a microdevice for multiplexed detection of t-cell-secreted cytokines

9
A microdevice for multiplexed detection of T-cell-secreted cytokines He Zhu, a Gulnaz Stybayeva, ac Monica Macal, b Erlan Ramanculov, c Michael D. George, b Satya Dandekar b and Alexander Revzin * a Received 17th June 2008, Accepted 19th August 2008 First published as an Advance Article on the web 30th September 2008 DOI: 10.1039/b810244a Cytokines are produced by immune cells in response to viral or bacterial pathogens and therefore have significant diagnostic value. The goal of the present study was to develop a miniature device for detection of interleukin (IL)-2 and interferon (IFN)-g cytokines secreted by a small population of CD4 and CD8 T-cells. Microarrays of T-cell- and cytokine-specific Ab spots were printed onto poly(ethylene glycol) (PEG) hydrogel-coated glass slides and enclosed inside a microfluidic device, creating a miniature (3 mL) immunoreaction chamber. Introduction of the red blood cell (RBC) depleted whole human blood into the microfluidic device followed by washing at a pre-defined shear stress resulted in isolation of pure CD4 and CD8 T-cells on their respective Ab spots. Importantly, the cells became localized next to anti-IL-2 and -IFN-g Ab spots. Mitogenic activation of the captured T-cells was followed by immunofluorescent staining (all steps carried out inside a microfluidic device), revealing concentration gradients of surface-bound cytokine molecules. A microarray scanner was then used to quantify the concentration of IFN-g and IL-2 near CD4 and CD8 T-cells. This study represents one of the first demonstrations of a microdevice for capturing desired T-cell subsets from a small blood volume and determining, on-chip, cytokine profiles of the isolated cells. Such a microdevice is envisioned as an immunology tool for multi-parametric analysis of T-cell function with direct applications in diagnosis/monitoring of HIV and other infectious diseases. Introduction Activated T-lymphocytes (T-cells) are central participants of immune response to viral or bacterial infections. T-helper cells (CD4 T-cells) and cytotoxic T-lymphocytes (CD8 T-cells) are major T-cell subsets that regulate immune cell recruitment and proliferation through a program of cytokine production. 1,2 Based on the types of secreted cytokines, CD4 T-helper cells are further subdivided into T-helper 1 (Th1), Th2, Th17 and other subsets, making cytokine profiling an important part of determining Th phenotype. Importantly, cytokines secreted by T-cells have been reported to correlate with positive or negative disease outcomes and thus provide important diagnostic information. In human immunodeficiency virus (HIV) infections, Th1 phenotype is associated with protection against the pathogen and is preserved in long-term nonprogressor patients, while Th2 phenotype is correlated with rapid progression of the infection and onset of AIDS. 3–5 Similarly, vigorous cytotoxic response of CD8 T-cells is needed to keep HIV viremia in check. 6 Therefore, detecting cytokines (e.g. IFN-g and IL-2) associated with Th1 and cyto- toxic responses of T-cells is important for HIV diagnosis and monitoring. 3,7 Cytokine production is traditionally determined using several bioanalytical approaches including flow cytometry coupled with intracellular cytokine staining, enzyme-linked immunospot (ELISPOT) method, enzyme linked immunosorbent assay (ELISA) and polymerase chain reaction (PCR). 4,8,9 ELISA and PCR are robust technologies for detecting either cytokines or cytokine mRNA in the blood stream but they can not be used to identify specific populations of cytokine producing cells. In contrast, flow cytometry-based approach allows one to connect cytokine production, determined by intracytoplasmic staining, to cell phenotype based on Ab-labeling of leukocyte surface antigens. 9,10 However, this method reports the frequency of cytokine positive cells and not the cytokine concentration; it requires a large number of cells for analysis and analyzes fixed (as opposed to live) cells. ELISPOT is another technology employed to detect cytokine production in leukocytes. 8,11 In this approach, leukocytes are seeded into micro-titer plates pre- coated with anti-cytokine Abs and are stimulated to commence cytokine production. The cells are then washed away and the numbers of spot forming units are quantified in a manner analogous to ELISA. While it provides information about the number of cytokine-secreting cells, the ELISPOT experiment must be preceded by a sorting step to isolate the desired leukocyte subset. Such an extra step is required due to ‘‘promiscuity’’ in leukocyte cytokine production where IFN-g, for example, may be secreted by CD4 and CD8 T-cells. In addition, the ELISPOT technique does not quantify the amount of secreted cytokines. Robotic printing has emerged as an important enabling tech- nology for proteomics research in general 12 and immunoassay development in particular. 13–18 Printed arrays of antibodies (Abs) have been employed to capture pathogens or proteins such as a Department of Biomedical Engineering, University of California, Davis, 451 East Health Sciences St. #2619, Davis, CA, 95616, USA. E-mail: [email protected]; Fax: +1 530-754-5739; Tel: +1 530-752-2383 b Medical Microbiology and Immunology, University of California, Davis, USA c National Center for Biotechnology, Astana, Republic of Kazakhstan This journal is ª The Royal Society of Chemistry 2008 Lab Chip, 2008, 8, 2197–2205 | 2197 PAPER www.rsc.org/loc | Lab on a Chip Downloaded by University of California - Davis on 22 November 2010 Published on 30 September 2008 on http://pubs.rsc.org | doi:10.1039/B810244A View Online

Upload: independent

Post on 23-Nov-2023

2 views

Category:

Documents


0 download

TRANSCRIPT

PAPER www.rsc.org/loc | Lab on a Chip

Dow

nloa

ded

by U

nive

rsity

of

Cal

ifor

nia

- D

avis

on

22 N

ovem

ber

2010

Publ

ishe

d on

30

Sept

embe

r 20

08 o

n ht

tp://

pubs

.rsc

.org

| do

i:10.

