interactions, 83–85surface energy and solvation interactions,

82–83physiological changes, 78porin crystals, 74–75purple membranes, 73–74

Microdissection, atomic force microscopy,259–260

Microfibrils, plant cell walls, basic properties,95–96

Micromechanical measurements:atomic force microscopy imaging, inner ear

tissue studies, 146–150sonicated unilamellar vesicle bilayers,

191–197Molecular mechanisms:

cell secretion, porosome structure and functionand, 17, 19–20

microbial cell surface imaging, atomic forcemicroscopy analysis, 81–85

photonic force microscopy, thermal fluctuationmeasurements, 153–157

plant cell walls, atomic force microscopicimaging, 96–97

secretory vesicle content expulsion:basic principles, 37–38cellular secretion swelling, 38–44

neuron secretion swelling, 44–46SNARE-induced membrane fusion, 25–34

bilayer interaction circular array,conducting channel formation, 25–27

calcium ion participation, 27–34cell fusion machinery with, 27

Molecular weight calculations, lignin formation,103–108

Monoclonal antibodies, antiphospholipidsyndrome, annexin A5, 278–279

Motilin, growth-hormone release and synthesis,ghrelin mediation, 54

Multilamellar vesicle (MLV) solution, sonicatedunilamellar vesicle bilayers, thickness andmorphology, 188–191

Nanobioscience:antiphospholipid syndrome, 268–272atomic force microscopy, cytogenetic

applications, 259–262historical evolution of, 2–3

Nanodrug delivery systems:cellular interactions:

basic principles, 113–116dendrimers, 122–130

chemistry, 124drug conjugation, 126–129free dendrimer interactions, 124–126ibuprofen nanodevices, 126–127methyl prednisolone nanodevices,

127–129lipid-based systems, 116–120polymer-based systems, 120–130

micelles, 121–122nanoparticles, 120–121

clinical applications and future research,130–132

Nanoextraction, atomic force microscopy,259–262

Nanoparticles:AFM manipulation and dissection, 259polymer-based nanodrug delivery, 120–121scanning probe microscopy, liquid droplets,

212–214Nanostructures:

microbial cell surface imaging, atomic forcemicroscopy analysis, 76–79

physiological changes, 78photonic force microscopy imaging, 151–157

Near-field scanning optical microscopy(NSOM):

antiphospholipid syndrome, 268–272lignin imaging, 104–108plant cell wall imaging, 108–109

Neuroendocrine regulation, in vivogrowth-hormone secretion, 49–50

Page 299: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

296 INDEX

Neurons:porosome structure and function in, 8–17secretory vesicle content expulsion, 44–46

Neuropeptide Y (NPY), growth-hormone releaseand synthesis, ghrelin mediation, 54

Noncontact mode:cytogenetics research, 252–253scanning probe microscopy, soft surface

imaging, 210NSF-ATP assays, SNARE-induced membrane

fusion, atomic-level calcium participationin, 31–34

Nyquist relation, atomic force microscopy,thermal cantilever noise, 236–237

OmpF porin, microbial cell surface imaging,atomic force microscopy analysis,immobilization strategies, 71

Opposing bilayers, SNARE-induced membranefusion, 25–27

Optical beam deflection, atomic forcemicroscopy cantilevers, low-noise detection,239–241

Optical trapping techniques, photonic forcemicroscopy, 151–157

Pancreatic acinar cells:immunogold localization, 5isolation, 3–4porosome structure in, 6–17secretory vesicle content expulsion:

basic principles, 37–38vesicle swelling during secretion, 38–44

transmission electron microscopy imaging, 5Pancreatic plasma membrane:

immunoprecipitation and Western blotanalysis, 5–6

isolation, 4porosome structural analysis, 10–17secretory vesicle content expulsion, basic

principles, 37–38Patch-clamp integration, atomic force

microscopy, 141–144inner ear tissue studies, 144–150

Pectins:atomic force microscopic imaging, 97–98basic properties, 96

PEO-b-poly(L-amino acids) (PLAA) blockcopolymers, polymer-based nanodrugdelivery, 122

multidrug resistance and, 131Phagocytosis, nanodrug delivery systems,

114–116liposome structure, 117–118

Phase contrast microscopy, metaphasechromosome karyotyping, 254–257

Phase shift detection, scanning probemicroscopy, soft surface imaging, 209–210

Phenolic alcohols, lignin synthesis, 100–108Phorbol myristate acetate (PMA), leukocyte

adhesion:avidity modulation, 170–171cell spreading, 177–178integrin lateral redistribution, 174–175

