design of artificial microenvironment for cord blood cd34

93
Design of Artificial Microenvironment for Cord Blood CD34+ Cells Expansion ex vivo Yue Zhang (M.Sc. Bioengineering, NUS) A THESIS SUBMITTED FOR THE DEGREE OF MASTER OF SCIENCE GRADUATE PROGRAM OF BIOENGINEERING NATIONAL UNIVERSITY OF SINGAPORE 2005 brought to you by CORE View metadata, citation and similar papers at core.ac.uk provided by ScholarBank@NUS

Upload: others

Post on 08-Feb-2022

4 views

Category:

Documents


0 download

TRANSCRIPT

Design of Artificial Microenvironment for Cord Blood

CD34+ Cells Expansion ex vivo

Yue Zhang

(M.Sc. Bioengineering, NUS)

A THESIS SUBMITTED

FOR THE DEGREE OF MASTER OF SCIENCE

GRADUATE PROGRAM OF BIOENGINEERING

NATIONAL UNIVERSITY OF SINGAPORE

2005

brought to you by COREView metadata, citation and similar papers at core.ac.uk

provided by ScholarBank@NUS

Acknowledgements

This thesis is the result of one and half years of work whereby I have been

accompanied and supported by many people. It is a pleasant aspect that I have now

the opportunity to express my gratitude for all of them.

The first people I would like to thank are my supervisors Professor Teoh Swee

Hin and Professor Kam W. Leong, who always kept an eye on the progress of my

work and always were available when I needed advice. Their enthusiasm on research

and mission for providing high-quality work has made a deep impression on me. I

owe them lots of gratitude for having me shown this way of research. I also would

like to thank the helpful discussion of Professor Ian McNiece, Professor Hai-Quan

Mao, and Dr Chai Chou.

My colleagues of Tissue Therapeutic Engineering lab in Division of Johns

Hopkins in Singapore gave me a lot of help in research and life. I would like to thank

Dr Xue-Song Jiang, for many discussions and collaboration in animal work. He also

provided me advice and tips that helped me a lot in staying at the right track. I thank

Mr Kian-Ngiap Chua, for the valuable technical advice in SEM imaging. I thank Mr

Roy Lee and Ms Peggy Tang for being good colleagues during the last one and half

years. They helped me a lot in animal experiment. I also want to specially thank Mr

Thong-Beng Lu and Mr Yan-Nan Du from National University of Singapore, XPS

staff from Institute of Materials Research & Engineering, for their valuable technical

support in AFM and Confocal imaging and XPS test.

In the end, I would like to thank the members of the school council for their

valuable input. I also want to thank Agency for Science, Technology and Research

(A*STAR) of Singapore and the Division of Johns Hopkins in Singapore who funded

the thesis project.

i

Table of Contents

Acknowledgements……………...…………………………………..……..………..... i

Table of Contents……………………………………………………………………...ii

Summary.......................................................................................................................vi

List of Tables..............................................................................................................viii

List of Figures...............................................................................................................ix

List of Symbols.............................................................................................................xi

Chapter 1 Introduction

1.1 Purpose.....................................................................................................................1

1.2 Motivation................................................................................................................2

1.3 Objectives and Scope...............................................................................................3

Chapter 2 Literature Survey

2.1 Review of HSC and clinic application.....................................................................5

2.1.1 Hematopoiesis and HSC..................................................................................5

2.1.2 HSC clinic application.....................................................................................6

2.1.3 Different sources of HSC in transplantation...................................................8

2.2 Review of HSC expansion.....................................................................................10

2.2.1 Selection of candidate cells for expansion....................................................10

2.2.2 Evaluation of ex vivo expansion...................................................................11

2.2.3 Expansion systems........................................................................................12

2.2.3.1 Stroma dependent expansion................................................................12

2.2.3.2 Stroma independent expansion.............................................................14

2.3 Review of HSC microenvironment........................................................................15

2.3.1 Cytokine microenvironment..........................................................................15

2.3.2 Stroma microenvironment.............................................................................18

ii

2.3.3 ECM microenvironment................................................................................19

2.3.4 3D topography...............................................................................................19

2.4 Protein immobilization and bioactivity study........................................................21

Chapter 3 Materials and methods

3.1 Materials:....................................................................................................................23

3.1.1 Biological materials...........................................................................................23

3.1.2 Chemical materials............................................................................................23

3.2 MSC and CB CD34+ cells co-culture ......................................................................24

3.2.1 MSC culture......................................................................................................24

3.2.2 Ex vivo expansion of CB CD34+ cells in co-culture systems.........................24

3.2.3 CFC....................................................................................................................26

3.2.4 Animal engraftment...........................................................................................26

3.3 MSC and CB CD34+ cells 3D co-culture..................................................................27

3.3.1 MSC culture in 3D system and assays..............................................................27

3.3.2 Ex vivo expansion of CB CD34+ cells in 3D co-culture systems and

functional assays...............................................................................................28

3.4 Study of FN immobilization bioactivity....................................................................28

3.4.1 FN conjugation and adsorption.........................................................................28

3.4.2 Chemical and physical characterization of the surface.................................29

3.4.3 Biological characterization of the surfaces...................................................30

3.5 Statistical analysis..................................................................................................32

Chapter 4 MSC and CB CD34+ cells co-culture

4.1 Screening of co-culture conditions.............................................................................33

4.1.1 MSC metabolic level in StemSpan medium..................................................33

4.1.2 Optimize expansion scale..............................................................................33

iii

4.1.3 Co-culture over MSC feeder layer................................................................35

4.1.3.1 Irradiated and non irradiated feeder layer............................................35

4.1.3.2 Feeder layer exchanged and non exchanged co-culture ......................36

4.1.4 Co-culture over MSC pellet..........................................................................36

4.2 Study of the contact and non-contact co-culture...................................................38

4. 3 Function assays of contact and non-contact co-culture.........................................40

4.3.1 Phenotype screening of the expanded cells I.................................................40

4.3.2 CFC I.............................................................................................................42

4.3.3 Animal engraftment I........................................................................................42

4.4 Discussion..............................................................................................................44

Chapter 5 MSC and CB CD34+ cells 3D co-culture

5.1 Screening of 3D scaffolds......................................................................................47

5.2 Study of 3D and 2D co-culture..............................................................................48

5.3 Functional assays of 3D and 2D co-culture...........................................................49

5.3.1 Phenotype screening of the expanded cells II..................................................49

5.3.2 CFC II...........................................................................................................51

5.3.3 Animal engraftment II...................................................................................52

5.4 3D culture of MSC.....................................................................................................53

5.5 Discussion..............................................................................................................54

Chapter 6 Study of FN immobilization bioactivity

6.1 Physical and chemical characterization of conjugated and adsorbed FN..............58

6.1.1 FN immobilization efficiency.......................................................................58

6.1.2 XPS assay......................................................................................................59

6.1.3 Surface contact angle measurements.............................................................60

6.1.4 AFM assay.....................................................................................................60

iv

6.2 Biological characterization of conjugated and adsorbed FN..................................62

6.2.1 ELISA test for RGD domain.........................................................................62

6.2.2 Ex vivo cell experiment.................................................................................63

6.3 Discussion..............................................................................................................63

6.3.1 FN conformation and its bioactivity........................................................63

6.3.2 FN adsorption..........................................................................................64

6.3.3 FN conjugation and fibrillogenesis.........................................................64

Chapter 7 Conclusions

7.1 Conclusions……………………………..…………………………………………68

7.2 Future prospect……………………………………………………………….……71

References........................................................................................................................73

v

Summary

Allogeneic transplantation of CB is limited by insufficient number of HSC as

well as PHPC. An efficient ex vivo expansion of CB can overcome this limitation. An

ideal ex vivo expansion condition should mimic the in vivo hematopoietic

microenvironment, which can efficiently expand the cell number while still

maintaining the “stemness” of the cells.

MSC, as non hematopoietic, well-characterized skeletal and connective tissue

progenitor cells within the bone marrow stroma, have been investigated as support

cells for the culture of HSC/PHPC. They are attractive for the rich environmental

signals that they provide and their immunologic compatibility and supporting effect in

transplantation. In this study we clearly demonstrated the benefit of MSC inclusion in

HSC expansion ex vivo. We also compared the contact and non-contact cultures of

the two cell types; the cell-cell direct interaction was necessary for the efficient

expansion of HSC.

HSC-MSC co-cultures have only been performed in 2D configuration. We

postulate that a 3D culture environment that resembles the natural in vivo

hematopoietic compartment would be more conducive for regulating HSC expansion.

The result showed that the direct contact between MSC and HSC in 3D cultures led to

statistically-significant higher expansion of CB CD34+ cells compared to 2D co-

cultures: 891- versus 545-fold increase in total cells, 96- versus 48-fold increase of

CD34+ cells, and 230- versus 150-fold increase in CFC. NOD/SCID mouse

engraftment assays also indicated a high success rate of hematopoiesis reconstruction

with these expanded cells. This enhanced expansion result was probably due to the

ECM secreted by MSC, especially FN, which facilitated the interaction with the

seeded CD34+ cells.

vi

Based on this finding, we hypothesize that the immobilization of ECM protein

FN onto the surface of culture systems might represent another stroma free expansion

strategy. A key problem in FN immobilization is the change of bioactivity during the

immobilization process. By comparing the FN bioactivity after two common

immobilization pathways: conjugation and adsorption, we found that the adsorption

method maintained a normal conformation of FN, leading to less change of its

bioactivity in the access of RGD domains and cell adhesion ability. The covalent

conjugation process, on the other hand, induced FN unfolding and fibrillogenesis ex

vivo, which blocked the access of active RGD domains and suppressed fibroblast

cells adhesion. This bioactivity variance on the level of immobilization methods may

provide us a way to control the function of the immobilized protein, which can be

helpful for the construction of an artificial niche.

vii

List of Tables

Number Page

1. Surface markers expression of expanded cells I ...................................................41

2. The engraftment and lineage expression by human cells in the bone marrow of NOD/SCID mice I..................................................................................................43

3. Surface markers expression of expanded cells II ..................................................50

4. The engraftment and lineage expression by human cells in the bone marrow of NOD/SCID mice II.................................................................................................53

5. Element proportion on FN immobilized surface....................................................60

viii

List of Figures

Number Page

1. Hematopoietic system hierarchy..................................................................................6

2. Scheme of FN immobilization on aminated PET surface.........................................29

3. MSC metabolic level in MSCGM medium or Stemspan serum free medium with cytokines.................................................................................................................34

4. CB CD34+ cells expansion in 96-well or 24-well tissue culture plates..................34

5. CB CD34+ cells expansion on irradiated or non irradiated MSC feeder layers... 35

6. CB CD34+ cells expansion with or without exchange of MSC feeder layer in the middle of co-culture...................................................................................................36

7. CB CD34+ cells expansion on MSC feeder layer or adherent MSC pellet..............37

8. MSC pellet morphology under phase contrast microscope.......................................37

9. CB CD34+ cells expansion in contact co-culture or non-contact co-culture.........39

10. Cell-cell contact study by CMFDA and CD34 expression.......................................39

11. CFC properties of expanded cells I............................................................................42

12. SEM image of PET meshes and co-culture in meshes...........................................47

13. CB CD34+ cells expansion in PET woven or unwoven mesh...............................48

14. CB CD34+ cells expansion in 2D or 3D systems..................................................49

15. CFC properties of expanded cells II...........................................................................51

16. MSC attachment, proliferation and protein production in 2D or 3D systems...........54

17. SEM and Confocol image of MSC culture and co-culture in 2D or 3D system.......55

18. FN immobilization efficiency....................................................................................59

19. XPS survey of FN immobilized PET surface............................................................59

20. Surface contact angle on clean, modified or FN immobilized PET surface.............61

21. AFM image of FN immobilized PET surface............................................................61

22. ELISA measurement of active RGD domains in immobilized FN compared with Micro-BCA result.......................................................................................................62

23. Cell adhesion on FN immobilized PET surface.........................................................63

ix

24. Hypothetical schemes of FN fibrillogenesis in conjugation process ........................66

x

List of Symbols

BM: Bone marrow

BMP: Bone morphogenetic protein

CB: Cord blood

CFC: Cloning forming cell

CFU-E: Colony forming unit erythrocyte colony-forming units

CFU-GEMM: Colony forming unit granulocyte, erythrocyte, monocyte,

megakaryocyte

CFU-GM: Colony forming unit granulocyte/macrophage colony-forming units

CLP: Common lymphocyte progenitor

CMP: Common megacytes progenitor

ECM: Extra cellular matrix

EPO: Erythropoietin

FBS: Fetal bovine serum

FGF: Fibroblasts growth factor

FL: Flt-3 ligand

FN: Fibronectin

G-CSF: Granulocyte-colony stimulating factor

GM-CSF: Granulocyte/macrophage colony-stimulating factor

GMP: Granulocyte / monocyte progenitor

GVHD: Graft-versus-host disease

HGF: Hematopoietic growth factor

HLA: Human leukocyte antigen

HSC: Hematopoietic stem cells

IL: Interleukin

xi

LIF: Leukemia inhibitory factor

LTC-IC: Long-term culture initiating cell

LTR: Long term repopulating

M-CSF: Macrophage-colony stimulating factor

MkEP: Megakaryocytes, erythrocytes progenitor

MNC: Mono-nucleus cells

MSC: Mesenchymal stem cells

NOD/ SCID: Non-obese diabetic/ severe combined immunodeficiency

PB: Peripheral Blood

PET: Polyethylene terephthalate

PHPC: Primitive hematopoietic progenitor cell

SCF: Stem cell factor

STR: Short term repopulating

TCPS: Tissue culture plates

TGF: Transforming growth factor

TNF: Tumor necrosis factor

TPO: Thrombopoietin

VLA-4/5: Very late antigen-4/5

2D: Two dimensional

3D: Three dimensional

xii

CHAPTER 1

INTRODUCTION

1.1 Purpose

All mature cells in the blood and immune system are derived from HSC and

all of these cell types can be generated from a single HSC [Osawa et al (1996)]. This

tremendous potential for reconstituting the hematopoietic system has allowed the

development of transplantation of HSC as a clinical strategy. HSC obtained directly

from the patient (autologous HSC) are used for rescuing the patient from the effects of

high doses of chemotherapy or used as a target for gene therapy vectors. HSC

obtained from another person (allogeneic HSC) are used to treat hematological

malignancies by replacing the malignant hematopoietic system with normal cells.