1039

/B81

0244

AView Online

A microdevice for multiplexed detection of T-cell-secreted cytokines

He Zhu,a Gulnaz Stybayeva,ac Monica Macal,b Erlan Ramanculov,c Michael D. George,b Satya Dandekarb

and Alexander Revzin*a

Received 17th June 2008, Accepted 19th August 2008

First published as an Advance Article on the web 30th September 2008

DOI: 10.1039/b810244a

Cytokines are produced by immune cells in response to viral or bacterial pathogens and therefore have

significant diagnostic value. The goal of the present study was to develop a miniature device for

detection of interleukin (IL)-2 and interferon (IFN)-g cytokines secreted by a small population of CD4

and CD8 T-cells. Microarrays of T-cell- and cytokine-specific Ab spots were printed onto poly(ethylene

glycol) (PEG) hydrogel-coated glass slides and enclosed inside a microfluidic device, creating

a miniature (�3 mL) immunoreaction chamber. Introduction of the red blood cell (RBC) depleted

whole human blood into the microfluidic device followed by washing at a pre-defined shear stress

resulted in isolation of pure CD4 and CD8 T-cells on their respective Ab spots. Importantly, the cells

became localized next to anti-IL-2 and -IFN-g Ab spots. Mitogenic activation of the captured T-cells

was followed by immunofluorescent staining (all steps carried out inside a microfluidic device),

revealing concentration gradients of surface-bound cytokine molecules. A microarray scanner was then

used to quantify the concentration of IFN-g and IL-2 near CD4 and CD8 T-cells. This study represents

one of the first demonstrations of a microdevice for capturing desired T-cell subsets from a small blood

volume and determining, on-chip, cytokine profiles of the isolated cells. Such a microdevice is

envisioned as an immunology tool for multi-parametric analysis of T-cell function with direct

applications in diagnosis/monitoring of HIV and other infectious diseases.

Introduction

Activated T-lymphocytes (T-cells) are central participants of

immune response to viral or bacterial infections. T-helper cells

(CD4 T-cells) and cytotoxic T-lymphocytes (CD8 T-cells) are

major T-cell subsets that regulate immune cell recruitment and

proliferation through a program of cytokine production.1,2Based

on the types of secreted cytokines, CD4 T-helper cells are further

subdivided into T-helper 1 (Th1), Th2, Th17 and other subsets,

making cytokine profiling an important part of determining Th

phenotype. Importantly, cytokines secreted by T-cells have been

reported to correlate with positive or negative disease outcomes

and thus provide important diagnostic information. In human

immunodeficiency virus (HIV) infections, Th1 phenotype is

associated with protection against the pathogen and is preserved

in long-term nonprogressor patients, while Th2 phenotype is

correlated with rapid progression of the infection and onset of

AIDS.3–5 Similarly, vigorous cytotoxic response of CD8 T-cells is

needed to keep HIV viremia in check.6 Therefore, detecting

cytokines (e.g. IFN-g and IL-2) associated with Th1 and cyto-

toxic responses of T-cells is important for HIV diagnosis and

monitoring.3,7

Cytokine production is traditionally determined using several

bioanalytical approaches including flow cytometry coupled

aDepartment of Biomedical Engineering, University of California, Davis,451 East Health Sciences St. #2619, Davis, CA, 95616, USA. E-mail:[email protected]; Fax: +1 530-754-5739; Tel: +1 530-752-2383bMedical Microbiology and Immunology, University of California, Davis,USAcNational Center for Biotechnology, Astana, Republic of Kazakhstan

This journal is ª The Royal Society of Chemistry 2008

with intracellular cytokine staining, enzyme-linked immunospot

(ELISPOT) method, enzyme linked immunosorbent assay

(ELISA) and polymerase chain reaction (PCR).4,8,9 ELISA and

PCR are robust technologies for detecting either cytokines or

cytokine mRNA in the blood stream but they can not be used

to identify specific populations of cytokine producing cells. In

contrast, flow cytometry-based approach allows one to connect

cytokine production, determined by intracytoplasmic staining,

to cell phenotype based on Ab-labeling of leukocyte surface

antigens.9,10 However, this method reports the frequency of

cytokine positive cells and not the cytokine concentration; it

requires a large number of cells for analysis and analyzes fixed

(as opposed to live) cells. ELISPOT is another technology

employed to detect cytokine production in leukocytes.8,11 In

this approach, leukocytes are seeded into micro-titer plates pre-

coated with anti-cytokine Abs and are stimulated to commence

cytokine production. The cells are then washed away and the

numbers of spot forming units are quantified in a manner

analogous to ELISA. While it provides information about the

number of cytokine-secreting cells, the ELISPOT experiment

must be preceded by a sorting step to isolate the desired

leukocyte subset. Such an extra step is required due to

‘‘promiscuity’’ in leukocyte cytokine production where IFN-g,

for example, may be secreted by CD4 and CD8 T-cells. In

addition, the ELISPOT technique does not quantify the

amount of secreted cytokines.

Robotic printing has emerged as an important enabling tech-

nology for proteomics research in general12 and immunoassay

development in particular.13–18 Printed arrays of antibodies (Abs)

have been employed to capture pathogens or proteins such as

Lab Chip, 2008, 8, 2197–2205 | 2197

Dow

nloa

ded

by U

nive

rsity

of

Cal

ifor

nia

- D

avis

on

22 N

ovem

ber

2010

Publ

ishe

d on

30

Sept

embe

r 20

08 o

n ht

tp://

pubs

.rsc

.org

| do

i:10.

1039

/B81

0244

AView Online

cytokines for subsequent detection with sandwich immuno-

assay.13,15,17,18

In addition to detection of biomolecules, protein microarrays

have been used for high-throughput screening of cell–surface

interactions.19–21 Belov et al. used robotic printing to create

arrays of leukocyte-specific Abs for isolation or ‘‘panning’’ of

multiple leukocyte subsets on the same surface and for subse-

quent correlation of cell binding patterns to blood malignan-

cies.19 More recently functionality of printed microarrays has

been expanded by incorporating sensing elements in addition to

cell-capture molecules. Chen et al. described a microarray with

individual spots containing T-cell specific antigens and cytokine-

specific Abs.22 This method allowed one to detect cytokine

secreting T-cells bound on the spot but was unsuitable for

quantifying levels of secreted cytokines. Another important

recent study described the use DNA-encoded Ab arrays inte-

grated with microfluidics for detection of cells, proteins and

nucleic acids in the same microdevice.23 While highlighting the

potential use of the same microarray for cell sorting and protein

detection, this study employed purified T-cells and stopped short

of detecting cell-secreted cytokines.