Phosphatidylserine (PS), antiphospholipidsyndrome, 276–277

Phospholipase C-IP3-protein kinase C, signaltransduction, 55–56

Photodetection sensitivity, atomic forcemicroscopy cantilevers, 240–241

Photonic force microscopy (PFM):development of, 151–157position-sensing systems, 151–153thermal fluctuation measurements:

membrane viscosity, 155–157single molecules, 153–155

Physicochemical properties, microbial cellsurface imaging, atomic force microscopy,81–85

Physiological changes, microbial cell surfaceimaging, atomic force microscopy analysis,78

Phytohemagglutinin (PHA), cytogeneticsapplications, atomic force microscopy, 249

Piezoelectric crystals:atomic force microscopy:

antiphospholipid syndrome, 269–272fast scanners, 241–242

scanning probe microscopy, soft surfaceimaging, 202–203

Pipette structures, atomic force microscopy,138–141

Pituitary adenylate cyclase-activating peptide(PACAP):

growth-hormone release and synthesis, 54signal transduction mechanisms, 55–56

Pituitary gland, growth-hormone cell fusionpores:

atomic force microscopy analysis, 59–60hypothalamic hormone control, 50–55

ghrelin, 53–54growth-hormone-releasing factor, 50–52peptide regulation, 54–55somatostatin, 52–53

immunocytochemical somatotrophdistribution, 57–58

overview, 49post-secretion secretory vesicle amounts,

60–63signal transduction mechanisms, 55–56in vivo neuroendocrine regulation, 49–50

Pixel acquisition, time-resolved imaging,227–228

Page 300: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

INDEX 297

Planck’s constant, leukocyte adhesion molecules,single-molecule unbinding theory, 160–166

Plant cell wall:atomic force microscopy (AFM), 96–97

cellulose, 99–100pectins, 97–99

scanning probe microscopic imaging:basic properties, 95–96future research techniques, 108–109lignin, 100–108

Plasma membrane:growth hormone secretory vesicles, secretion

effects, 61–63porosome structure and function and, 8–17

Poisson’s ratio, sonicated unilamellar vesiclebilayer micromechanics

Poly(acrylic acid) (PAA), soft tissue imaging,swollen surfaces, 214–218

Polyamidoamine (PAMAM) dendrimers,nanodrug delivery systems:

basic properties, 123–124cellular interactions, 125–126dendrimer-drug conjugates, 126–129

Poly(DL-lactide-co-glycolide) (PLGA),polymer-based nanodrug delivery, 120–121

Poly(ethylene oxide) (PEO):polymer-based nanodrug delivery, 122supported lipid bilayer micromechanics,

195–197supported lipid bilayer thickness

measurements, 184–186Polylactides, polymer-based nanodrug delivery,

120–121Polymer-based systems:

nanodrug delivery, cellular interactions,120–130

micelles, 121–122nanoparticles, 120–121

soft tissue imaging, swollen surfaces, 214–218Polysaccharides:

pectin macromolecules, atomic forcemicroscopic imaging, 97–98

plant cell walls, basic properties, 95–96Porin crystals, microbial cell surface imaging,

atomic force microscopy, 74–75Porosomes:

atomic force microscopy, 4–5basic properties, 1–3growth-hormone secretion in pituitary gland:

atomic force microscopy analysis, 59–60hypothalamic hormone control, 50–55

ghrelin, 53–54growth-hormone-releasing factor, 50–52peptide regulation, 54–55somatostatin, 52–53

immunocytochemical somatotrophdistribution, 57–58

overview, 49

post-secretion secretory vesicle amounts,60–63

signal transduction mechanisms, 55–56in vivo neuroendocrine regulation, 49–50

immuno-atomic force microscopy:fixed cells, 5live cells, 5

immunoprecipitation and Western blotanalysis, 5–6

lipid membrane reconstitution, mica-based, 4molecular-based cell secretion, 17–22pancreatic acinar cell isolation, 3–4pancreatic plasma membrane preparation, 4secretory vesicle content expulsion, vesicle

swelling in pancreatic acinar cells, 41–44structural properties, 6–17synaptosome/synaptosomal/synaptic vesicle

isolation, 4transmission electron microscopy, 5zymogen granule isolation, 5

PPO complexes, supported lipid bilayermicromechanics, 195–197

p53 protein-DNA interaction, time-resolvedimaging, 224–226

Probe surface chemistry:atomic force microscopy, antiphospholipid

syndrome, 269–272microbial cell surface imaging, atomic force

microscopy, 81–82Proportional-integral-differential (PID) feedback

loop, atomic force microscopy, 242Prostaglandin (PGE2), nanodrug delivery

systems:dendrimer-ibuprofen nanodevices, 127methyl prednisolone-dendrimer nanodevices,