Allogeneic HSC can be obtained from siblings (matched sibling transplants), parents

or unrelated donors (mismatched unrelated donor transplants). Currently, about

45,000 patients each year are treated by HSC transplantation, a number that has been

increasing during the past decade.

There are several different sources for obtaining HSC for transplantation. One

important new source of HSC is CB that is collected during newborn deliveries. In

addition to their widespread availability, these HSC have several useful properties,

including their decreased ability to induce immunological reactivity against the

patient because of increased levels of immune tolerance in the fetus. Interest in this

approach has increased since the first successful transplantation of cord-blood HSC in

1988 [Gluckman et al (1989)], and there are now an estimated 70,000 units of CB that

are stored and available for transplantation [Gluckman (2001)]. However, their use is

limited by the number of HSC that can be collected, and it is clear that engraftment is

1

closely correlated with the number of cells that are infused [Wagner et al (2002)]. For

optimum engraftment, current transplantation requires at least 2,500,000 CD34+ cells

per kilogram of patient body weight [Koller et al (1993)]. An optimally collected CB

donation generates approximately 10,000,000 CD34+ cells, which is just 10-15% of

the optimal dose for an adult. For this reason, CB transplantation is limited to

pediatric patients right now. Furthermore, because the stem cells in CB are more

primitive than those in marrow, the hematopoietic recovery using CB stem cells is

approximately 25 days for neutrophils and 43 days for platelets [Leary et al (1988)],

which is significantly slower than that obtained in BM transplantation. And it leaves

the patient vulnerable to a fatal infection for a longer period of time.

To address these limitations, an effective strategy to expand CB cells ex vivo,

increasing stem cells for transplantation would considerably improve the therapeutic

potentials. What’s more, the ex vivo expansion produces more mature precursors,

supplementing stem cells graft, which help to shorten and potentially prevent

chemotherapy induced pancytopenia. The purpose of the research presented here is to

develop a more compatible and efficient system of increasing CB ex vivo expansion,

while still keeping the stem cells properties.

1.2 Motivation

The ability of environmental cues to affect stem cell fate is a concept from

embryology that has been extended to the adult organism with the notion that a highly

specialized set of conditions exists to regulate the ultimate fate of anatomically

restricted primitive cells. The physical interaction of stem cells with stroma cells,

matrix components and supportive factors is thought to define a niche critical for the

homeostatic maintenance of stem cell populations. For example the HSC changes the

2

location during development moving from extra embryonic regions to the aorto-

gonadal-mesonephros in the embryo and then to the liver in the first trimester. By the

second trimester, BM, spleen and thymus become populated and eventually become

the exclusive sites of hematopoiesis. With each change in location is an associated

change in the production emphasis of the stem cell population, with stem cell

expansion and erythropoiesis in great dominance early and a more balanced, full

immune repertoire of expansion later. Inspired by the research in hematopoiesis

microenvironment, we planned to reconstruct the hematopoiesis niche ex vivo for

more reliable expansion of CB cells.

1.3 Objectives and Scope

The objective of reconstructing hematopoiesis microenvironment ex vivo for

more efficient HSC expansion is achieved by the following specific approaches:

• The incorporation of MSC as stroma in supporting CB expansion (Chapter 4).

MSC are well characterized skeletal and connective tissue progenitor cells

within bone marrow. They can provide rich environment signals and show good

immunological compatibility in transplantation. For these reasons, we incorporated

MSC into expansion system. First of all, we screened and optimized the co-culture

conditions, and then we studied on the direct cell-cell contact effect during the co-

culture. The expansion result was evaluated by functional assays ex vivo and in vivo.

• The incorporation of 3D scaffold in co-culture of CB and MSC (Chapter 5).

We postulated that 3D culture environment that resembles the natural niche

would be more conductive for CB expansion, thus we incorporated the 3D scaffold

into MSC and CB co-culture system. We first screened and optimized the 3D scaffold

for co-culture, and then we compared the 2D co-culture with 3D co-culture through

3

expansion and functional assays. Finally we studied MSC culture and its ECM

secretion in 3D scaffold, which probably explained the higher expansion result in 3D.

• The bioactivity study of immobilized FN, as a supportive research for its

further application in CB expansion (Chapter 6)

Inspired by the co-culture result that FN was specially secreted by MSCs in

supporting CB’s expansion, we hypothesized that the immobilization of FN onto the

surface of culture system may represent another stroma free expansion strategy. A key

problem in this strategy is how to control the bioactivity of immobilized FN. In this

study, we aimed to study the bioactivity of FN after different immobilization

pathways: conjugation and adsorption. The chemical and physical properties of the

immobilized surfaces were studied by Micro-BCA, surface contact angle with water

and AFM. The biological properties of the immobilized surfaces were evaluated by

fibroblast adhesion assay and ELISA test for active RGD domain.

4

CHAPTER 2

LITERATURE SURVEY

2.1 Review of HSC and its clinic application

2.1.1 Hematopoiesis and HSC

Blood cells are responsible for constant maintenance and immune protection

of every cell type of the body, which can be classified into two main classes,

lymphoid cells (T, B, and natural killer cells) and myeloid cells (granulocytes,

monocytes, erythrocytes, and megakaryocytes). All blood cells have a limited lifespan:

several hours for granulocytes, several weeks for red blood cells, and up to several

years for memory T cells. Thus, a vast amount of blood cells must be permanently

produced. In normal adults, the body produces about 2.5 billion red blood cells, 2.5

billion platelets, and 10 billion granulocytes per kilogram of body weight per day

[Williams et al (1990)]. Hematopoiesis: the regulated production of eight different

blood cells is a continual process that is the result of proliferation and differentiation

of a small common pool of pluripotent cells called HSC, committed progenitor cells,

and differentiated cells. Within these three stages, extensive expansion of cell

numbers occurs through cell division. A single stem cell has been proposed to be

capable of more than 50 cell divisions or doublings and has the capacity to generate

up to 1015 cells, or sufficient cells for up to 60 years [Kay (1965)].

Alexander Maximow, working in St. Petersburg as a military doctor in 1909,

was the first to suggest that there is a HSC with the morphological appearance of a

‘lymphocyte’ capable of migrating through the blood to micro-ecological niches that

would allow them to proliferate and differentiate along lineage specific pathways

[Maximow (1909]]. The first evidence and definition of blood-forming stem cells

5

came from studies of people exposed to lethal doses of radiation in 1945. Basic

research soon followed. After duplicating radiation sickness in mice, scientists found

they could rescue the mice from death with bone marrow transplants from healthy

donor animals. In the early 1960s, Till and McCulloch began analyzing the bone

marrow to find out which components were responsible for regenerating blood [Till

and McCullough (1961)]. They defined what the two hallmarks of an HSC remain: it

can renew itself and it can produce cells that give rise to all the different types of

blood cells. For these characters, HSC has huge application in clinics.

Figure 1 Hematopoietic system hierarchy. Dividing pluripotent stem cells (LTR and STR) may undergo self-renewal to form daughter cells without loss of potential, and also experienced a concomitant differentiation to form oligopotent daughter cells (CLP, CMP, and CMP lead to GMP and MkEP) with more restricted potential. Continuous proliferation and differentiation result in the production of many mature cells. Macrophages are obtained from monocytes and similarly platelets are derived from megakaryocytes.

2.1.2 HSC clinic application

A: Leukemia and Lymphoma

6

Among the first clinical uses of HSC were the treatment of cancers of the

blood—leukemia and lymphoma, which result from the uncontrolled proliferation of

white blood cells. In these applications, the patient’s own cancerous hematopoietic

cells were destroyed via radiation or chemotherapy, and then replaced with a bone

marrow transplant, or with a transplant of HSC collected from the peripheral

circulation or the CB of matched donors.

B: Inherited Blood Disorders

Another use of allogeneic bone marrow transplants is in the treatment of

hereditary blood disorders, such as different types of inherited anemia (failure to

produce blood cells), and inborn errors of metabolism (genetic disorders characterized

by defects in key enzymes needed to produce essential body components or degrade

chemical byproducts). The blood disorders include aplastic anemia, beta-thalassemia,

Blackfan-Diamond syndrome, globoid cell leukodystrophy, sickle-cell anemia, severe

combined immunodeficiency, X-linked lympho proliferative syndrome, and Wiskott-

Aldrich syndrome.

C: Cancer Chemotherapy

Chemotherapy aimed at rapidly dividing cancer cells inevitably hits another

target—rapidly dividing hematopoietic cells. Doctors may give cancer patients an

autologous stem cell transplant to replace the cells destroyed by chemotherapy. They

do this by mobilizing HSC and collecting them from peripheral blood. The cells are

stored while the patient undergoes intensive chemotherapy or radiotherapy to destroy

the cancer cells. Once the drugs have washed out of a patient’s body, the patient

receives a transfusion of his or her stored HSC.

D: Other Applications

7

For example: One of the most exciting new uses of HSC transplantation puts

the cells to work attacking otherwise untreatable tumors.

Most of the therapies above require the use of allogeneic HSC Transplantation,

as the patients’ own HSC were damaged or dysfunctional. However, there was

significant risk of patient death soon after the transplantation either from infection or

from GVHD. Clinics usually try to find a matched donor who is typically a sister or

brother of the patient who has inherited similar HLA on the surface of their cells to

reduce the risk. In a study where all patients received an HLA-matched transplant

from a sibling, and the results were controlled for age, the relative risk of CB versus

bone marrow was 0.41 for acute GVHD and 0.35 for chronic GVHD[Kondo et al,

2003].

2.1.3 Different sources of HSC in transplantation

A: BM

The classic source of HSC is bone marrow. For more than 40 years, doctors

performed bone marrow transplants by anesthetizing the stem cell donor, puncturing a

bone—typically ilium or iliac bone of pelvis —and drawing out the bone marrow

cells with a syringe. About 1 in every 100,000 cells in the marrow is a long-term,

blood-forming stem cell; other cells present include stroma cells, blood progenitor

cells, and mature and maturing white and red blood cells.

B: PB

It has been known for decades that a small number of stem and progenitor

cells circulate in the bloodstream, but in the past 10 years, researchers have found that

they can coax the cells to migrate from marrow to blood in greater numbers by

injecting the donor with a cytokine, such as GCSF. The donor is injected with GCSF a

8

few days before the cell harvest. To collect the cells, doctors insert an intravenous

tube into the donor’s vein and pass his blood through a filtering system that pulls out

CD34+ white blood cells and returns the red blood cells to the donor. For clinical

transplantation of human HSC, doctors now prefer to harvest donor cells from

peripheral, circulating blood which is easier on the donor—with minimal pain, no

anesthesia, and no hospital stay. The peripherally harvested cells contain twice as

many HSC as stem cells taken from bone marrow and engraft more quickly. This

means patients may recover white blood cells, platelets, and their immune and clotting

protection several days faster than they would with a bone marrow graft.

C: CB

In the late 1980s and early 1990s, physicians began to recognize that blood

from the human umbilical cord and placenta was a rich source of HSC. This tissue

supports the developing fetus during pregnancy, is delivered along with the baby, and,

is usually discarded. Since the first successful umbilical CB transplants in children

with Fanconi anemia, the collection and therapeutic use of these cells has grown

quickly.

Scientists in the laboratory and clinic are beginning to measure the differences

among HSC from different sources. In general, they find that HSC taken from tissues

at earlier developmental stages like CB may hold an advantage in telomere length for

a greater ability to self-replicate, and are less likely to be rejected by the immune

system—making them potentially more useful for therapeutic transplantation. The

accepted explanation is that CB carries much less GVHD than bone marrow because

the newborn baby is "immunologically immature" – for example, the immune system

has not had time to be exposed to various foreign bodies and develop reactions against

them [Rocha (2000)].

9

2.2 Review of HSC expansion

There are three major aspects of stem cell expansion that impact clinical

feasibility.

• First is the ability to isolate and characterize an optimal candidate cell

population for expansion.

• Second is to define the desired characteristics of the population to be used for

engraftment including ways to measure those characteristics.

• Third is the development of reagents and reproducible procedures to support

the culture of a highly defined cell product for transplantation.