Recently, we described the use of Ab microarrays for

capturing pure (>95%) CD4 and CD8 T-cells from whole human

blood and quantifying T-cell proportions based on cell binding

to Ab spots.24 The goal of the present study was to integrate

multiplexed immunoassay for detection of secreted cytokines

with captured T-cells. To achieve this goal, microarrays of cell-

and cytokine-specific Ab spots were printed side-by-side so as to

position T-cells in the immediate vicinity of the immunosensors

for IFN-g and IL-2 (Fig. 1A,B). To further increase sensitivity of

the cytokine immunoassay and to minimize the blood volume

requirement, the microarrays were enclosed inside a microfluidic

chamber using reversible sealing strategy that was non-destruc-

tive to printed biomolecules (Fig. 1C,D). Incubation of Ab

microarrays with RBC-depleted whole human blood inside

a microfluidic chamber resulted in binding of CD4 and CD8

T-cells on their respective Ab spots next to Ab regions for

detection of IL-2 and IFN-g. Activation of T-cells followed by

immunofluorescent staining revealed cytokine concentration

gradients extending from T-cells. Overall, a microdevice enabling

isolation of pure T-cell subsets from a heterogeneous cell sample

and on-chip detection of cytokines has important applications in

vaccine development as well as diagnosis/treatment of HIV and

other infections.

Experimental

Materials

10� Phosphate-buffered saline (PBS) without calcium and

magnesium, paraformaldehyde (PFA), surfactant TWEEN� 20,

rabbit anti-mouse IgG antibody (2nd labeling antibody)

Na4EDTA, KHCO3, NH4Cl, poly(ethylene glycol)diacrylate

(PEG-DA) (MW575), anhydrous toluene (99.9%), sodium azide,

bovine serum albumin (BSA), 0.1% poly-L-lysine were purchased

from Sigma-Aldrich (Saint Louis, MO). Silane adhesion

promoter, 3-acryloxypropyl trichlorosilane, was from Gelest,

Inc. (Morrisville, PA). Monoclonal antibodies used for capturing

T-lymphocytes and cytokines consisted of the following: purified

2198 | Lab Chip, 2008, 8, 2197–2205

mouse anti-human CD4 Abs (13B8.2) and CD8 (B9.11) from

Beckman-Coulter (Fullerton, CA), and purified mouse anti-

human IFNg Ab (clone K3.53), anti-IL2 Ab (clone 5355), bio-

tinylated goat anti-human IFN-g Ab and biotinylated anti-IL2

Ab from R & D Systems (Minneapolis, MN). Mouse IgG2a

(OX34) was purchased from Serotec Antibodies (Raleigh, NC).

Antibodies used for immunostaining of surface bound cells or

flow cytometry analysis listed below: anti-human CD33 -FITC

(UCHT1), anti-CD4-PE (L120) and anti-CD8-PE (RPA-T8)

were all purchased from BD Pharmingen. Streptavidin conju-

gated with Alexa 546 was purchased from Invitrogen (Carlsbad,

CA). Human recombinant IFN-g and IL-2 were from R & D

Systems (Minneapolis, MN) and Endogen (Woburn, MA),

respectively. T-cell activation reagents: Phorbol 12-myristate 13-

acetate (PMA), Phytohemagglutinin (PHA), Staphylococcal

enterotoxin B (from Staphylococcus aureus) and ionomycin were

purchased from Sigma-Aldrich. Cell culture medium RPMI

1640: 1X, with L-glutamine was purchased from VWR. Poly-

(dimethylsiloxane) (PDMS) and its curing agents were purchased

from Dow Corning (Midland, MI).

Fabrication of multi-functional Ab microarrays

Robotic printing of protein microarrays on PEG gel-coated glass

slides was described previously.24 Briefly, a standard microscope

slide (75� 25 mm) was modified with acrylated silane and coated

with poly(ethylene glycol)-diacrylate (PEG-DA)-based prepol-

ymer. Exposure to UV resulted in cross-linking of the pre-poly-

mer and formation of PEG hdyrogel layer on a glass slide. PEG

gel-coated glass slides were lyophilized for 24 h in order to ensure

rapid absorption and uniform distribution of Abmolecules in the

hydrogel upon printing. 8 � 20 microarray of Ab spots (�150–

250 mm diameter, 375 mm center-to-center spacing) was printed

on hydrogel-coated glass slides using contact microarrayer

(GMS 417 Affymetrix, Santa Clara, CA). Prior to printing, anti-

CD4, -CD8, -IFN-g, -IL-2 and mouse IgG (negative control)

Abs were dissolved in DI water at a concentration of 0.2 mg

mL�1 and were supplemented with BSA (0.5% v/v) and Tween20

(0.005% v/v) (anti-CD4 and -CD8 Abs contain only 0.005%

Tween20). Components of the microarray were loaded into

a 384-well plate (Genetix, Boston, MA) and then robotically

imprinted onto a hydrogel-coated glass slide. After printing,

surfaces were air dried and stored in a sealed box at 4 �C prior to

further use.

Fig. 1B shows design of the microarray with columns of anti-

CD4 and -CD8 Ab spots placed next to rows of cytokine

immunoassay spots. This microarray arrangement ensured that

all cytokine immunoassay spots were exposed to the same cyto-

kine concentration. In addition to capture Abs, cytokine

immunoassay microarray included several negative and positive

controls. Specifically, anti-mouse IgG-2a was employed as

a negative control while biotin molecules were printed to provide

a positive control for the binding of the streptavidin-Alexa-546

complex used during immunofluorescent labeling.

Design of a microfluidic platform

A microfluidic platform was developed to enable isolation of

T-cells from a small blood sample and to increase local

This journal is ª The Royal Society of Chemistry 2008

Fig. 1 (A) The conceptual design of microarrays for detection of T-cell-secreted cytokines. Printing of cell- and cytokine-specific Ab spots side-by-side

allowed one to capture T-cells next to IL-2 and IFN-g sensing regions. T-cell-secreted cytokines were detected on the adjacent anti-cytokine Ab spots.

(B) A map of the 8 � 20 microarray for capturing T-cells and detecting T-cell-secreted IL-2 and IFN-g. (C) Design of a microfluidic platform employed

for integration with Ab microarrays. The device containing two reaction chambers was secured on top of microarrays by applying vacuum suction

through a web of auxiliary channels. This approach allowed Ab microarrays to be enclosed inside a microfluidic chamber in a reversible and protein-

friendly manner. (D) An image of a PDMSmicrodevice employed for T-cell capture and cytokine detection experiments with one reaction chamber filled

with unlysed whole blood.

Dow

nloa

ded

by U

nive

rsity

of

Cal

ifor

nia

- D

avis

on

22 N

ovem

ber

2010

Publ

ishe

d on

30

Sept

embe

r 20

08 o

n ht

tp://

pubs

.rsc

.org

| do

i:10.