128–129Protein immobilization, leukocyte adhesion,

avidity modulation, 172Protein kinase A, signal transduction, 55–56Protein kinase C:

leukocyte adhesion, avidity modulation,170–171

signal transduction, 55–56Protein-protein interactions, atomic force

microscopic imaging, 222–223Proton nuclear magnetic resonance, plant cell

wall imaging, lignin structures, 106–108Pulronic copolymers, supported lipid bilayer

thickness measurements, 184–186Purple membranes, microbial cell surface

imaging, atomic force microscopy analysis,73–74

immobilization strategies, 71

Q-banding, cytogenetics research,249–250

Page 301: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

298 INDEX

Q control:atomic force microscopy:

cantilever resonant frequency, 232–233thermal cantilever noise, 235–237

scanning probe microscopy:liquid droplets, 214soft surface imaging, 207–209

Quantitative analysis, growth hormone cells,57–58

Rack1 phosphorylation, leukocyte adhesion,avidity modulation, 170–171

“Raft” lipids, viscosity measurements, 156–157Raman spectroscopy, plant cell wall imaging,

lignin structures, 106–108Rayleigh range, atomic force microscopy

cantilevers, focused spot dimensions,238–239

Receptor affinity modulation, leukocyteadhesion, 169–171

Receptor-mediated endocytosis, nanodrugdelivery systems, 114–116

Recrystallization techniques, microbial cellsurface imaging, atomic force microscopyanalysis, 71

Repulsive force, sonicated unilamellar vesiclebilayer micromechanics, 195–197

Resonant frequencies, atomic force microscopycantilevers, 232–233

Reverse hemolytic plaque assay, growthhormone fusion pore structure anddynamics, 59–60

RMS-DC converter, atomic force microscopy:antiphospholipid syndrome, 270–272fast feedback loop, 242

RPMI 1640 medium, leukocyte adhesion, aviditymodulation, 171

Scanner design, atomic force microscopy,241–242

Scanning electron microscopy (SEM):metaphase chromosomes, 257–258simultaneous AFM/patch-clamp recordings,

145–147Scanning probe microscopy (SPM):

plant cell wall:basic properties, 95–96future research techniques, 108–109lignin, 100–108

soft surface imaging:basic principles, 201–202cantilevers and probes, 210–211contact mode, 203–205dynamic mode, 205–209experimental setup, 202–203liquid droplets, 212–214

phase shift technique, 209–210physics and methods, 202–211swollen polymer surfaces, 214–218

Scanning tunneling microscopy (STM):antiphospholipid syndrome, 268–272cytogenetics research, 250lignin structure, 101–108scanner speed, 242

Secretagogue exposure, cell secretion and, 6–17Secretory vesicles:

content expulsion, molecular mechanisms:basic principles, 37–38swelling process in cellular secretion,

38–44swelling process in neuron secretion, 44–46

growth hormone cells, secretion effects,60–63

Selectin/sLeX complexes, atomic forcemicroscopic imaging, 162

Signal-to-noise ratio, atomic force microscopy:low-noise detection, 240–241thermal cantilever noise, 236–237

Signal transduction:adenylate cyclase/cAMP/protein kinase A and

phospholipase C-IP3-protein kinase C,55–56

growth-hormone release and synthesis:growth-hormone-releasing hormone, 51–52somatostatin, 52–53

Silicon nitride cantilevers:atomic force microscopy:

antiphospholipid syndrome, 268–272resonant frequency, 233

scanning probe microscopy, soft surfaceimaging, 210–211

Silicon oxide substrates, microbial cell surfaceimaging, atomic force microscopy analysis,70–71

Single molecule force spectroscopy, microbialcell surface elasticity, 86–88

Single-molecule unbinding theory, leukocyteadhesion molecules, 160–166

SNARE complex:membrane fusion, molecular mechanisms,

25–34bilayer interaction circular array,

conducting channel formation, 25–27calcium ion participation, 27–34cell fusion machinery with, 27

porosome structure and function and, 13–17Soft surface imaging, scanning probe

microscopy:basic principles, 201–202cantilevers and probes, 210–211contact mode, 203–205dynamic mode, 205–209experimental setup, 202–203liquid droplets, 212–214

Page 302: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

INDEX 299

phase shift technique, 209–210physics and methods, 202–211swollen polymer surfaces, 214–218

Solvation interactions, microbial cell surfaceimaging, atomic force microscopy analysis,82–83

Somatostatin (SRIF), growth-hormone releaseand synthesis, 52–53

Somatotrophs:growth-hormone release and synthesis, 55immunocytochemical distribution, 57–58

Sonicated unilamellar vesicle (SUV) bilayers:micromechanical properties, 191–197thickness and morphology, 187–191

Stalk sectioning techniques, in vivogrowth-hormone secretion, 49–50

“Stealth” liposomes, nanodrug delivery systems,118–120

Stereocilia properties, atomic force microscopyimaging, inner ear tissue studies, 144–150