2.2.1 Selection of candidate cells for expansion

There are a number of issues related to the selection of a candidate stem

population for ex vivo expansion. Before determining if a protocol results in the

expansion of a stem cell, one must define what a stem cell is. Based on transplantation

of marked stem cells in murine model system, a “stem cell” is defined as a cell with

multi-lineage in vivo hematopoietic reconstituting potential, but has not yet been

proven in humans. Instead, monoclonal antibodies that react with cell-surface proteins

on hematopoietic cells have been used to define and isolate populations of candidate

stem cells phenotypically for further analysis. It is reasonable, though, to assume that

under ideal conditions one would try to expand the most primitive population that one

could identify. However, because of the close timing between preparative regimen

and re-infusion, the standard long-term biological assays for stem cells are impractical

for quantitating the level of expansion. Therefore, the only practical criteria for

infusion would likely be based on phenotypic analysis of the expanded product with

retrospective determination of hematopoietic potential ex vivo. Consequently, it is

10

important to choose a population for expansion that displays a strong correlation

between phenotype and functional activity.

It is widely held that the cell-surface glycoprotein CD34 is expressed on

almost all cells with discernable hematopoietic potential [Krause et al (1996)], as

determined by ex vivo models of progenitor cell proliferation such as CFC and

LTCIC assays [Sutherland et al (1990)]. In more recent years, though, additional

markers have been used to fractionate the CD34+ cells further into lineage-committed

progenitors and more primitive “stem” cells [Terstappen et al (1991)]. Data from

evaluation of sub fractions of CD34+ cells indicated that those cells expressing a

CD34+/CD38- phenotype are highly enriched for very primitive hematopoietic cells

[Baum et al (1992)]. In general, there is a strong correlation between these cell-

surface phenotypes and biological activity in primary cells isolated from different

tissue sources (fetal liver and BM, CB, PB).

2.2.2 Evaluation of ex vivo expansion

Ultimately, preservation of cell function is the determining criteria in the

success of ex vivo human HSC culture systems. Cell function can be queried ex vivo

using the CFC and LTC-IC assays, techniques that detect the presence of committed

and multi potent cells on the basis of the formation of morphologically distinguishable

colonies either directly in methylcellulose cultures (CFC), or after greater than 5

weeks on stroma feeder layers (LTC-IC) [Hogge et al (1996)]. In vivo functional

assays for HSC have been developed on the basis of the ability of these cells to

reconstitute hematopoiesis in the BM of SCID/NOD mice following intravenous

injection [Vormoor et al (1994)]. These so-called NOD/SCID-repopulating cells are

considered human HSC that home to and engraft the murine bone marrow, where they

11

subsequently proliferate and differentiate into multiple lineages [Cashman et al

(1997)].

2.2.3 Expansion systems

HSC must maintain a balance between self-renewing cell divisions (needed to

ensure their existence throughout the life of an organism) and differentiation (needed

to replace the loss of mature cell populations).The successful maintenance and

expansion of transplantable HSC ex vivo requires triggering stem cells to proliferate

without undergoing differentiation. Cultured HSC must additionally avoid apoptosis

and maintain the ability to home, engraft, and differentiate into appropriate receptive

tissues. Because the regulation of these processes involves interactions between cell-

surface receptors and a large group of soluble and matrix bound modulators of cell

function, many investigators have focused on developing culture systems

supplemented with stroma, appropriate combinations of stimulatory factors to

maintain and expand HSC ex vivo. Primarily the ex vivo expansion processes can be

divided into two groups: stroma dependent and stroma independent.

2.2.3.1 Stroma dependent expansion

Before the development of recombinant human cytokines, in the traditional

cultivation process, a stroma layer was known to produce ECM components and

growth factors to support PHPC expansion. The first successful hematopoietic culture

was performed by Dexter’s group, who used murine BM to develop a confluent

stroma layer [Dexter et al (1973)]. The addition of hydrocortisone to the medium

allowed the development of human BM stroma layers, which were found to support

HSC expansion from a variety of sources. [Quesenberry et al (1991)]. A second

12

source of BM or CB cells could be used to initiate this two-step stroma culture.

Gartner [Gartner and Kaplan (1980)] first identified conditions for the simultaneous

development of a stroma layer and stroma dependent hematopoiesis, a system which

is now termed one-step stroma culture. Both types of stroma cultures support HSC

expansion for a prolonged time period, but after approximately 6-8 weeks, the stroma

layer begins to lose integrity and hematopoiesis stopped. Recently the use of FGF-4

has been found to enable faster development and longer maintenance of healthy

stroma layer [Quito et al (1996)].

The finding that more frequent medium exchange increase the productivity of

stroma cultures [Schwartz et al (1991)] led to the development of perfusion bioreactor

systems. The first two perfusion systems consisted of flat bed chambers containing

stroma co-cultures that were perfused with media. They successfully increased the

expansion of total cells, CFC, and LTC-IC compared with static controls [Koller et al

(1993)]. A number of other reactor systems have been used with less success.

Sardonini and Wu investigated the use of stirred micro carrier, airlift and hollow fiber

systems for the culture of BM MNC and CD34+ cells, in all cases they found poor

expansion compared with static cultures [Sardonini and Wu (1993)]. An airlift

packed-bed system was used for murine BM cultures, but this required the use of

exogenous stroma-conditioned medium, and the cells were difficult to harvest from

the packed bed [Highfill et al (1996)]. A micro porous collagen-sphere perfusion

system has been developed for BM cultures, but an unidentified factor has been

shown inhibit the production of CFU-GM colonies [Wang et al (1995)].

Though use of stroma cells in ex vivo culture is advantageous for maturation

and attainment of functional properties in progenitor cells, stroma may pose several

disadvantages in clinical applications: (1) the use of allogeneic source of stroma cells

13

could be potentially harmful due to additional loading of miss-matched tissue, for

example GVHD and donor-mediated infection, (2) ex vivo expansion of PB/CB stem

cells for autologous transplantation requires an additional invasive harvest of patient

bone marrow to produce a preformed stroma layer, and (3) due to the adherent nature

of progenitor stem cells harvesting of cells may require harsh treatment that could be

detrimental to the desired cells.

2.2.3.2 Stroma independent expansion

With the advent of recombinant cytokines and CD34+ selection technologies,

it became possible to develop stroma-free culture systems with significant expansion

of hematopoietic cells. Most of these cultures have been performed in well-plates and

T-flasks utilizing CD34+ cells. For the scale-up of these cultures for clinical trials,

most investigators have chosen to use large culture bags, in which cells grow

undisturbed until harvest. It has been demonstrated that CD34+ cells or MNC cultured

in these environments produce qualitatively indistinguishable cells [Koller et al

(1995)].

A modification of a flat-bed culture chamber has allowed the development of

stroma-free hematopoietic perfusion culture [sandstorm et al (1996)]. The chamber

contains multiple microgrooves (perpendicular to the direction of the flow of medium)

that allow the diffusion of nutrition but retain cells in the chamber. Considerable

success has also been demonstrated in a stirred spinner flask culture at low agitation

rates using a high seeding density of BM MNC. Although there is no stroma layer in

this stirred system, it was demonstrated that the culture maintained and expanded

stroma cells precursors in suspension along with hematopoietic progenitors. Stirred

14

suspension systems have also been successfully used for both serum containing and

serum free culture of PB cells and CB cells.

Despite many of the positive effects of stroma cells can be partly substituted

by either stroma conditioned medium or recombinant growth factors, in the absence

of stroma support, the expansion of CB based on HGF-supplemented culture has been

suboptimal. Phase I clinical trials conducted on UCB expanded in cytokine-

supplemented culture have failed to demonstrate more rapid hematopoietic recovery

in CB recipients [Pecora et al (2000); Kogler et al (1999); Koller et al (1998)]. This

suggests a loss of stem cell function in the cytokine-based expansion in spite of an

increase in CD34+ cell dose. These observations corroborate recent advances in the

cellular and molecular mechanisms underlying the regulation of stem cell

differentiation, indicating that microenvironment or stem cell “niche” is crucial for

maintenance of self-renewal capacity of stem cells [Gupta et al (2000); Kiger et al

(2000); Tran et al (2000)].

2.3 Review of HSC microenvironment

In mammals, hematopoiesis occurs in the extra vascular spaces between

marrow sinuses, in a specialized area, known as niche. The stem cells niche is yet to

be described in the ultra structure level, but it is believed that niches provide

microenvironment to meet certain functional requirements of hematopoiesis. The

microenvironment is characterized by the local geometry and cellular arrangement, by

stroma cells, by the chemical nature of the microenvironment in terms of cytokines

and ECM components.

2.3.1 Cytokine microenvironment

15

The cytokine microenvironment in these cultures is dynamic [Zandstra et al

(1997)], with multiple cell types competing for cytokines and nutrients, many of

which are capable of influencing stem cell fate directly or indirectly. These complex

interactions make the cytokine composition of HSC expansion medium particularly

challenging to optimize. Furthermore, as culture time progresses, the composition of

the culture system can change dramatically and the endogenous production of factors

that have a suppressing effect on HSC self-renewal may significantly influence

culture output. Designing successful HSC expansion cultures thus requires strategies

to maintain an appropriate balance between stimulatory and inhibitory modulators of

HSC function.

Positive modulators of HSC function

Among the different stimulatory cytokines identified in the above mentioned

and other studies, combinations of SCF, FL, and TPO have been shown to be

particularly effective in eliciting self-renewal divisions of human HSC.

SCF, the ligand for the tyrosine kinase receptor c-Kit, promotes the survival

and growth of primitive hematopoietic progenitors when used in combination with

other stimulatory cytokines [Broudy (1997)]; little expansion is observed in cultures

supplemented with SCF alone [Ohmizono et al (1997)].

FL, a factor secreted by stroma cells and which has documented similarities to

SCF, has also been extensively investigated as a potential mediator of HSC expansion

and maintenance [Lyman and Jacobsen (1998)]. FL is believed to act by recruiting

HSC into the cell cycle [Haylock et al (1997)] and by promoting survival,

presumably through inhibition of apoptosis [Meyer and Drexler (1999)].

TPO supports the proliferation and long term maintenance of primitive

hematopoietic progenitors and repopulating HSC when used in combination with

16

other stimulatory cytokines [Won et al (2000)]. When used alone, TPO has little

effect on the expansion of HSC in suspension cultures. However, in the presence of

stroma cells, TPO may promote sustained HSC expansion. Thus, TPO may play a

key role in the long-term maintenance and development of primitive HSC in vivo, and

may be a useful additive for successful HSC expansion ex vivo [Luens et al (1998)].

Although most studies have focused on “hematopoietic” cytokines, other

groups have identified novel factors, not originally identified as a result of their

effects on hematopoietic cells that may regulate HSC responses. For example, Bhatia

et al. [Bhatia et al (1999)] reported that BMP can modulate the proliferation and

differentiation of primitive human hematopoietic cells and that BMP type-1 receptors

were expressed, along with downstream transducers of the BMP signaling pathway, in

subpopulations of hematopoietic progenitors. Recently, the addition of the Notch

ligand, Jagged-1, to cultures of primitive progenitor cells induced the survival and

expansion of repopulating HSC [Karanu et al (2000)] implicating Jagged-1 as another

novel growth factor for the modulation of human HSC function [Varnum et al (2000)].

Negative modulators of HSC function

In spite of extensive investigations, ex vivo expansion systems supplemented

with combinations of stimulatory cytokines have not, to date, yielded sustained HSC

growth, suggesting that HSC expansion may be limited by other factors. Ex vivo

production of inhibitory proteins is one potential reason for the transient expansion

observed in many cultures. Protein factors, such as TGF-b, TNF-a, and, more recently,

IL-3 have been shown to decrease repopulating potential by inducing differentiation

or apoptosis, as well as by decreasing the ability of repopulating stem cells to migrate

to the bone marrow microenvironment. For these inhibitory cytokines to affect HSC

growth ex vivo, they must be generated in culture. Evidence showing that this occurs

17

has been obtained from the analysis of hematopoietic progenitor cultures, where both

mRNA and protein expression of inhibitory and stimulatory factors have been

measured from stroma and progenitor cell populations [Majka et al (2001)].

2.3.2 Stroma microenvironment

The stroma cells include endothelial cell that line the marrow sinuses,

adventitial reticular cells, macrophages, and adipocytes. These stroma cells synthesize

growth and differentiating factors, and ECM components. The stroma cells and the

ECM form 3D scaffolding upon which the HSC lodge. The stroma cells through their

intimate physical contacts with the hematopoietic cells, through the ECM components

and the growth factors they secrete create an intricate hematopoietic inductive

microenvironment, which promotes and regulates hematopoiesis [Dexter (1989)]. It is

this type of inductive microenvironment is termed hematopoietic niche.

Endothelial cells have been observed to produce constitutively G-CSF and

GM-CSF [Brockbank et al (1986)]. They are also capable of transporting plasma

derived substances through their cytoplasm into the hematopoietic space. Adventitial

reticular cells are fibroblastoid cells that ectopically synthesize a number of growth

factors, e.g., G-CSF, M-CSF, GM-CSF, IL-1, IL-6, SCF, FL, LIF [Bianco et al

(2001)].Adventitial reticular cells are capable of taking up fat in the presence of

corticosteroids and converted into adipocyte [Greenberger (1978)]. In young age

marrow is red in color and cellular, with the age hematopoietic activity is reduced and

the marrow is composed of large proportion of fat cells (yellow marrow) [Tavassoli et

al (1974)]. Macrophages are critical in controlling erythropoiesis, which proceeds in

isolated islets surrounding an associated macrophage nurse cell in the BM [Crocker

and Gordon (1987)].Macrophages are considered to be a major source of cytokines

18

under normal conditions; these are, GM-CSF, IL-1 EPO [Rich (1988)]. They also

produce hematopoiesis inhibitory cytokine like, TNFa and IFNg.