1039

/B81

0244

AView Online

concentration of secreted cytokines. Standard soft lithography

approaches were used to fabricate poly(dimethyl siloxane)

(PDMS)-based microfluidic devices.25 A transparency photo-

mask (CAD Art Services, Poway, CA) was generated based on

an AutoCAD drawing of the device. This photomask was then

employed to micropattern SU-8 on a 4 inch silicon (Si) wafer in

order to create a negative replica of the fluidic network. Poly(-

dimethylsiloxane) (PDMS) was mixed 10 : 1 with a curing agent,

poured onto a Si wafer containing SU-8 features and cured for 12

h at 60 �C. The elastomer with imbedded channel architecture

was released and inlet/outlet holes were punched with a blunt 16

gauge needle. The microfluidic device, shown in Fig. 1C, con-

tained two immunoreaction chambers with width–length–height

dimensions of 3 � 10 � 0.1 mm as well as a network of inde-

pendently addressed auxiliary channels. The auxiliary channels

were used to apply negative pressure (vacuum suction) to the

PDMS mold and reversibly secure it on top of a glass substrate.

This strategy of reversible sealing of microfluidic devices, first

described by Schaff et al.,26 allowed us to enclose protein

microarrays inside fluidic conduits without damaging or dena-

turing Ab molecules.

A 5 mL syringe was connected to silicone tubing (1/32 inch id,

Fisher) which was then attached to the outlet of the flow chamber

with a metal insert cut from a 20 gauge needle. A blunt shortened

This journal is ª The Royal Society of Chemistry 2008

20 gauge needle carrying a plastic hub with volume of 100 mLwas

inserted in the inlet. A pressure-driven flow in the microdevice

was created by withdrawing the syringe positioned at the outlet

using a precision syringe pump (Harvard Apparatus, Boston,

MA).

The shear stress (t) inside the channel was estimated assuming

an expression for an infinitely wide parallel-plate flow chamber:

t ¼ 6mQ

wh2(1)

where Q is flow rate, m is viscosity of the medium (approximately

10�2 dyn s cm�2), w is channel width and h is channel height. For

a channel 3 mm in width and 100 mm in height, flow rates of 3 and

50 mL min�1 would yield shear stress values of 0.1 and 1.67 dyn

cm�2, respectively. In our experiments, low flow rate was used for

seeding and capturing leukocytes on Ab microarrays, while

higher flow rate was employed for removal of non-specific cells.

Capture of T-cells on Ab microarrays

Red blood cell (RBC) depleted whole human blood was

employed in T-cell isolation experiments. Blood was collected

from healthy adult donors through venipuncture under sterile

conditions with informed consent and approval of the

Lab Chip, 2008, 8, 2197–2205 | 2199

Dow

nloa

ded

by U

nive

rsity

of

Cal

ifor

nia

- D

avis

on

22 N

ovem

ber

2010

Publ

ishe

d on

30

Sept

embe

r 20

08 o

n ht

tp://

pubs

.rsc

.org

| do

i:10.

1039

/B81

0244

AView Online

Institutional Review Board of the University of California at

Davis (protocol number 200311635–6). RBCs were removed

using ammonia chloride based erythrocyte lysis solution (89.9 g

NH4Cl, 10.0 g KHCO3, and 370.0 mg tetrasodium EDTA in 10 L

of deionized water) as described previously.24,27 RBC-depletion

produced blood cell suspension comprised of granulocytes,

peripheral blood mononuclear cells and RBC debris. The cells

were concentrated by centrifugation and re-suspended in

RPMI1640 medium containing 10% FBS and 1% penicillin/

streptomycin but without L-glutamine and Phenol Red (Medi-

atech, Herndon, VA). The leukocyte suspension was used

immediately for microfluidic cytokine detection experiments.

Prior to introduction of cells, PDMS molds containing fluidic

and vacuum channels were sterilized by 15 min UV exposure in

a tissue culture hood. Sterile 1� PBS was first injected into the

flow chamber to remove air bubbles, then 50 mL of RBC-lysed

blood resuspended in phenol red-free RPMI1640 was added into

the inlet reservoir (hub of a 20 gauge needle) and drawn into

a microfluidic channel at shear stress of 0.3 dyn cm�2 (9 mL

min�1). Upon entry of cells into a channel the shear stress was

decreased to 0.1 dyn cm�2 (3 mL min�1) and was maintained for

10 min allowing cells to interact with Ab microarrays. This time

was found by us to be sufficient for binding of cells on Ab spots.27

In order to wash away nonspecific cells while retaining T-cells

captured on Ab microarrays flow rate was increased to 50 mL

min�1, creating a shear stress of 1.7 dyn cm�2. The shear stress

window of 1–2 dyn cm�2 was previously found to be optimal for

removal of nonspecifically bound cells as well as contaminating

monocytes, while retaining CD4 and CD8 T-cells.24,28

Detection of T-cell secreted cytokines in a microfluidic device

After capturing T-cells, a microfluidic device was flushed with 1�PBS and filled with mitogenic agents to commence cell activation

and cytokine production. The mitogenic solution consisted of

PMA and ionomycin dissolved to concentrations of 50 ng mL�1

and 2 mM respectively in phenol red-free RPMI1640 media

supplemented with 10% FBS. A surgical clamp was secured

around the inlet/outlet tubing to eliminate convective mixing and

to establish diffusion gradient of cytokine concentration. A

microfluidic chamber with captured T-cell was then placed into

a tissue culture incubator (37 �C, 5% CO2 and 90% humidity) for

periods of time ranging from 1 to 6 h.

At the end of the desired activation period T-cells and anti-

cytokine Ab spots enclosed in a microfluidic chamber were

exposed to reagents for immunofluorescent staining of cells and

cytokines. After flushing away mitogenic solution with 1� PBS

fluidic chambers were filled with biotinylated anti-IFN-g and

-IL-2 Abs (5 mg mL�1 in 1� PBS) and incubated for 1 h at room

temperature. The chambers were then flushed with 1� PBS again

and filled with streptavidin-Alexa546 (10 mg mL�1 in 1� PBS) for

30 min in order to reveal the presence of biotin-Ab-cytokine-Ab

sandwich complex. Immunoassay strategy employed in this

study is described pictorially in Fig. 1A.

The immunofluorescently labeled anti-cytokine Ab spots were

visualized and imaged inside a fluidic chamber using a confocal

microscope (Zeiss LSM 5 Pascal, Carl Zeiss, Inc.). Quantifying

cytokine signals using a microarray scanner required removal of

a PDMS chamber. Prior to opening a microfluidic device cells

2200 | Lab Chip, 2008, 8, 2197–2205

adherent on Ab spots were fixed by exposure to 1% para-

formaldehyde (PFA) for 15 min. Glass slides containing T-cell

arrays and cytokine immunoassays were air dried and analyzed

using a microarray scanner.