“Sterically stabilized” liposomes, nanodrugdelivery systems, 118–120

Stiffness measurement, atomic force microscopyimaging, inner ear tissue studies, 148–150

Stokes drag, membrane viscosity, thermalfluctuation measurements, 156–157

Substrate requirements:leukocyte adhesion, avidity modulation,

170–171microbial cell surface imaging, atomic force

microscopy analysis, 70–71Supercritical fluids, polymer-based nanodrug

delivery, 120–121Supermodule structural unit, lignin formation,

101–108Supported lipid bilayer (SLB), thickness

measurements, 182–186Supramolecular activation clusters, leukocyte

adhesion, avidity modulation, 170–171Surface charges, microbial cell surface imaging,

atomic force microscopy analysis, 83–85Surface energy, microbial cell surface imaging,

atomic force microscopy analysis, 82–83Surface-enhanced Raman spectroscopy, plant

cell wall imaging, 108–109Synaptic vesicles:

isolation, 4porosome structure and function in, 12–17

Synaptosomal membrane:isolation, 4porosome structural analysis, 11–17

Synaptosome:isolation, 4porosome structure and function in, 12–17

Tapping mode atomic force microscopy(TMAFM):

antiphospholipid syndrome, 271–272biomolecular motion, basic principles,

221–223feedback limits, 231genetic material imaging, 251–253microbial cell surface imaging:

biofilms, 76external agent effects, 79purple membranes, 74

pancreatic acinar cells, 5porosome imaging, 5soft surface imaging, 205–209

swollen polymer surfaces, 216–218sonicated unilamellar vesicle bilayers,

thickness and morphology, 188–191Thapsigargin, leukocyte adhesion stimulation,

175–176Thermal cantilever noise, atomic force

microscopy, 235–237low-noise detection, 240–241

Thermal fluctuation measurements, photonicforce microscopy, single molecules,153–157

3A9 cell line, leukocyte adhesion:avidity modulation, 171–172

force measurements, 172–173integrin clustering, 176–178

Three-dimensional morphological analysis,atomic force microscopic imaging,metaphase chromosomes, 257–259

Three-dimensional topographic imaging,supported lipid bilayer thickness, 183–186

Thyrotropin-releasing hormone (TRH),growth-hormone release and synthesis, 54

Time-resolved imaging, biomolecular motion,223–228

alignment, 225–227moving images, 227–228p53-DNA dynamic interactions, 224–225recording sequences, 223–224

Tip effect, sonicated unilamellar vesicle bilayers,thickness and morphology, 190–191

Topographic imaging, scanning probemicroscopy, soft surface imaging, 205

Transmission electron microscopy (TEM):growth hormone fusion pore structure and

dynamics, 59–60growth hormone secretory vesicles, 61–63metaphase chromosomes, 257–258pancreatic acinar cell isolation, 5plant cell walls, 97porosome structure and function and, 10–17

Ultraviolet (UV) radiation, plant cell wallimaging, lignin structures, 106–108

Page 303: FORCE MICROSCOPY - The Eyethe-eye.eu/public/Books/Medical/texts/Force Microscopy - Applicatio… · Care Department within the United States at (800) 762-2974, outside the United

300 INDEX

Vertical drift, biomolecular motion,time-resolved imaging, 223–228

Very late antigen-4 (VLA-4), leukocyte adhesionmolecules:

basic properties, 159–160integrin activation, 165–166

Vesicles:secretory vesicle content expulsion, swelling

mechanisms:basic principles, 37–38cellular secretion, 38–44neuron secretion, 44–46

sonicated unilamellar vesicle:bilayer thickness and morphology,

187–191micromechanical properties, 191–197

synaptic vesicles:isolation, 4porosome structure and function in,

12–17Video-rate imaging, atomic force microscopy,

231“Virosome,” nanodrug delivery systems,

118–120Viscosity of membranes:

photonic force microscopy, thermal fluctuationmeasurements, 155–157

scanning probe microscopy, soft surfaceimaging, 212–214

Viscous damping, atomic force microscopy,thermal cantilever noise, 235–237

Water transport, plant cell wall imaging:basic properties, 96–97lignin mechanisms for, 105–108

Western blot analysis, pancreatic plasmamembrane, 5–6

Worm-like chain (WLC) model, microbial cellsurface elasticity, 87–88

X-ray diffraction, SNARE-induced membranefusion, atomic-level calcium participationin, 27–34

Young’s modulus:leukocyte adhesion molecules:

elasticity, 173integrin lateral redistribution, 174–175

sonicated unilamellar vesicle bilayermicromechanics, 191–197

Zymogen granules (ZG):isolation, 5porosome structure and function and, 10–17secretory vesicle content expulsion:

basic principles, 37–39neuron vesicle swelling, 44–46pancreatic acinar cell vesicle swelling,

38–44