2.3.3 ECM microenvironment

In BM, ECM components are mainly produced by endothelial and adventitial

reticular cells. The ECM of BM consists of collagen types I, II, IV [Zuckerman and

Wicha (1983)], laminin, FN [Bentley and Tralka (1983)], vitronectin [Coulombel et al

(1988)], thrombospondin [Long and Dixit (1990)], hemonectin [Campbell et al

(1987)], and proteoglycans with glucosaminoglycan side chains, such as heparan

sulfate and hyaluronic acid [Gordon et al (1988)]. ECM provides support and

cohesiveness for the marrow structure. There is a growing body of evidence

indicating that ECM is important for the regulation of hematopoiesis. For example

glucosaminoglycan side chains of heparan sulfate can sequester and compartmentalize

certain cytokines in local areas and present them to HSC [Roberts et al (1988)]. This

is the mechanism for presenting high, localized concentrations of growth factors that

are protected from proteolysis. Another important function of ECM is to provide

anchorage for immature hematopoietic cells.

2.3.4 3D topology

The successful reconstitution of long-term ex vivo hematopoiesis can perhaps

be best simulated by mimicking in vivo conditions. As mentioned earlier, the

hematopoietic microenvironment is composed of different stroma cells that secrete

ECM components and various soluble growth factors distributed in three-dimensional

space. Thus it was hypothesized that growth of stem cells on a three-dimensional

culture system will be a close replica of hematopoietic microenvironment.

19

Following the concept of the three-dimensional culture system, in 1998,

Mantalaris et al. reported culture of human bone marrow cells on porous collagen

sphere in a packed-bed bioreactor [Mantalaris et al (1998)]. Mononuclear cells of

healthy adult donors were cultured on collagen macro porous spheres in the presence

of SCF, IL-3, GM-CSF, IGF-I, and EPO for four weeks. A few significant

observations extracted from this report are: (a) microscopic examination of the thin

section reveals that the marrow cells populated the pores of the micro spheres in a 3D

fashion with cell-to-cell contact and the resulting interactions resembled the bone

marrow tissue structure in vivo; (b) morphological and staining techniques ensured

the presence of erythroblastic islands of matured red cells and monocytes; (c) no

adipocytes and blanket cells were observed in this culture. In this culture system, cells

of all lineages were present in significant levels, as compared to the 2D Dexter’s

culture.

In the recent past, two different three-dimensional culture systems were

reported for hematopoietic stem cells. The first was tantalum coated porous carbon

matrix, known as Cellfoam [Banu et al (2001)], and the other was non woven PET

fabric [Li et al (2001)]. It was shown that low concentrations of early acting cytokines

SCF and FL result in higher yields of cells that were enriched with primitive

progenitor and multipotent hematopoietic progenitor cells in a 3D Cellfoam bioreactor

as compared to plastic culture flasks. Similarly, 3D non woven PET matrix was

reported to produce two- to three fold more CD34+ cell when compared with the 2D

system in seven to nine weeks culture in the presence of TPO and FL. It was also

shown that both stroma and hematopoietic cells were distributed spatially within the

scaffold and facilitated cell-cell and cell-matrix interactions closure to the in vivo

bone marrow microenvironment. Despite the feasibility of culturing progenitor cells

20

on 3D supports, this system was limited by the problems such as insufficient

expansion of mononuclear cells and difficulties in harvesting cells from the scaffold

matrix.

2.4 Protein immobilization and bioactivity study

In the past twenty years different strategies were utilized to immobilize

functional protein like FN onto varied biomaterials surface. Adsorption was the most

commonly adopted method. Although less stable and less controllable, adsorption is

partially reversible leading to a relatively slow change of conformation of the

adsorbed molecules and a slow release of adsorbed molecules through exchange

reactions. The pioneering work of Grinnell and Feld in the study of adsorption

characteristics of plasma FN reported the higher adsorption and lower bioactivity of

FN on the hydrophobic substrate than on the hydrophilic surfaces [Grinnell and Feld

(1982)]. And it was confirmed by the following studies [George et al (2004);

Kowalczynska et al (2005)].

Recently there has been interest to immobilize FN to surfaces using covalent

coupling chemistries via the amine, thiol, and aldehyde groups [Staffan et al (1997);

Biran et al (2001)]. Covalent coupling presumably would be more efficient and stable

against detachment. A critical issue of the covalent strategy is the conservation of the

desired bioactivity of the immobilized FN. The degree of activity depends on the

binding procedure and the substrate. A combination of conjugation and adsorption can

sometimes produce the optimal results. By conjugating FN to the terminals of

PluronicTM F108 adsorbed on polystyrene, Biran et al. concluded that the FN was

more bioactive than the FN adsorbed directly onto polystyrene as revealed by the

neurite adhesion and outgrowth assay [Biran et al (2001)].

21

However, comparing the adsorption method with the covalent conjugation

approach in the literature is difficult because of the different chemistries and surfaces

used. In addition, the presence of serum and trypsin in the cell adhesion study, which

may alter the composition of the immobilized protein [Andrade and Hlady (1986)] or

degrade the cells surface integrins, can also confound the comparison.

22

CHAPTER 3

MATERIALS AND METHODS

3.1 Materials

3.1.1 Biological materials

CB CD34+ cells were purchased from AllCell Inc. (San Mateo, CA, USA) and

thawed according to reference [Rubinstein et al (1995)]. The purity of CD34+ cell was

91% with a viability of 95%. Recombinant human SCF, FL-3, TPO and IL-3, the

media used in hematopoietic stem cell expansion (StemSpan SFEM Medium) and

CFC assay (MethoCult GF+ H4435) were all purchased from StemCell Technologies

(Vancouver, BC, Canada). Purified human bone marrow MSC and MSC Growth

Medium - MSCGM PoieticsTM were purchased from Cambrex Bio Science

(Walkersville, MD, USA). BHK21 cell line was obtained from Cambrex Walkersville

Inc. (Maryland, USA). RPMI 1640 medium, FBS, penicillin and streptomycin were

purchased from Sigma Aldrich (St. Louis, USA).

3.1.2 Chemical materials

Bi-axially oriented PET films about 100um thickness were purchased from

Goodfellow Inc. (Cambridge, UK). Sterilized SEFAR PETEX 07-1/2 PET woven

mesh was purchased from Sefar Ltd. (Singapore) or Non woven PET Fibra-Cel® was

purchased from New Brunswick Scientific Co. Inc. Human plasma lyophilizated and

sterile FN, MTT were purchased from Roche Ltd. (Indiana, USA). Ethylenediamine,

Glutaradehyde, Ethanolamine, cyanoborohydride, were obtained from Sigma-Aldrich

Co. (St. Louis, USA). Micro BCA, BCA, Goat anti-rabbit IgG peroxidase conjugated

were obtained from PIERCE Biotechnology Inc. (Rockford, USA). 24/96 wells tissue

23

culture plate was from NUNC. Anti-VCIP, RGD domain, Human (rabbit) was

purchased from Oncogene Science Inc. (Cambridge, USA). Monoclonal anti-collagen

type I and IV, monoclonal anti-laminin, monoclonal anti-FN, and secondary antibody

were purchased from Sigma (Saint Louis, Missouri USA). CellTracker Green

CMFDA was purchased from Molecular Probes, Inc (Eugene, OP, USA)

3.2 MSC and CB CD34+ cells co-culture

3.2.1 MSC culture

Cryopreserved cells were thawed and plated in tissue culture flask (Nunc, New

York, USA). Adherent MSCs were maintained in MSCGM, and were harvested when

they reached 80-90% confluence with 1X trypsin/EDTA solution PBS for 5 min, and

then were diluted and inoculated into a fresh tissue culture flask. Cells of passage

around 4 were used for co-culture.

3.2.2 Ex vivo expansion of CB CD34+ cells in co-culture systems

For the 2D (contact) co-culture, MSCs (2.5 x 104 cells) were seeded into each

well of a 24 well plates (Nunc, New York, USA) and grown to confluence. After

washing 3 times with PBS, 600 CB CD34+ enriched cells suspended in 600µl

StemSpan SFM containing 40ug/ml low density lipoprotein (from Academy

Biomedical Company; Houston, USA), 100ng/ml SCF, 100ng/ml FL-3, 50ng/ml TPO,

and 20ng/ml IL-3 were seeded onto the MSC layer in each well. The cells were

cultured for 10 days without medium change at 37oC and 5% CO2. Plain 24-well plate

without the MSC layer was used as a control (2D or TCPS control).

For non-contact co-cultures (Transwell co-culture), MSC feeder layer was

prepared in 24-well plates as described above and incubated in 600ul of supplemented

24

StemSpan medium. A Nunc CC Insert (sterile Polycarbonate membrane, 0.4um pore

sized, pore density 1.5 x 108 pores per cm2, film thickness 20 microns, height above

the base of well 1mm, culturing area 0.5cm2), was placed into each well and seeded

with 600 CB CD34+ enriched cells in an additional 600 ul of supplemented StemSpan

medium. 10 days expansion at 37oC and 5% CO2. Plain 24-well plate with cell insert

was used as a control (Transwell control).

For irradiated MSC co-culture: MSCs were harvested and given irradiation at

15 Gy from a 60Co source. 1 x 105 irradiated cells were seeded into each well of 24-

well plates and formed confluent layer. After 24 hours adhesion, the adherent layers

were washed 3 times with PBS. 600 CB CD34+ enriched cells were added to the

feeder layer, with 600µl supplemented StemSpan medium as described above.

For MSC pellet co-culture: harvested MSCs were incubated in high density at

37ºC over night. After the formation of stable pellets, the MSC pellets were

transferred to the tissue culture plates. After 24 hours adhesion, the adherent pellets

were washed 3 times with PBS. 600 CB CD34+ cells were added to each well of 24-

well plates, suspended in 600µl supplemented StemSpan medium as described above.

At the end of 10-day expansion, cells were harvested by repeated pipetting and

counted using a hemacytometer. Antibodies against CD34, CD45 and other mature

cell surface markers (CD38, Glycophorin A [GlyA], CD15, CD3, CD19 from Becton-

Dickinson and CD41 from Dako) were incubated with cells on ice for 30 min and

analyzed using the FACSCalibur (Becton-Dickinson). At least 20,000 events were

acquired for each analysis. Analysis of flow cytometry data was carried out using

CellQuest Pro. CD34+, CD34+CD38- percentages were calculated as percentages of

blood cells gate (set on CD45+, low side scatter cells of the expanded cells, and

maintained throughout analysis).

25

3.2.3 CFC

After a 10-day culture, 1-3×103 expanded cells or 300-500 unexpanded cells

were harvested and transferred to 1.1 ml H4435 medium and plated 35-mm sterile

Petri dishes (StemCell Technologies). After incubation for 14 days at 37°C in a

humidified atmosphere at 5% CO2, granulocyte-macrophage, erythroid and multi-

lineage colony-forming cells (CFC-GM, CFC-E and CFC-GEMM) were counted

under an inverted microscope.

3.2.4 Animal engraftment

NOD/ SCID mice were purchased from Jackson Laboratories (Animal

Resource Centre in Perth, Australia) and maintained at the animal holding unit of

National University of Singapore. All animals were handled according to Institutional

regulations, under sterile conditions and maintained in micro-isolator cages. Mice to

be transplanted were given total body irradiation of 350 cGy from a 60Co source at

age of 6 to 8 weeks. Within 24 hours, expanded cells were harvested and injected into

the tail vein of the irradiated mice. 2×105 human bone marrow CD34 depleted cells

were irradiated with 1500cGy and injected together as carrier cells.

To assess the engraftment efficiency of the expanded human cells, mice were

sacrificed 6 weeks after transplantation. Bone marrow cells were flushed from both

femurs and tibias with HBSS containing 2% FBS and 5% human serum using a

syringe and a 26-gauge needle. Red blood cells were lysed with 0.16M ammonium

chloride and the remaining cells were washed with HBSS supplemented with 2% FBS.

To facilitate blocking of Fc receptors and prevent nonspecific binding of antibodies,

cells were suspended in HBSS with 2% FBS and 5% human serum and incubated

with mouse 2.4G2 MAb (Becton-Dickinson) for 10 min at 4°C. Cells were then

26

incubated with PerCP-Cy5.5-labeled monoclonal antibodies specific for human CD45

for 30 min at 4°C. Analysis was performed using a FACScalibur and more than

40,000 events were acquired. Successful engraftment of human hematopoietic stem

cell was defined by the presence of at least 0.2% of human CD45+ cells. To verify the

multi-lineage re-constitution of human cells, additional staining was performed with

anti-human CD19-PE /CD41-FITC / CD13-PE / GlyA-PE in combination with anti-

human CD45-PerCP-Cy5.5.

3.3 MSC and CB CD34+ cells 3D co-culture

3.3.1 MSC culture in 3D system and assays

Sterilized SEFAR PETEX 07-1/2 poly PET woven mesh (Sefar Singapore Pte.

Ltd., Singapore) or Non woven PET Fibra-Cel® (New Brunswick Scientific Co. INC)

were used as scaffolds for cell culture. The PET mesh was cut and fitted into the wells

of a 24-well plate. MSCs (2.5 x 104 cells) were seeded into the mesh, grown to

confluence.

SEM:

The cultured samples were fixed with 3% glutaraldehyde in PBS for 24 hours,

washed with PBS, followed by osmification in 1% OsO4 for 30 minutes. After

dehydration in a graded ethanol series, the samples were dried in a critical-point dryer.