Calibration curves were constructed to translate fluorescence

signal to concentration of IFN-g and IL-2. To construct cali-

bration curves, microarrays of anti-Il-2 and anti-IFN-gAbs were

incubated for 30 min with recombinant human IFN-g and IL-2

dissolved in 1� PBS supplemented with 0.5% v/v BSA and

0.005% Tween20. Cytokine concentration range tested was 1 to

400 ng mL�1. Importantly, printed arrays contained both types

of anti-cytokine Abs but were exposed to one type recombinant

cytokine to ensure lack of cross-reactivity between the assays.

These microarrays were incubated with biotinylated anti-cyto-

kine Abs and streptavidin-Alexa546 to reveal cytokine-concen-

tration dependent fluorescence signal. In order to create

a quantitative readout of fluorescence signal emanating from the

array, the laser microarray scanner was employed to scan the

glass slides at a spot pixel resolution of 5 mm (Agilent, Santa

Clara, CA). The fluorescence intensity of each array element was

determined using GenePix Pro 6.0 data analysis software

(Molecular Devices, Downingtown, PA).

Flow cytometry analysis of T-cell cytokine production

RBC-lysed whole blood was stimulated with single mitogens or

media alone (negative control) for 6 h at 37 �C. Brefeldin A

(Sigma, St. Louis, MO) was added for the final 5 h to prevent

Golgi transport of cytokines out of cells. Specifically the cells

were stimulated in a fashion identical to that described in the

previous section using 50 ng mL�1 PMA, or 2 mM mL�1 Ion-

omycin and incubated at 37 �C with 5% CO2 for 6 h.

Following stimulation, cells were washed in 1 mL of 1� PBS

for 5 min at 1600 rpm and incubated with Aqua (Amcyan) Live/

Dead Dye (Invitrogen, Carlsbad, CA) at a concentration of 0.5

mL million cells�1 in 1 mL of 1� PBS for 30 min at 4 �C. Cells

were washed with staining buffer (filter sterilized 1� PBS with

3% heat-inactivated FBS (Gibco) and 0.1% sodium azide

(Sigma)). Cells were re-suspended in staining buffer and incu-

bated with Abs for surface markers CD3 APC Cy7, CD4 APC

(Beckton Dickinson (BD), Mountain View, CA), and CD8 FITC

(Ebioscience, San Diego, CA) for 30 min 4 �C. Cells were then

fixed in 1% PFA for 15 min at room temperature. Fixative was

washed out and cells were permeabilized with Caltag Per-

meabilization Solution B (Invitrogen, Carlsbad, CA) and stained

with monoclonal antibodies IFN-g PE Cy7 (Ebioscience) and

IL-2 PE (BD) for 25 min at room temperature. Samples were

washed and fixed with 1% PFA before analysis with LSR II (BD)

at the UC Davis Optical Core Facility. A minimum of 300 000

events were collected for each sample and analyzed using Flow Jo

software (Treestar, Inc, San Carlos, CA).

Results and discussion

The goal of this study was to develop a strategy for on-chip

detection of cytokines secreted from captured T-cells. An

approach utilizing a printed microarray of cell- and cytokine-

specific Ab spots enclosed inside a microfluidic device enabled

placement of T-cells next to cytokine immunosensors inside

This journal is ª The Royal Society of Chemistry 2008

Dow

nloa

ded

by U

nive

rsity

of

Cal

ifor

nia

- D

avis

on

22 N

ovem

ber

2010

Publ

ishe

d on

30

Sept

embe

r 20

08 o

n ht

tp://

pubs

.rsc

.org

| do

i:10.

1039

/B81

0244

AView Online

a miniature fluidic chamber. Mitogenic stimulation of T-cells

then resulted in detection of IL-2 and IFN-g on the adjacent anti-

cytokine Ab spots (Fig. 1A). A miniature cytometry platform

described here may have a wide range of applications in immu-

nology research including monitoring/diagnostics of infectious

diseases and development of vaccines.

Integrating Ab microarrays into microfluidic devices

In order to minimize blood sample required for T-cell analysis

and to increase local concentration of secreted cytokines it was

important to enclose Ab microarrays inside a miniature flow-

through cell. Microfluidic devices have been used extensively for

cell sorting and manipulation25,29,30 and have recently found an

application in blood cell analysis.31 Several studies described the

use of Ab-modified microfluidic chambers for capturing model

immune cells as well as primary T-lymphocytes from heteroge-

neous cell suspensions.28,32,33 In these studies, microfluidic devices

were irreversibly sealed using oxygen plasma treatment and then

modified with a uniform layer of Ab molecules, thus, allowing

one to avoid exposure of Ab molecules to potentially destructive/

denaturing effects of oxygen plasma. In contrast, Abmicroarrays

employed for cell capture in our studies had to be printed prior to

assembly of microfluidics and had to be removed after cell

capture experiment for scanning of cytokine arrays. Therefore,

a reversible and protein-friendly method for sealing a micro-

fluidic chamber was required. Such a reversible seal was created

by applying negative pressure (vacuum suction) to PDMS mold

through a network of auxiliary channels, in a manner similar to

that described recently by Schaff et al.26 This method entailed

creating two networks of channels in PDMS, one for blood

Fig. 2 Immunofluorescent staining of cells captured on anti-CD4 and -CD8

clusters formed on 150 mm diameter anti-CD4 spots showing that captured c

identifying these cells as CD4+ T-cells. (C,D) Immunofluorescent image of a 3�showing that the majority of captured cells are stained with anti-CD3 FITC

T-lymphocyte phenotype.

This journal is ª The Royal Society of Chemistry 2008

sample analysis, another for applying vacuum, addressed with

separate inlet/outlet ports (see Fig. 1C for pictorial description).

An example of a microfluidic device containing two immuno-

reaction chambers (one of which is filled with blood) as well as

a network of auxiliary vacuum channels used to achieve revers-

ible seal between a PDMS device and a glass substrate is shown

in Fig. 1D. This microfluidic platform was employed for all cell

seeding, washing and immunofluorescent staining steps

described in the paper.

Capturing T-cells and detection of secreted cytokines on Ab

microarrays

It was important to ensure purity of the captured T-cell pop-

ulation in order to correctly assign a cytokine profile to a desired

cell subset. This requirement was motivated by the fact that

production of IL-2 and IFN-g may be associated with Th1

response of CD4 T-cells or cytotoxic function of CD8 T-cells.