After sputter coating with gold, the samples were examined using a SEM (JSM-

5660LV).

Confocol imaging:

For confocal microscopy (Zeiss 510 meta), the cultured samples were first

stained with CMFDA, then fixed with 1% PFA. After that they were stained with

27

antibody for 30 minutes (repeat staining for the use of secondary antibody).After

extensively washing, the samples were examined.

Cell attachment, proliferation and protein production assay for MSC

MSCs (25,000 per well) were seeded into 24-well tissue culture plate with or

without PET mesh. After 24-hr incubation, cells were harvested and counted in

hemacytometer. The result was expressed as attachment percentage of the seeded cells.

Cell proliferation was determined after 4 days of incubation, when the cells reached

~90% confluence. Cell protein production was determined by the BCA™ protein

assay (Pierce, IL, USA). After 4 days incubation, the confluent MSC layer was

washed by PBS and lysed in 300ul of 4% SDS. After centrifugation, 50ul supernatant

was mixed with 200ul BCA working solution and incubated at 37ºC for 30 min. The

total protein level was then measured by UV absorbance at 562nm. The result was

expressed as equivalent albumin production (ug) per 10000 cells.

3.3.2 Ex vivo expansion of CB CD34+ cells in 3D systems and functional assays

MSCs (2.5 x 104 cells) were seeded into the mesh, grown to confluence,

washed 3 times with PBS, and then 600 CD34+ enriched cells were seeded in the

same medium described above. Plain PET woven mesh without MSC layer was used

as a control (3D control). The other functional assays followed the above procedures.

3.4 Study of FN immobilization bioactivity

3.4.1 FN conjugation and adsorption

Figure 2 showed the scheme of FN immobilization. In conjugation pathway,

clean PET surface aminated by ethylenediamine solution (ethylenediamine:

ethanol=1:1) at 50ºC for 40 minutes, was immersed in freshly prepared 2%

28

glutaraldehyde solution with 0.05M sodium cyanoborohydride, reacted for 12-20

hours at room temperature. After washing, the glutaradehyde conjugated surface was

transferred to FN solution in sodium carbonate buffer with 0.05M sodium

cyanoborohydride, reacted for 24 hours at room temperature. To block unreacted

aldehyde sites, 0.06M ethanolamine was added and reacted for another 12 hours at

room temperature. Finally the surface was washed with 5M sodium chloride solution

for 24 hours, and then washed with the desired buffer. In FN adsorption pathway, the

same procedures as above were used, except that no FN was added in the protein

immobilization steps. After surface was blocked by the ethanolamine, FN was added

into the system, reacted for 24 hours, and then washed with 5M sodium chloride for

24 hours, and then washed with desired buffer.

Figure 2 Scheme of FN immobilization on aminated PET surface. (a) FN conjugation pathway (b) FN adsorption pathway

3.4.2 Chemical and physical characterization of the surface

Immobilized FN quantification:

2b) Wash

3b) Block

4b) + FN

5b) Wash

H2C

CH2

CH2

CH2

H2CN

H NH

H2C

CH2

OH

H2C

CH2

CH2

CH2

H2CN

H NH

H2C

CH2

OH

H2C

CH2

CH2

CH2

H2CN

H NH

H2C

CH2

OH

H2C

CH2

CH2

CH2

H2CN

H NH

H2C

CH2

OH

FN

H2C

CH2

CH2

CH2

CH

O

NH2a) Wash

3a) + FN

4a) Block

5a) Wash H2C

CH2

CH2

CH2

H2CN

H NH

H2C

CH2

CH2

CH2

H2CN

H NH

H2C

CH2

OH

H 2C

C H 2 C H 2

C H 2 H2CN

H NH

FN

FN

FN == Protein: FN

NH2HC

CH2

CH2

CH2

CH

OO

H2C

CH2

CH2

CH2

CH

O

NH

1) + Glutaraldehyde (GA)

29

The prepared surfaces were added to micro-BCA working solution (A: B: C:

sodium carbonate buffer=25:24:1: 50), incubated at 37ºC for 2 hours. The total

protein level was then measured by absorbance at 562nm.

Surface contact angle measurements:

Clean PET surface and FN immobilized PET surfaces were measured at room

temperature and 60% relative humidity, using sessile drop method on a telescope

goniometer. More than five measurements were carried out and the resulting values

were averaged.

XPS measurements:

FN immobilized PET surfaces were measured by VG ESCALAB MkII

spectrometer with a Mg Kα X-ray source (1253.6Ev photons) at a constant retard ratio

of 40Ev. The PET films were mounted on the standard sample studs by means of

double-sided adhesive tapes. The core-level signals were obtained at the

photoelectron takeoff angle of 90º. A survey scan spectrum was taken and surface

elemental stoichiometrics were determined from peak area rations.

AFM measurements:

The surface topography of FN conjugated PET, FN adsorbed PET was

examined on a Nanoscope IIIa AFM (Digital Instruments Inc. USA.) in air. Atomic

force microscope images were obtained by scanning surface in the tapping mode. The

driven frequency was 330 ± 50 KHz. The applied voltage was between 3.0 and 4.0 V,

and the drive amplitude was about 300Mv, scanning rate was 1.0 Hz. An arithmetic

mean of the surface roughness profile was determined.

3.4.3 Biological characterization of the surfaces

ELISA

30

FN-immobilized PET surfaces were first captured by primary antibody, rabbit

anti-human VCIP/RGD domain, (1:5000 dilution in BSA solution for 2 hours at 37ºC).

After washing, the surfaces were bound by secondary antibody, goat anti-rabbit IgG

conjugated with peroxidase (1:5000 dilutions in BSA solution for 2 hours at 37ºC).

After washing off the unbound secondary antibody, Slow-TMB-solution was added to

each well incubated for 5-30 min at room temperature in darkness, which was

oxidized by peroxidase, showing the absorbance value at wavelength of 450nm. The

ELISA results for active RGD domains were expressed as equivalent FN according to

the linear FN adsorption isotherm on ethanolamine blocked PET surface as a function

of total FN surface density (by micro-BCA).

In vitro cell culture experiment

The BHK21 cell line was maintained in RPMI 1640 medium supplemented

with 10% fetal calf serum, penicillin (100U/ml), and streptomycin (10mg/ml). The

cell studies were carried below passage 20. Before the cell assay on the surface, the

BHK 21 cells were pretreated with serum-free medium for 2 hours, and then detached

by incubation with non- enzyme cell dissociation solution, in order to avoid the serum

effect and protect cell surface receptor from degradation.

In cell viability assay, cells (seeding density 10,000 per well [96-well plate])

were cultured for 24 hours, and then measured by MTT test. In cell adhesion assay,

cells (seeding density 200,000 per well [24-well plate]) were incubated for 2 hours.

The unattached cells were washed by warmed PBS several times. The adherent cells

were lysed by 4% SDS and centrifuged. The supernatant was subjected to BCA assay.

Based on the assumption that the cells had equal amount of proteins, the total protein

content quantified by BCA was correlated with cell numbers. As the FN surface

density was very low, it would not affect the cell number determination.

31

3.5 Statistical analysis

Data were presented as the mean ± SD. The statistical significance of the data

was determined by student’s t test. The significant level was set at 0.05.

32

CHAPTER 4

MSC AND CB34+ CELLS CO-CULTURE

4.1 Screening of co-culture conditions

The expansion of CB CD34+ cells for clinic application requires serum free

condition. And as the CB cells are non adherent cells, the exchange of medium is not

allowed during the expansion process, which will lead to the loss of cells and the

increase the consumed cost of cytokines. In this project, we chose StemSpan serum

free medium combined with 40ug/ml low density lipoprotein, 100ng/ml SCF,

100ng/ml FL, 50ng/ml TPO, and 20ng/ml IL-3. Then we studied MSC metabolic level

in this serum free medium, and optimized the co-culture conditions.

4.1.1 MSC metabolic level in StemSpan medium

Alamarblue was used as an assay to continuously test MSC metabolic level in

MSCGM and StemSpan serum free medium, which were presented as Alamarblue

reduction percentages. Figure 3 showed that after 10 days incubation in StemSpan

serum free medium, MSC metabolic level (Alamarblue reduction percentages)

decreased from 60.8±0.95% to 55.4±2.81%. It suggested that MSC could survive in

this cord blood expansion condition, and maintain its basic metabolic level. Based on

this result, we concluded that the CB expansion condition was applicable to MSC.

4.1.2 Optimize expansion scale

To optimized the expansion scale of CB CD34+ cells, first of all, we

compared the CB CD34+ cells expansion in different scales: 600 cells expansion in

600ul medium per well of 24-well tissue culture plate and 100 cells expansion in

33

100ul medium per well of 96-well tissue culture plate. Figure 4 showed that the

expansion in 24 well tissue culture plate resulted higher total cells and CD34+ cells

expansion fold ( 342.2±22.3 fold increase of total cells, 9±0.7 fold increase of CD34+

cells) than the expansion in 96 well tissue culture plate (155.2±11.6 fold increase of

total cells, 3.3±0.5 fold increase of CD34+ cells). In the following experiment, we

followed larger expansion scale in 24-well tissue culture plates

0

10

20

30

40

50

60

70

0 3 6 9 12 15

Culturing days

Alam

arbl

ue re

duct

ion

%

Figure 3 MSC metabolic level in MSCGM medium or StemSpan serum free medium with cytokines. 25000 MSCs were seeded into each well of 24-well plate at day 0. After 4 days culture with MSCGM medium, MSCs reached the confluence, subjected to StemSpan serum free medium for another 10 days incubation. Cell metabolic level were evaluated by Alamarblue test at day 4 and day 10, expressed as Alamarblue reduction percentages (n>5).

0

50

100

150

200

250

300

350

400

96 w ell TCPS 24 w ell TCPS

Tota

l cel

ls e

xpan

sion

fold

0

0.5

1

1.5

2

2.5

3

96 w ell TCPS 24 w ell TCPS

CD

34+

cells

per

cent

age

%

0

2

4

6

8

10

12

96 w ell TCPS 24 w ell TCPS

CD

34+

cells

exp

ansi

on fo

ld

Figure 4 CB CD34+ cells expansion in 96-well or 24-well tissue culture plates. 96-well TCPS: each well 100 CB 34+ cells expanded in 100 ul medium. 24-

34

well TCPS: each well 600 CB34+ cells expanded in 600 ul medium. (n=3, p<0.05 for total cells and CD34+ cells expansion fold).

4.1.3 Co-culture over MSC feeder layer

4.1.3.1 Irradiated and non irradiated feeder layer

Usually feeder layers were irradiated before co-culture, in order to suppress

feeder layer’s overgrowth and its competition with expanding cells. Considering the

relatively low metabolic level of MSC in StemSpan medium, we hypothesized that

irradiation was unnecessary in this experiment. We compared CD34+ cells expansion

over the irradiated feeder layer and non irradiated feeder layer. The result (Figure 5)

showed that no difference existed between these two kinds of feeder layers in total

cells expansion fold, the non irradiated feeder layer even lead to higher CD34+

percentages (8.99±1.50 % to 5.97±0.31%) and higher CD34+ cells expansion fold

(47.4±4.8 to 31.9±1.6) than irradiated feeder layer. So MSC feeder layers could be

directly used for co-culture, and supported CB CD34+ cells expansion better than

irradiated feeder layer.

0

100

200

300

400

500

600

Non irradiatedfeeder layer

Irradiatedfeeder layer

Tota

l cel

ls e

xpan

sion

fold

0

2

4

6

8

10

12

Non irradiatedfeeder layer

Irradiatedfeeder layer

CD

34+

cells

per

cent

age

%

0

10

20

30

40

50

60

Non irradiatedfeeder layer

Irradiatedfeeder layer

CD

34+

cells

exp

ansi

on fo

ld

Figure 5 CB CD34+ cells expansion on irradiated or non irradiated MSC feeder

layers. (n=3, p<0.05 for CD34+% and CD34+ cells expansion fold)

35

4.1.3.2 Feeder layer exchanged and non exchanged co-culture

Inspired by the above result that non irradiated MSC feeder layers supported

higher expansion of CB CD34+ cells, we hypothesized that the higher metabolic level

of MSC may support CB expansion better. We designed an experiment that at the 5th

day of co-culture, the culture medium and expanded cells were all transferred to a

freshly prepared MSC feeder layer, then continued the left 5 days of co-culture. The

presence of freshly prepared MSC feeder layer in the mid of co-culture, could

enhanced the average MSC metabolic level. But the expansion result (Figure 6 )was

negative. The exchange of freshly prepared feeder layer lead to the decrease of total

cells expansion fold and CD34+ cells expansion fold. The possible explanation might

be the loss of the cells during transferring and metabolic competition from the freshly

prepared MSC.

0

100

200

300

400

500

600

MSCcoculture

MSC addedcoculture

Tota

l cel

ls e

xpan

sion

fold

0

1

2

3

4

5

6

7

8

9

10

MSC coculture MSC addedcoculture

CD

34+

cells

per

cent

age

%

0

10

20

30

40

50

60

70

MSC coculture MSC addedcoculture

CD

34+

cells

exp

ansi

on fo

ld

Figure 6 CB CD34+ cells expansion with or without exchange of MSC feeder layer in the middle of co-culture. (n=3, p<0.05 for total cells expansion fold and CD34+ cells expansion fold).

4.1.4 Co-culture over MSC pellet

During the high density incubation, MSCs can aggregate, forming stable pellet.

Compared with the monolayer, pellet can accommodate more cells in the same

36

culturing surface. Here I tested if MSC pellets can support CB CD34+ cells expansion

better than MSC feeder layer. The result showed that despite of twice amount of the

MSCs in pellet co-culture, MSC pellet co-culture failed to produce significant

difference compared with MSC feeder layer co-culture. We found that once the pellet

adhered to the surface, MSCs tended to migrate from pellet to surface, forming new

monolayer. This kind of unstable morphology probably affect the expansion result,

which may explained the big variance of the data.

0

100

200

300

400

500

600

MSC feederlayer

MSC pellet

Tota

l cel

ls e

xpan

sion

fold

0

2

4

6

8

10

12

MSC feederlayer

MSC pellet

CD

34+

cells

per

cent

age

%

0

10

20

30

40

50

60

70

80

MSC feederlayer

MSC pellet

CD

34+

cells

exp

ansi

on fo

ld

Figure 7 CB CD34+ cells expansion on MSC feeder layer or adherent MSC pellet. (n=3)

A B

Figure 8 MSC pellet morphology under phase contrast microscope (Magnification: 100X). A: MSC pellet adhered to the surface after 24 hours incubation. B: after another 2 days co-culture, MSC pellet spread to the surrounding surface.

37

Summary: The screening experiments showed that MSC can survive in the

serum free StemSpan medium supplemented with growth factors and LDL. MSC’s

metabolic level affected the expansion result. Neither too low or too high metabolic

level was favorable for CB CD34+ cells expansion. The aggregated conformation of

MSCs did not show any significant effect to CB expansion. In conclusion, we set the

optimal co-culture condition as non irradiated MSC feeder layers culturing in 24-well

tissue culture plate.

4.2 Study of the contact and non-contact co-culture

Most of the co-culture research focused on identifying MSC secreted factors

in supporting HSC expansion. Here we focused on another aspect of the problem: the

effect of cell-cell contact in co-culture. We adopted a system for non-contact co-

culture (transwell co culture). In that system MSC feeder layers were separated from

the CB cells by cell culture inserts, but the molecular signals and factors still could

communicate through the porous membranes (pore size: 0.4um) of the cell culture

inserts. So the only difference to normal feeder layer co-culture would be the block of

the cell-cell contact.

In this study, 600 CB CD34+ cells (CD34+CD45+%: 91%; CD34+CD38-

CD45+%: 16.4%) were cultured in the presence of the cytokines SCF, FL, TPO and

IL-3 for 10 days either in direct contact with a confluent layer of MSCs or physically

separated from the MSC layer by transwell culture. The expansion result showed the

incorporation of MSC feeder layer in both conditions was beneficial for CD34+ cells

expansion. When blocked the cell-cell contact between CB cells and MSCs, the non-

contact co-culture resulted 316.5±55.1, 21.1±3.7 fold increase in total cells,

CD34+CD45+ cells respectively, as compared to 545.4±82.0, 47.9±5.2 fold increase

38

in contact co-culture system. The significant decrease of total cells expansion fold,

CD34+ cells expansion fold in non-contact system suggested cell-cell contact must

play a crucial role in supporting CB cells expansion. Due to the system error in

analysis CD34+CD38-CD45+ cells (another combination of markers to identify HSC),

no significant difference was observed between the contact and non-contact system.

0

100

200

300

400

500

600

700

Contact Non contact

Tota

l cel

ls fo

ld in

crea

se

With MSC Without MSC

0

20

40

60

80

100

120

Contact Non contact

CD

34+C

D45

+ ce

lls fo

ld in

crea

se With MSC Without MSC

0

100

200

300

400

Contact Non contact

CD

34+C

D38

-CD

45+

cells

fold

incr

ease With MSC Without MSC

Figure 9 CB CD34+ cells expansion in contact co-culture or non-contact co-culture.

(n>10, p<0.05).

A

0

0.5

1

1.5

2

2.5

3

Contact co culture Non contact coculture

CMFD

A +

cells

%

B

0

10

20

30

40

50

60

70

80

90

Contact co culture Non contact coculture

CD34

+ ce

lls %

CMFDA+ CMFDA-

Figure 10 Cell-cell contact study by CMFDA and CD34 expression. A: Percentage of CMFDA stained cells. B: CD34+ expression over CMFDA+ cells and CMFDA- cells in contact co-culture and non-contact co-culture system. (n=4).

Then we tried to quantify this kind of cell-cell contact, and its direct result to

CD34 surface markers expression. We used long term tracing staining- CMFDA

stained MSC feeder layer first, then co-culturing CB cells for 10 days. If there was

39

cell-cell communication between MSCs and CB cells, CMFDA would be transferred

to the interacted CB cells through plasma exchanges. The non-contact co-culture was

used as a control, to test if there was CMFDA diffusion existed between non-contact

cells. The result (Figure 10)showed that about 1. 65±0.55 % expanded cells were

stained with CMFDA in contact co-culture system, while nearly no expanded cells

stained with CMFDA in non-contact system. 63.9±17.4% of the CMFDA+ cells

expressed CD34, which was about 8 times higher than the CMFDA- cells in contact

co-culture. This highly suggested that cell-cell communication existed between MSCs

and CB cells in contact co-culture, and that kind of cell-cell communication helped to

reserve stem cell primitivity.

4. 3 Function assays of contact and non-contact co-culture

4.3.1 Phenotype screening of the expanded cells I

To investigate the differentiation status of the expanded cells, we analyzed the

surface markers expression on the expanded cells by flow cytometry. The

incorporation of MSC in both systems displayed similar phenotypic surface markers

expression profile in increasing CD34+CD45+ cells %, CD38+CD45+ cells %,

CD19+cells % and decreasing Gly-A+ cells % compared to the TCPS control and

transwell control. These results suggest that the MSC incorporated co-cultures

favored the expansion of primitive cells (CD34) and lymphocytes (CD38), in

particular B-lymphoid cells (CD19) and slowed down erythrocyte (Gly-A)

differentiation. On the other hand, when contact co-culture and non-contact co-culture

systems were compared, direct cell-cell contact resulted in the increase of

CD34+CD45+ %, CD45+ cells %, CD38+CD45+ cells %, CD15+CD45+ cells %

and the decrease of Gly-A+ cells %, CD3+ cells %, CD19+ cells %, but there was no

40

significant difference in CD34+CD38-CD45+ cells % and CD41+ cells %. The results

suggested that direct cell-cell contact favored monocytes / granucytes (CD15)

differentiation, slowed down erythrocyte (Gly-A), B-lymphoid cells (CD19), T-

lymphoid cells (CD3) differentiation, and maintained slightly higher percentages of

more primitive cells, compared with non-contact co-culture.

In control experiments, we investigated whether the doubling the volume of

culture medium in the non-contact system could affect the expansion of CB CD34+

cells. No significant differences in terms of cell expansion and cell phenotype were

observed when compared to cultures without MSC (data not shown). This confirmed

that the medium volume was not confounding factor.

Table 1: Surface markers expression of expanded cells I:

Percentage % CD45+ CD34+CD45+ CD34+CD38-CD45+ CD38+CD45+

Contact co-culture 85.87±5.85* 8.22±1.12 3.15±1.73 29.37±6.24*

TCPS control 71.45±8.45 3.42±1.01 2.00±0.75 10.56±2.07

Non-contact co-culture 73.22±4.02 6.14±0.78 2.77±1.98 14.23±2.38

Transwell control 69.05±18.27 2.74±0.67 1.98±0.83 7.83±1.91

Percentage % CD15+CD45+ Gly-A+ CD3+ CD19+ CD41+

Contact co-culture 43.27±8.87* 20.99±5.84* 0.30±0.07* 1.08±0.33* 5.83±1.74

TCPS control 27.36±5.32 30.56±3.25 0.44±0.15 0.41±0.11 4.19±1.48

Non-contact co-culture 25.43±0.23 28.51±0.41 1.06±0.35 3.39±1.64 6.06±2.52

Transwell control 25.74±2.83 44.8±9.91 0.91±0.10 0.40±0.09 2.44±0.50

CD15: Monocytes and Granulocytes; Gly-A: precursors of Erythrocytes, CD3+: T cells, CD19+: B cells, CD41+: Megakaryocytes, the precursor of plateles. (n>6 for the first 4 categories, n>3 for the left categories, *p<0.05 compared to non-contact co-culture)

41

4.3.2 CFC I

The CFC assay evaluates the multipotency of the cells following expansion.

Figure 11 shows the colony counts derived from the expanded cells, expressed as fold

increase over the number of colonies derived from 600 freshly thawed CB CD34+

cells (total CFC colonies: 132.8 ±8.5; CFC-GM: 72±3.2; CFC-E: 20±3.5; CFC-

GEMM: 40.8±8.7). The contact co-culture produced higher fold increase in CFC-GM

colonies and total CFC colonies, which were consistent with the expansion and

phenotype screening result. In terms of CFC profile (Figure 11B) ex vivo expansion

appear to have induced an increase of CFC-E at the expense of CFC-GEMM in

comparison with unexpanded cells. No significant effect on the CFC-GEMM% was

observed.

A0

50

100

150

200

250

CFC-GM CFC-E CFC-GEMM CFC total

CFU

fold

incr

ease

Contact coTCPS controlNon contact coTranswell control

B0

10

20

30

40

50

60

70

CFC-GM CFC-E CFC-GEMM

Perc

enta

ges

*100

%

UnexpandedContact coTCPS controlNon contact coTranswell control

Figure 11 CFC properties of expanded cells I. 600 CB CD34+ enriched cells (CFC-GM: 72±3.17; CFC-E: 20±3.46; CFC-GEMM: 40.8±8.65) were expanded in contact or non-contact system with or without MSC. A: Colonies fold increase. B: Colonies distribution. (Mean + SD; n>4; *p<0.05 compared with 2D co culture)

4.3.3 Animal engraftment I

The Repopulation assay of human hematopoietic stem cell in NOD/SCID

mouse can quantitatively measure the dual ability of stem cells to self-renew and

42

differentiate. From the literature, the lowest number of CD34+ CB cells that can

engraft NOD/ SCID mice is around 2×104 [Holyoake et al (1999)].

Table 2: The engraftment and lineage expression by human cells in the bone marrow of NOD/SCID mice I

Human Cells (%) Mouse injection Positive

Mice * CD45+ CD41+ CD19+CD45+ CD13+CD45+ GlyA+

20000 uncultured CD34+ CB cells 3/3 0.232;

10.094; 1.132 0.723±0.01 3.056±4.36 0.672±0.74 3.476±0.26

600 uncultured CD34+ CB cells 3/3 2.386;

0.248; 0.739 -0.106±0.3 0.532±0.55 0.606±0.83 1.521±1.60

Injected cells were expanded from 600 CD34+ CB cells in the following systems:

2D co-culture 5/5 1.435; 1.570; 1.683; 1.770; 1.461

0.664±0.9 1.193±0.41 1.268±0.35 3.972±1.06

Non-contact co-culture 4/4 1.041; 0.625;

1.246; 0.589 0.193±0.1 0.86±0.24 0.981±0.32 2.245±0.51

Control (Tissue culture plates) 4/7

0.013; 0.603; 0.487; 1.045; 1.379; 0; 0

0.732±0.4 0.667±0.28 1.082±0.70 3.767±0.58

*Positive mice were identified by at least 0.2% of human CD45+ cells of total lysed bone marrow cells. The lineage expression of human megakaryocytes CD41+; human B cells CD19+CD45+; human monocytes and granulocytes CD13+CD45+; human erythrocytes Gly-A were studied by FACS in all the positive mice.

The ex vivo results suggested that the co-culture produced about 550-320 fold

increase of CD34+CD45+ cells, 50-100 fold increase of CD34+CD38-CD45+ cells and

80-90 fold increase of CFC-GEMM colonies. Based on this reasoning, cells expanded

from about 600 initial CD34+ CB cells would be expected to contain enough

progenitors to reconstitute the hematopoiesis of irradiated NOD/SCID mice. As no

significant difference in expansion results was observed among all the non co-culture

systems, we chose cells expanded from the 2D control (TCPS) as a common control

for all the non co-culture systems. In addition, un-expanded CD34+ CB cells at two

different doses (600 and 20,000 cells) were also transplanted for comparison. Table 2

show the engraftment efficiency of the expanded cells as well as their differentiation

43

potential in the positive mice. The contact co-culture system produced an engraftment

percentage above 1%, which were better than non-contact co-culture system and

TCPS control. The engraftment of the unexpanded cells, particularly at low dose, was

unexpected. Nevertheless, there is high variability among animals in these two groups.

The results highlight the fact that ex vivo expansion would impair stem cells

transplantation ability.

4.4 Discussion

Hematopoietic stem cells, defined by their ability to self-renew and to

differentiate into all mature blood cell types, have by far been the most widely applied

stem cells in the clinic for treatment of such diseases as leukemias and other

malignant cancers as well as several non-malignant blood disorders. However, the

clinical applications of HSC are severely hampered by our limited understanding of

the mechanisms that control HSC self-renewal and differentiation and by the inability

to expand HSC in culture. However, over the past 15 years, rapid advances have been

made in the understanding of HSC biology, particularly in the roles of cytokines and

signaling molecules such as Wnt, Notch etc [Zhang et al (2003); Calvi et al (2003)].