Attaining high purity of the desired leukocyte type was highly

dependent on optimization of washing protocols in order to

remove loosely bound non-specific cells without dislodging CD4

or CD8 T-cells. Employing a microfluidic device for T-cell

capture experiments allowed to precisely control shear stress

inside a fluidic conduit. T-cell panning step was performed at

a low flow rate of 3 mL min�1 (0.1 dyn cm�2) while the washing

step was carried out at 50 mL min�1 (1.7 dyn cm�2). The latter

value of shear stress was previously shown to result in removal of

loosely bound non-specific cells and elimination of CD4-

expressing monocytes.24,27,28 Coupled with nonfouling properties

of PEG hydrogel coating, the washing procedure described

above allowed to isolate T-cells on Ab spots with minimal

Ab microarrays. (A,B) Fluorescent image of the 3 � 4 array of leukocyte

ells are stained with both anti-CD3 FITC (green) and anti-CD4 PE (red)

4 array of leukocyte clusters formed on 150 mm diameter anti-CD8 spots

(green) but not with anti-CD4 PE (red) pointing to CD3+CD4� or CD8+

Lab Chip, 2008, 8, 2197–2205 | 2201

Fig. 3 Detection of secreted cytokines using Ab microarrays. (A) Cells

residing on 300 mm diameter anti-CD4 Ab spots next to cytokine-sensing

spots after 6 h mitogenic activation. Fluorescent staining of the micro-

arrays with CD3 FITC (green), Dapi (blue) and anti-cyotkine Ab-biotin/

streptavidin-Alexa 546 (red) shows that CD3+ T-cells were captured on

cell-specific Abs and minimal cell attachment was observed on anti-

cytokine spots or elsewhere on a PEG hydrogel coated glass surface.

Mitogenic activation resulted in cytokine production detected with IFN-

g and IL-2 sensing spots (red). (B) CD4+ human T-cells captured next to

anti-IL-2 and –IFN-g Ab spots. Mitogenic stimulation of T-cells with

PMA and ionomycin was followed by incubation with biotinylated anti-

cytokine Abs, followed by incubation with streptavidin-Alexa546. There

2202 | Lab Chip, 2008, 8, 2197–2205

Dow

nloa

ded

by U

nive

rsity

of

Cal

ifor

nia

- D

avis

on

22 N

ovem

ber

2010

Publ

ishe

d on

30

Sept

embe

r 20

08 o

n ht

tp://

pubs

.rsc

.org

| do

i:10.

1039

/B81

0244

AView Online

nonspecific cell binding. Phenotypic purity of the captured cells

was analyzed by immunofluorescent staining. Representative

images shown in Fig. 2A,B demonstrate that cells captured on

anti-CD4 Ab spots were CD3+CD4+, definitely identifying them

as CD4 T-cells. Conversely, cells captured on anti-CD8 Ab spots

were CD3+CD4� which is logical given that CD4 antigen is not

expressed on CD8 T-cells (Fig. 2C,D). Because the majority

(over 95%) of mature T-cells express either CD4 or CD8 antigen,

the CD3+CD4� cells were assigned CD8 T-cell phenotype.

Direct immunostaining for CD8 antigen was performed by us

previously, confirming the presence of CD8+ T-cells on the Ab

spots.24

The capture of pure T-cell subsets next to cytokine immuno-

sensors inside a miniature (�3 mL) immunoreaction chamber was

followed by cell activation and cytokine detection steps. T-cells

were mitogenically stimulated using a combination of PMA and

ionomycin for a period of time ranging from 1 to 6 h under

physiological conditions (37 �C and 5% CO2). Activation with

PMA and ionomycin was previously reported to result in robust

production of IL-2 and IFN-g as verified by intracellular staining

and flow cytometry.34 During T-cell activation, flow in the

microfluidic channel was stopped and care was taken to eliminate

convective mixing, and ensure that diffusion was the sole mass

transport mechanism. Fig. 3A demonstrates immunofluorescent

labeling of CD4 T-cell- and cytokine-capture microarray after 6h

activation inside a microfluidic device. T-cells were stained with

anti-CD3 FITC (green) and Dapi (blue) whereas immnuno-

sensing spots were stained with biotinylated anti-cytokine Abs,

followed by streptavidin-Alexa 546 (red). Fig. 3A highlights

several important aspects of the cytokine detection platform: (1)

the majority of cells panned from RBC-depleted whole blood on

anti-CD4 Ab spots were CD3+, suggesting T-cell phenotype, (2)

minimal cell adhesion was observed on anti-cytokine Abs or

PEG hydrogel-coated glass substrate, underscoring the possi-

bility of placing pure cell population next to immunosensing

regions, (3) IL-2 and IFN-g could be detected using microarray

immunoassay format.

Fig. 3B,C demonstrates another cytokine detection experiment

where CD4 and CD8 T-cells were captured in different chambers

of a microfluidic device and were activated for 4 h. As seen from

Fig. 3B, activation of CD4 T-cells resulted in cytokine secretion

and binding on anti-IL-2 and anti-IFN-g Ab spots. A concen-

tration gradient in signal intensity was observed with signal

becoming undetectable � 2 mm away from the cytokine-

secreting T-cells. This underscores the importance of placing

immunosensing domains next to T-cells and miniaturizing the

reaction volume. Fig. 3B,C also illustrates a difference in cyto-

kine profiles of CD4 T-cells and CD8 T-cells, with former

producing both cytokines and latter producing only IFN-g. It

should be also be noted that cytokine production was observed

only when T-cells were mitogenically activated (data not shown).

The detection of IL-2 and IFN-g after 4 h mitogenic stimu-

lation described in Fig. 3B,C was confirmed by intracellular

was a clear fluorescence signature for both IL-2 and IFN-g secreted by

CD4 T-cells. (C) CD8+ human T-cells captured next to anti-IL-2 and

–IFN-g Ab spots. Mitogenic stimulation of these cells pointed to the

presence of IFN-g and lack of IL-2 production.

This journal is ª The Royal Society of Chemistry 2008

Fig. 4 Analysis of cytokine production using flow cytometry. RBC-depleted whole blood was mitogenically activated and cytokine production was

analyzed by intracellular staining and flow cytometry. Lymphocyte population determined by forward and side scatter was gated to single cells and then

further gated to CD3+ T-cells. (A) Flow cytometry analysis of IFN-g and IL-2 in CD4+ T-cell population points to robust production of both cytokines

by this leukocyte subset. (B) Flow cytometry analysis showed that CD8+ T-cells robustly produced IFN-g but not IL-2. These data corroborate results

observed with our microarray immunosensing strategy.