These findings have led to the development of stroma-free ex vivo expansion systems,

which are clinically more well-defined and accepted [Lazzari et al (2001)]. Recently

however, interest has been revived in stroma-supported culture systems and various

cell types including several primary cell types [Breems et al (1998)], immortalized

stroma cell lines [Nishioka et al (2003)], or genetically engineered cytokine-secreting

cells [Balduini et al (1998)]. Feeder cultures are able to provide a more

physiologically relevant repertoire of factors that promotes HSC self-renewal and

maintenance.

44

MSC is a non hematopoietic, well-characterized homogeneous population of

adherent skeletal and connective tissue progenitor cells within the bone marrow

stroma [Haynesworth et al (1992); Majumdar et al (1998); Pittenger et al (1999)]. It is

one of the most promising stem cell types due to their availability and the relatively

simple requirements for ex vivo expansion. Importantly, MSC provides a rich

environment of signals, including cytokines, ECM proteins, adhesion molecules, and

cell-cell interactions, controlling the proliferation, survival, and differentiation of

early lympho- hematopoietic stem cells [Haynesworth et al (1996), Majumdar et al

(2000)]. Accordingly, MSC has been shown to be capable of supporting HSC

expansion ex vivo [Kadereit et al (2002); Wang et al (2004); McNiece et al (2004)].

Similar to Dexter-type stroma cells, MSC can also support LTC-IC colonies during

prolonged ex vivo bone marrow culture [Majumdar et al (2000)].

In addition to the abovementioned advantages, MSC, like other stem cells

which are poor antigen presenting cells, are relatively immunologically compatible,

and show positive augmentative role in HSC transplantation. High dose chemotherapy

or total body irradiation often results in the ablation of the bone marrow stroma,

which is critical for the maintenance of hematopoiesis. Previous animal studies

suggest that the transplantation of healthy stroma components including MSC

enhances the ability of the bone marrow micro environment to support hematopoiesis

following transplantation [Devine et al (2001)]. Recent studies have shown that

autologous MSC expanded ex vivo could accelerate hematopoietic recovery after

HSC transplantation [Koc et al (2000); Westerman and Leboulch (1996)]. Allogeneic

MSC expanded in culture has been successfully co-infused with T-cell depleted

peripheral blood hematopoietic progenitors into a haploidentical receipient and is

currently under further clinical evaluation [Lee et al (2002)]. In summary, all these

45

studies have shown that MSC is a good stroma candidate. We therefore incorporated

it into our CB expansion systems.

Most of the studies which focus on the secretion effect of MSC showed that

MSC secretes, at baseline, IL-6, IL-7, IL-8, IL-11, IL-12, IL-14, and IL-15, MCSF, FL,

and SCF, similar to the cytokines and growth factors expressed by marrow derived

stroma cells [Majumdar et al (1998)]. We wondered whether conditioned medium

could replace the total function of MSC ex vivo, or is the cell-cell contact needed. We

adopted the transwell co-culture system, which blocks the cell-cell contact, but still

permits the molecular signal exchange through porous membrane. Our results

indicated that the contact co-culture increased the expansion of total cells (545.4

vs.316.5 fold), CD34+ cells (47.9 vs. 21.1 fold), CD34+CD38- cells (100.4 vs. 52.8

fold) compared with the non-contact co-culture in transwell system. Similarly, the

pluripotency of cells expanded in contact co-culture as evaluated by CFC assay was

also superior compared to those expanded in non-contact co-culture (CFU-GEMM:

84.1 vs. 76.0 fold; total CFU 150.1 vs. 116.5 fold). The animal engraftment assays

also showed that cells expanded in contact co-culture system appeared to be more

competent in reconstructing short term hematopoiesis. The CMFDA staining showed

that the 63.9% of the anchored cells to the stroma layer (CMFDA+), expressed CD34

surface marker, which is about 8 times higher than suspension cells. The findings

suggested that contact co-culture is effective in maintaining stem cell function,

protecting them from differentiation.

46

CHAPTER 5

MSC AND CB CD34+ CELLS 3D CO-CULTURE

5.1 Screening of 3D scaffolds

In this study we incorporated MSC stroma into the 3D system supporting CB

CD34+ cells expansion, in order to more faithfully replicating the in vivo 3D marrow

configuration.

A B

C D

Figure 12 SEM image of PET meshes and co-culture in meshes. A: PET unwoven mesh; B: CB and MSC co-culture over PET unwoven mesh (Day 10 ). C: PET woven mesh; B: CB and MSC co-culture over PET woven mesh (Day 10 ).

First of all we screened two commonly used 3D systems: unwoven PET mesh

and woven PET mesh. Although serum coated unwoven PET mesh achieved good

expansion result in the literature, the MSC co-culture in non woven PET fibrous

matrices induced quite limited expansion of CD34+ cells. It was probably because of

47

the space limit in structure for co-culture (expanded MSC size: about 20*60 um, CB

cells diameter 3-10 um, compared to the controlled pore size 10-60 um in fibrous

matrices), and difficulties in harvesting the entrapped CB expanded cells from the MSC

stroma in the scaffold. For the application purpose we gave up this scaffold, chose

woven PET mesh, which was relatively easy to isolate cells, while still maintaining part

of the 3D structure. The PET woven mesh comprises of fibers (mesh opening 1um, wire

diameter 34 um, thickness 70 um, and 6.29 fold increase of surface area compared to

the 2D surface, detected by micro CT).

0

100

200

300

400

500

600

700

800

900

1000

Wovenmesh

Unwovenmesh

Tota

l cel

ls e

xpan

sion

fold

With MSC

Without MSC

0

2

4

6

8

10

12

14

Wovenmesh

Unwovenmesh

CD34

+ ce

lls p

erce

ntag

e %

With MSC

Without MSC

0

20

40

60

80

100

120

Wovenmesh

Unwovenmesh

CD

34+

cells

exp

ansi

on fo

ld

With MSC

Without MSC

Figure 13 CB CD34+ cells expansion in PET woven or unwoven mesh. (n=4, p<0.05 compare woven mesh co-culture with the other groups).

5.2 Study of 3D and 2D co-culture

In this study, 600 CB CD34+ cells (CD34+CD45+%: 91%; CD34+CD38-

CD45+%: 16.4%) were cultured in the presence of the cytokines SCF, FL, TPO and

IL-3 for 10 days in direct contact with a confluent layer of MSC growing on 2D or 3D

scaffolds. As shown in Figure 14, the expansion of total cells, CD34+CD45+ cells and

CD34+CD38-CD45+ cells in 3D co-culture were all significantly higher than those of

the other groups. Co-culture in 3D configuration resulted in approximately

48

891.2±163.6, 96.0±20.2, 273.2±106.1 fold increase in total cells, CD34+CD45+, and

CD34+CD38-CD45+ cells, respectively, as compared to 545.4±82.0, 47.9±5.2,

100.4±51.7 fold increase in the 2D co-culture system. These results also suggest that

3D co-culture is superior to 2D co-culture in supporting the expansion of CB CD34+

cells.

0

200

400

600

800

1000

2D 3D

Tota

l cel

ls fo

ld in

crea

se

With MSC

Without MSC

0

20

40

60

80

100

120

2D 3D

CD

34+C

D45

+ ce

lls fo

ld in

crea

se

With MSC

Without MSC

0

100

200

300

400

2D 3DC

D34

+CD

38-C

D45

+ ce

lls fo

ld in

crea

se

With MSC

Without MSC

Figure 14 CB CD34+ cells expansion in 2D or 3D systems. (n>10, p<0.05 compare woven mesh co-culture with the other groups).

5.3 Functional assays of 3D and 2D co-culture

5.3.1 Phenotype screening of expanded cells II

To investigate the differentiation status of the expanded cells, we analyzed the

surface markers expression on the expanded cells by flow cytometry. Cells expanded

in the 2D and 3D co-cultures displayed very similar phenotypic surface marker

expression profiles. The notable exceptions are the increase in the CD34+CD38-

CD45+ (5.08% vs. 3.15%; p: 0.019) and CD19+ (2.94% vs. 1.08%; p: 0.041)

populations in the 3D co-culture systems (Table 3). In comparison to the TCPS

controls, the co-cultures in 2D and 3D systems showed increases in CD45+,

CD34+CD45+, CD38+CD45+ and CD19+ cell population but a decrease in Gly-A+

cell population, There is however no significant difference in the CD15+CD45+,

49

CD3+%, and CD41+ populations after expansion. These results suggest that the

contact co-cultures favored the expansion of primitive cells (CD34) and lymphocytes

(CD38), in particular B-lymphoid cells (CD19). It also resulted in a reduction in

erythrocyte (Gly-A) population, and produced no significant effects on the

percentages of monocytes/granulocytes (CD15), T cells (CD3), and megakaryocytes

(CD41).

Table 3 Surface markers expression of expanded cells II

Percentage % CD45+ CD34+CD45+ CD34+CD38-CD45+ CD38+CD45+

2D co-culture 85.87±5.85 8.22±1.12 3.15±1.73 29.37±6.24

TCPS control 71.45±8.45 3.42±1.01 2.00±0.75 10.56±2.07

3D co-culture 85.54±3.77 10.06±2.63 5.08±1.83 26.71±5.39

3D control 61.51±15.44 2.82±0.69 1.80±0.76 8.32±4.32

Percentage % CD15+CD45+ Gly-A+ CD3+ CD19+ CD41+

2D co-culture 43.27±8.87 20.99±5.84 0.30±0.07 1.08±0.33 5.83±1.74

TCPS control 27.36±5.32 30.56±3.25 0.44±0.15 0.41±0.11 4.19±1.48

3D co-culture 37.01±5.68 21.01±2.45 0.99±0.64 2.94±0.87 4.96±1.41

3D control 27.39±6.33 47.77±11.57 0.43±0.10 0.19±0.19 2.34±0.20

CD15: Monocytes and Granulocytes; Gly-A: precursors of Erythrocytes, CD3+: T cells, CD19+: B cells, CD41+: Megakaryocytes, the precursor of platelets. *p<0.05 compared to its internal control: cytokine expansion only. (n>6 for the first 4 categories, n>3 for the left categories)

In control experiments, we investigated whether the presence of 3D PET mesh

alone could affect the expansion of CB CD34+ cells. No significant differences in

terms of cell expansion and cell phenotype were observed when compared to cultures

50

without MSC (data not shown). This confirmed that the PET mesh was not

confounding factor.

5.3.2 CFC II

The CFC assay evaluates the multipotency of the cells following expansion.

Figure 15A shows the colony counts derived from the expanded cells, expressed as

fold increase over the number of colonies derived from 600 freshly thawed CB

CD34+ cells (total CFC colonies: 132.8 ±8.5; CFC-GM: 72±3.2; CFC-E: 20±3.5;

CFC-GEMM: 40.8±8.7). The 3D co-culture produced the highest fold increase across

all CFU types assessed, being statistically significant from the 2D co-culture. The

trend is consistent with the cell expansion result. In terms of CFC profile (Figure 15B),

ex vivo expansion appear to have induced an increase of CFC-E at the expense of

CFC-GEMM in comparison with unexpanded cells. No significant effect on the CFC-

GM% was observed. The 3D co-culture showed a higher CFC-GEMM% than the 2D

co-culture (24.56±1.38% vs. 17.13±2.69%; p: 0.0012).

A0

50

100

150

200

250

300

350

400

CFC-GM CFC-E CFC-GEMM CFC total

CFU

fold

incr

ease

2D co 2D 3D co 3D

B0

10

20

30

40

50

60

70

CFC-GM CFC-E CFC-GEMM

Perc

enta

ges

*100

%

Unexpanded 2D co 2D 3D co 3D

Figure 15 CFC properties of expanded cells II. 600 CB CD34+ enriched cells (CFC-GM: 72±3.17; CFC-E: 20±3.46; CFC-GEMM: 40.8±8.65) were expanded in 2D or 3D system with or without MSC. A: Colonies fold increase. B: Colonies distribution. (Mean + SD; n>4; *p<0.05 compared with 2D co culture)

51

5.3.3 Animal engraftment II

The repopulation assay of human hematopoietic stem cell in NOD/SCID

mouse can quantitatively measure the dual ability of stem cell to self-renew and

differentiate. From the literature, the lowest number of CD34+ CBcell that can engraft

NOD/ SCID mice is around 2×104 [Holyoake et al (1999)]. The ex vivo results

suggested that the co-culture produced about 50-100 fold increase of CD34+CD45+

cells, 100-270 fold increase of CD34+CD38-CD45+ cells and 80-180 fold increase of

CFC-GEMM colonies. Based on this reasoning, cells expanded from about 600 initial

HSC would be expected to contain enough progenitors to reconstitute the

hematopoiesis of irradiated NOD/SCID mice. As no significant difference in

expansion results was observed among all the non co-culture systems, we chose cells

expanded from the 2D control (TCPS) as a common control for all the non co-culture

systems. In addition, un-expanded CD34+ CB cells at two different doses (600 and

20,000 cells) were also transplanted for comparison.

Table 4 showed the engraftment efficiency of the expanded cells as well as

their differentiation potential in the positive mice. The 3D co-culture system

consistently generated engraftment of human CD45+ cells at percentages above 1.5%

in all mice analyzed. Similarly, the 2D co-culture system produced an engraftment

percentage above 1%. The difference between these two systems is statistically

significant (p3D co to 2D co=0.008). Cells expanded in 3D co-culture also yielded higher

human B cells (CD19+CD45+) and monocytes and granunolocytes (CD13+CD45+)

engraftment percentages (B cells: P3D co to 2D co=0.035; monocytes and granulocytes:

P3D co to 2D co=0.008). This result was consistent with the observed higher CD34+ and

CFC-GEMM expansion results. The engraftment of the unexpanded cells, particularly

at low dose, was unexpected. Nevertheless, there is high variability among animals in

52

these two groups. The results highlight the fact that ex vivo expansion would impair

stem cells transplantation ability.