Dow

nloa

ded

by U

nive

rsity

of

Cal

ifor

nia

- D

avis

on

22 N

ovem

ber

2010

Publ

ishe

d on

30

Sept

embe

r 20

08 o

n ht

tp://

pubs

.rsc

.org

| do

i:10.

1039

/B81

0244

AView Online

staining and flow cytometry of the same blood sample. As shown

in Fig. 4A, 45% of CD4 T-cells stained positive for intracellular

IL-2 while 28% were positive for IFN-g. In contrast, as seen from

Fig. 4B, CD8 cells were observed to have a robust IFN-g

response (39%) but limited IL-2 production (4%). Therefore,

microarray- and flow cytometry-based detection methods

showed similar patterns in cytokine production by CD4 and CD8

T-cells. However, the flow cytometry reported the frequency of

appearance of cytokine-producing cells and did not reveal the

amount of cytokines produced by the cells. On the other hand,

microarray-based cytokine immunoassays enabled us to quantify

the concentration of cytokines produced by the captured T-cells.

These experiments are described in the following section.

Quantifying T-cell cytokine production

The use of the microarray scanner allowed us to rapidly and

quantitatively analyze fluorescence signals emanating from

immunosensing regions. Fig. 5A shows a region of the T-cell/Ab

microarray with a tracer line across cytokine sensing spots. These

line scans were converted into fluorescence intensity profiles

using a microarray scanner. Fig. 5B demonstrates scanned

fluorescence intensity data from anti-mouse IgG (negative

control), anti-Il-2 and anti-IFN-g spots located next to cells in

experiments where mitogenic activation of CD4 T-cells was

varied from 1 to 6 h. As seen from these data, IFN-g and IL-2

signal intensity increased as a function of activation time;

This journal is ª The Royal Society of Chemistry 2008

however, no IL-2 signal was observed after 1 h of mitogenic

activation. It should be noted that the negative control (mouse

IgG) spots had a low background signal contributing to high

signal-to-noise ratio of immunosensing microarrays.

In order to convert arbitrary fluorescence intensity units into

cytokine concentration, calibration curves using known amounts

of recombinant IFN-g and IL-2 were constructed. Fig. 5C,D

shows concentration dependent modulation in fluorescence

intensity of anti-IFN-g and anti-IL-2 spots. The higher sensi-

tivity of IFN-g immunosensor compared to IL-2 may be due to

differences in association/dissociation kinetics of antibody–

antigen interactions. Calibration curve for IFN-g was fitted with

a logarithmic best fit while IL-2 concentration was a linear

function of fluorescence with R2 ¼ 0.96 for both curves.

Employing calibration curves, the fluorescence intensity profiles

presented in Fig. 5B were converted into cytokine concentration,

thus allowing us to establish the amount of IL-2 and IFN-g

secreted by CD4 T-cells after varying extents of mitogenic acti-

vation. As shown in Fig. 5E, IFN-g concentration was estimated

to be �18 ng mL�1 after 1 and 4 h of mitogenic activation, and

increased further to �230 ng mL�1 after 6 h of stimulation inside

a microdevice. No IL-2 production was observed after 1 h

stimulation; however, this cytokine was detected after 4 h of

activation (70 ng mL�1) and was strongly expressed after 6 h of

stimulation (360 ng mL�1). The data presented in Fig. 5 point to

the possibility of using microarray immunoassays placed next to

arrays of T-cells for on-chip detection of secreted cytokines.

Lab Chip, 2008, 8, 2197–2205 | 2203

Fig. 5 Quantifying cytokine production by captured T-cells. (A) Staining of microarrays with biotinylated anti-cytokine Abs followed by streptavidin-

Alexa 546 revealed strong fluorescence signals from anti-IL-2 and anti-IFN-gAb spots and no signal from mouse IgG serving as a negative control. The

white trace shows a line scan from a microarray scanner. (B) Scans of fluorescence intensity profiles for different times of mitogenic activation of CD4

T-cells. (C–D) Calibration curves for converting fluorescence intensity into concentration of IFN-g and IL-2. (E) Concentration of IL-2 and IFN-g

detected from CD4 T-cells activated for different time periods. The concentration values correspond to fluorescence intensity scans shown in part (B).

Dow

nloa

ded

by U

nive

rsity

of

Cal

ifor

nia

- D

avis

on

22 N

ovem

ber

2010

Publ

ishe

d on

30

Sept

embe

r 20

08 o

n ht

tp://

pubs

.rsc

.org

| do

i:10.

1039

/B81

0244

AView Online

Another advantage of using microarray format for cytokine

detection is the simplicity of the signal readout. In our strategy,

anti-cytokine Abs, positive/negative controls and cytokine refer-

ence points were printed in discrete locations of the substrate so

that only one fluorescence color was needed to discern all signals.

In addition, using cell-specific microarrays allowed us to capture

different leukocyte subsets on defined Ab spots and to identify

a cell subset solely based on ‘‘location’’ within the microarray

without the use of fluorescence. In contrast, flow cytometry

analysis of multiple cytokines produced by different leukocyte

subsets requires each cell subset to have surface antigen- and

cytokine-specific fluorescence signature, necessitating the use of

complex instrumentation for multi-parametric analysis.

Conclusion

This paper describes the development of a microdevice for

isolating pure T-cell subsets and detecting cell-secreted cytokines:

IL-2 and IFN-g. Printedmicroarrays of cell- and cytokine-specific

Ab spots were enclosed inside a microfluidic device and were

employed to capture pure T-cell subsets next to cytokine-sensing

domains. Mitogenic activation of T-cells followed by immuno-

fluorescent staining of the microarrays revealed fluorescence

signals due to binding of IL-2 and IFN-g on anti-cytokine Ab

spots. An immunosensing strategy described here offers several

2204 | Lab Chip, 2008, 8, 2197–2205

advantages: (1) Ab microarrays allowed us to perform T-cell

sorting and cytokine detection in the same microdevice, elimi-

nating an off-chip leukocyte purification step employed by

conventional immunological methods, (2) placement of cells near

cytokine immunosensors inside a microfluidic chamber ensured

high local concentration of the analyte, (3) capturing pure

leukocyte subsets on specific Ab spots within the microarray

allowed us to identify cell phenotype solely based on location and

eliminated the need for fluorescent staining of cells, (4) printing

cytokine-sensing Abs in discrete locations of the surface enabled

the use of single fluorophore for microarray analysis. In the

future, a miniature cytometry platform described here may be

employed for multi-parametric analysis of T-cell function in

diagnosis and monitoring of infectious disease such as HIV.

Acknowledgements

We thank Prof. Louie’s lab in the Department of Biomedical

Engineering at UC Davis for providing assistance with confocal

microscopy. Scanning of the microarrays was performed at

Expression Analysis Facility at UC Davis. Financial support for

this work was provided in part by the California Research Center

for the Biology of HIV in Minorities, California HIV/AIDS

Research Program #CH05-D-606. Additional financial support

was provided in part by an NIH grant DK61297 awarded to SD.

This journal is ª The Royal Society of Chemistry 2008

Dow

nloa

ded

by U

nive

rsity

of

Cal

ifor

nia

- D

avis

on

22 N

ovem

ber

2010

Publ

ishe

d on

30

Sept

embe

r 20

08 o

n ht

tp://

pubs

.rsc

.org

| do

i:10.

1039

/B81

0244

AView Online

HZ was supported through an NIH Training Grant (EB003827).

GS was supported through a Biotechnology Fellowship from

National Center for Biotechnology, Republic of Kazakhstan.

References

1 S. Romagnani, Clin. Immunol. Immunopathol., 1996, 80(3), 225–235.2 P. Salgame, J. S. Abrams, C. Clayberger, H. Goldstein, J. Convit,R. L. Modlin and B. R. Bloom, Science, 1991, 254(5029), 279–282.

3 G. Pantaleo and R. A. Koup, Nat. Med., 2004, 10, 806–810.4 M. Clerici, F. T. Hakim, D. J. Venzon, S. Blatt, C. W. Hendrix,T. A. Wynn and G. M. Shearer, J. Clin. Invest., 1993, 91(3), 759–765.

5 M. Clerici and G. M. Shearer, Immunol. Lett., 1996, 51, 69–73.6 P. Shankar, M. Russo, B. Harnisch, M. Patterson, P. Skolnik andJ. Lieberman, Blood, 2000, 96, 3094–3101.

7 G. Pantaleo and A. Harari, Nat. Rev. Immunol., 2006, 6, 417–423.8 J. H. Cox, G. Ferrari and S. Janetzki, Methods, 2006, 38, 274–282.9 A. C. Karlsson, J. N. Martin, S. R. Younger, B. M. Bredt, L. Epling,R. Ronquillo, A. Varma, S. C. Deeks, J. M. McCune, D. F. Nixonand E. Sinclair, J. Immunol. Methods, 2003, 283, 141–153.

10 V. C. Maino and L. J. Picker, Cytometry, 1998, 34(5), 207–215.11 R. E. M. Guerkov, O. S. Targoni, C. R. Kreher, B. O. Boehm,

M. T. Herrrera, M. Tary-Lehmann, P. V. Lehmann andS. K. Schwander, J. Immunol. Methods, 2003, 279, 111–121.

12 G. MacBeath and S. L. Schreiber, Science, 2000, 289, 1760–1763.13 J. B. Delehanty and F. S. Ligler, Anal. Chem., 2002, 74, 5681–5687.14 C. R. Taitt, G. P. Anderson, B. M. Lingerfelt, M. J. Feldstein and

F. S. Ligler, Anal. Chem., 2002, 74(23), 6114–6120.15 J. R. Falsey, M. Renil, S. Park, S. Li and K. S. Lam, Bioconjugate

Chem., 2001, 12, 346–353.16 Y. Li, N. Nath and W. M. Reichert, Anal. Chem., 2003, 75, 5274.

This journal is ª The Royal Society of Chemistry 2008

17 Y. Li and W. M. Reichert, Langmuir, 2003, 19, 1557–1566.18 C. C. Wang, J. Proteome Res., 2002, 1, 337–343.19 L. Belov, O. de la Vega, C. G. dos Remedios, S. P. Mulligan and

R. I. Christopherson, Cancer Res., 2001, 61, 4483–4489.20 A. Revzin, P. Rajagopalan, A. W. Tilles, F. Berthiaume,

M. L. Yarmush and M. Toner, Langmuir, 2004, 20, 2999–3005.21 C. J. Flaim, S. Chien and S. N. Bhatia, Nat. Methods, 2005, 3, 119.22 D. S. Chen, Y. Soen, T. B. Stuge, P. P. Lee, J. S. Weber, P. O. Brown

and M. M. Davis, PLoS Med., 2005, 2(10), 1018–1030.23 R. C. Bailey, G. A. Kwong, C. G. Radu, O. N. Witte and J. R. Heath,

J. Am. Chem. Soc., 2007, 129, 1959–1967.24 H. Zhu, M. Macal, M. D. George, S. Dandekar and A. Revzin, Anal.

Chim. Acta, 2008, 608, 186–196.25 G.M.Whitesides, E. Ostuni, S. Takayama, X. Jiang andD. E. Ingber,

Annu. Rev. Biomed. Eng., 2001, 3, 335–373.26 U. Y. Schaff, M. M. Q. Xing, K. K. Lin, N. Pan, N. L. Jeon and

S. I. Simon, Lab Chip, 2007, 7(4), 448–456.27 K. Sekine, A. Revzin, R. G. Tompkins and M. Toner, J. Immunol.

Methods, 2006, 313, 96–109.28 X. H. Cheng, D. Irimia, M. Dixon, K. Sekine, U. Demirci, L. Zamir,

R. G. Tompkins, W. Rodriguez and M. Toner, Lab Chip, 2007, 7(2),170–178.

29 D. Irimia and M. Toner, Lab Chip, 2006, 6, 345–352.30 C. E. Sims and N. L. Allbritton, Lab Chip, 2007, 7, 423–440.31 M. Toner and D. Irimia, Annu. Rev. Biomed. Eng., 2005, 7, 77–103.32 S. K. Murthy, A. Sin, R. G. Tompkins and M. Toner, Langmuir,

2004, 20, 11649–11655.33 A. Sin, S. K. Murthy, A. Revzin, R. G. Tompkins and M. Toner,

Biotechnol. Bioeng., 2005, 91, 816–826.34 A. Rodriguez-Caballero, A. Garcia-Montero, C. Bueno, J. Almeida,

R. Varro, R. Chen, A. Pandiella and A. Orfao, Lab. Invest., 2004,84, 1387–1398.

Lab Chip, 2008, 8, 2197–2205 | 2205