Table 4 The engraftment and lineage expression by human cells in the bone marrow of NOD/SCID mice II

Human Cells (%) Mouse injection Positive

Mice * CD45+ CD41+ CD19+CD45+ CD13+CD45+ GlyA+

20000 uncultured CD34+ CB cells 3/3 0.232;

10.094; 1.132 0.723±0.01 3.056±4.36 0.672±0.73 3.476±0.26

600 uncultured CD34+ CB cells 3/3 2.386;

0.248; 0.739 -0.106±0.32 0.532±0.55 0.606±0.83 1.521±1.61

Injected cells were expanded from 600 CD34+ CB cells in the following systems:

3D co-culture 5/5 2.470; 2.952; 3.187; 2.370; 1.913

1.77±0.99 2.125±0.68 3.008±0.85 6.178±2.25

2D co-culture 5/5 1.435; 1.570; 1.683; 1.770; 1.461

0.664±0.86 1.193±0.41 1.268±0.35 3.972±1.06

Control (Tissue culture plates) 4/7

0.013; 0.603; 0.487; 1.045; 1.379; 0; 0

0.732±0.43 0.667±0.28 1.082±0.70 3.767±0.58

*Positive mice were identified by at least 0.2% of human CD45+ cells of total lysed bone marrow cells. The lineage expression of human megakaryocytes CD41+; human B cells CD19+CD45+; human monocytes and granulocytes CD13+CD45+; human erythrocytes Gly-A were studied by FACS in all the positive mice. (Mean ± SD; *p<0.05 compared with other expanded cells engraftment).

5.4 3D culture of MSC

In an attempt to understand the observed differences in the supportive role of

MSC in the 2D and 3D co-culture systems, we analyzed the attachment, proliferation

and ECM production of the cells cultured on the PET woven fabric and TCPS. The

result (Figure 16) showed that the 3D PET woven structure was more favorable for

cell attachment, or possibly entrapment (77.4±3.3% vs. 60.8±4.5%). Consequently,

the 3D system maintained more cells in confluence compared with the 2D system

(65,000 ± 5,000 cells per well vs. 52,000 ± 2,000 cells per well at day 4). MSCs

grown in the PET mesh also showed higher protein production compared to those

53

grown on TCPS. (7.3±0.6 vs. 6.2±1.7 ug per 10000 cells; p: 0.0157). Scanning

electron microscopy revealed that the MSCs grown on the PET mesh secreted copious

amount of ECM proteins that were organized into an extensive fibrous network

(Figure 17). In contrast, MSCs grown on TCPS displayed relatively sparse ECM

production. The immuno-staining showed that MSCs exhibited a more elongated

morphology in PET mesh than in TCPS surface. Cultured on TCPS, the MSCs

expressed FN but not collagens I, collagen IV, and laminin. Cultured on the PET

mesh, the MSCs expressed FN strongly and also detectable although low amount of

collagen I.

A

0

20

40

60

80

100

3D 2D

MSC

atta

chm

ent *

100%

B

0

20000

40000

60000

80000

3D 2D

No.

of c

ells

per

wel

l

C

0

2

4

6

8

10

3D 2D

Prot

ein

prod

uctio

n (u

g) p

er 1

0000

cel

ls

Figure 16 MSC attachment, proliferation, and protein production in 2D or 3D systems. A: MSC attachment on 3D PET mesh and 2D TCPS after 24 hours incubation. B: MSC proliferation after 4 days culture on 3D PET mesh and 2D TCPS. C: Protein production per 10,000 cells after 4 days culture on 3D PET mesh and 2D TCPS. (Mean ± SD, n=4; Pattachment: 0.0009; Pcell number:0.002; Pprotein production: 0.0157).

5.5 Discussion

Recently, the function of 3D micro environment in hematopoiesis has attracted

more attention and interests. Quite a few 3D systems were designed to enhance cell–

54

cell and cell–matrix interactions, in an effort to mimic the in vivo 3D bone marrow

structure in which more than 95% of normal hematopoiesis occurs; like collagen

coated nylon mesh [Naughton et al (1987) Naughton et al (1990)], porous collagen

micro spheres [Wang et al (1995); Mantalaris et al (1998)], tantalunm coated cellfoam

[Rosenzweig et al (1997)], serum coated unwoven PET fibrous matrices [Li et al

(2001)] in supporting CB or bone marrow cells expansion.

Figure 17 SEM and Confocal images of MSCs culture and co-culture in 2D or 3D systems. A-B: MSCs grown on 3D and 2D surface at confluence; C: MSCs and CB cells after 10 days co-culture; D-E: MSCs grown on 3D and 2D surface; F-G: Fibronectin expression in 3D and 2D culture; H-I: Collagen I expression in 3D and 2D culture. Cells were stained with CMFDA (green); each kind of ECM proteins was stained with TRITC (red) separately.

In this study, we investigated the effects of a combination of MSC feeder layer

with 3D architecture on CB CD34+ cells expansion. MSCs were cultured on woven

55

PET mesh (mesh opening 1um, wire diameter 34um, thickness 70um, and 6.29 fold

increase of surface area compared to the 2D surface, detected by micro CT). Our

results indicated that co-culture of CB CD34+ cells with MSCs in a 3D setting

produced increased expansion of total cells (891.2- vs 545.4-fold), CD34+ cells (96.0-

vs 47.9-fold) or CD34+CD38- cells (273.2- vs 100.4-fold) compared to control

culture. Similarly, the pluripotency of cells expanded on 3D co-cultures as assessed

by CFU assay was also superior compared to those expanded on 2D coculture (CFU-

GEMM:183.6- vs 84.1-fold; total CFU: 229.9- vs 150.1-fold). Additionally, animal

engraftment assays have shown that cells expanded in the 3D co-culture appear to be

more competent in reconstituting short term hematopoiesis. Cytochemical staining

revealed that culture of MSCs on 3D mesh led to increased proliferation which may

contribute to the increase in beneficial effects on CB expansion. More importantly, we

observed high expression of ECM fibrils on MSCs grown in 3D culture, a feature not

seen for cells grown on 2D culture. This result is consistent with the former findings

[Grayson et al (2004)] which showed that MSCs secreted more ECM protein such as

FN when cultured in 3D non-woven PET matrices. Scanning electron microscopy

reveals a tendency of hematopoietic cells to aggregate in crevasses formed by fibrils

and cell structures. This may provide 3D topological cues necessary to maintain the

primitiveness and pluripotency of the CD34+ cells.

In addition to providing mechanical support , the endogenous bone marrow

furnishes a biochemical environment by secreting a complex meshwork of ECM rich

in proteins such as collagen and elastin, specialized proteins such as FN and laminin,

and proteglycans, It has been reported that ECM glycosaminoglycan side chains can

bind soluble growth factors forming concentration gradients that among other roles

aid in cell anchorage, facilitate receptors dimerization and signal transduction,

56

modulate extra cellular proteolysis and protect signal molecules from being degraded

[Ramirez and Rifkinb (2003) ]. Cell adhesion motifs present in the ECM also play

important roles in the regulation of stem cell biology. FN which is secreted by bone

marrow stroma cells, have been found to have important influence on HSC cell cycle.

Its receptors, integrin α4β1 and α5β1, are highly expressed on HSC. This implied

that besides its traditional functions in HSC lodgment, FN may also take part in the

maintenance of the self-renewal ability of HSC within the hematopoietic niche.

Evidence comes from studies that showed that adhesion to FN during ex vivo culture

preserves the regenerative capacity of human PHPCs/HSCs [Dao et al (1998)]. Taken

together, we postulate that the influence of a 3D microenviroment coupled with

elevated deposition of ECM by MSC create a cellular niche more closely resembling

the natural bone marrow thus favoring CD34+ cell proliferation and maintenance of

pluripotency.

57

CHAPTER 6

STUDY OF FN IMMOBILIZATION BIOACTIVITY

Inspired by the 3D co-culture results that FN was specially secreted by MSCs

in the co-culture system and FN’s potential functions in controlling cells adherence

and cell cycles, we hypothesized that FN may play a very important role in CB’s

expansion. Immobilization of FN onto the surface of culture system may represent a

stroma free expansion strategy. The purpose of this sub project was to evaluate two

common protein immobilization methods, and study the change of FN bioactivity

during the immobilization process. The results of this study would be applied to

construct artificial HSC expansion niche in the future.

6.1 Physical and chemical characterization of conjugated and adsorbed FN

6.1.1 FN immobilization efficiency

The amount of the immobilized FN on modified PET surface was determined

by the micro BCA assay, which depends on the number of peptide bonds and the

presence of four amino acids (cysteine, cystine, tryptophan and tyrosine). Figure 18

shows the surface density of FN as a function of the input concentration in solution.

The conjugation method produced a higher FN content onto the surface than the

adsorption method. With adsorption, the content of FN adsorbed reached a saturation

level around 400 ng/cm2 once the FN input concentration exceeded 4 ug/ml. The

saturation level is consistent with conventional adsorption isotherms, representing

approximately the amount of FN necessary to produce a monolayer coating based on

the dimension of the molecules [Williams et al (1982)]. In contrast, the conjugation

method seemed to be able to form multiple layers of FN despite extensive washing.

At an FN input concentration of 30 ug/ml, 918±134 ng/cm2 of FN could be stably

immobilized onto the surface.

58

6.1.2 XPS assay

XPS instruments measure the number and kinetic energy of emitted

photoelectrons. Because the core electrons of each element have characteristic

binding energies, the peaks in the XPS spectra allow identification of all elements,

except H and He. Among the elements detected, the N1s signal percentage as a

function of the FN surface density was shown for both substrates in Figure 19 and

Table 5. The XPS data showed that FN covalent conjugated surface had higher

nitrogen percentages. This result was consistent with the micro-BCA result, that more

protein was immobilized through covalent conjugation method.

0

200

400

600

800

1000

1200

0 3 6 9 12 15 18 21 24 27 30FN input concentration (ug/ml)

FN d

ensi

ty o

n su

rfac

e ( n

g/cm2 )

ConjugationAbsorption

Figure 18 FN immobilization efficiency. FN surface density (by micro-BCA assay) was immobilization methods (conjugation & adsorption) and protein concentration (0.256ug/ml-30ug/ml) dependent. Mean ± SD was obtained from five separate experiments

A0

10000

20000

30000

40000

50000

02004006008001000

Cou

nts

/ s

Binding Energy (eV)

O

C

B0

5000

10000

15000

20000

25000

02004006008001000

Cou

nts

/ s

Binding Energy (eV)

N

C0

C0

5000

10000

15000

20000

25000

02004006008001000

Cou

nts

/ s

Binding Energy (eV)

O

N

C

Figure 19 XPS survey of FN immobilized PET surface. A) Clean PET surface; B) Fn conjugated surface; C) Fn adsorbed surface

59

Table 5 Element proportion on FN immobilized surface

Element Percentages Surface

C O N

Fn conjugated PET 67.4 26.47 6.13

Fn adsorbed PET 70.07 24.14 5.79

6.1.3 Surface contact angle measurements

The contact angle between water and a surface is an inverse measure of the

ability of water to ‘‘wet’’ the surface. A smaller contact angle indicates a hydrophilic

surface, on which water spreads to a greater extent; a larger contact angle indicates a

hydrophobic surface, on which water beads up.

The cleaned and unmodified PET surface showed a static contact angle of

74.1º ± 2.6º. Amination of PET surface with ethylenediamine lower the contact angle

to 62.9º ± 3.1º by introducing the more polar amino groups on the surface. The

conjugation of FN on aminated surface decreased the angle further to 56.7º ± 2.0º).

The ethanolamine blocked surface had similar contact angle with the aminated surface

(61.5º ± 2.9º), and the FN adsorption onto this surface did not change the contact

angle further (61.8º± 2.1º). While the mechanism is not clear, the difference in the

contact angles between the FN- conjugated and the FN-adsorbed surfaces (p<0.05)

suggest dissimilarity in the conformation of FN presented on the surface.

6.1.4 AFM assay

In AFM assay, a sharp tip attached to a cantilever scanned across a surface.

Changes in surface topography that were encountered as the tip moved over the

material’s surface changed inter atomic attractive or repulsive forces between the

60

surface and tip. These forces were sensed and recorded to construct images of surface

3D topography.

50

55

60

65

70

75

80

Clean PET Aminated PET Fn conjugatedPET

Ethanolamineblocked PET

Fn adsorbedPET

Surf

ace

cont

act a

ngle

Figure 20 Surface contact angle on clean, modified or FN immobilized PET surface (n>5). (p<0.05. between Fn conjugated PET and Fn adsorbed PET)

A) B)

Figure 21 AFM image of FN immobilized PET surface. (A) FN conjugated PET surface, scanning area: 4.8um*4.8um. Ra: 119 nm, Rmax: 522 nm. (B) FN adsorbed PET surface, scanning area: 10um*10um Ra: 11.2nm, Rmax: 217nm.

Different morphology was observed when FN was conjugated or adsorbed to

the PET surfaces in the AFM topographic images (Figure 21). On the FN-adsorbed

PET surface, the protein appeared evenly distributed, with a mean roughness of

Ra=11.2 nm and a maximal roughness of 217 nm. In contrast, the protein on the FN-

conjugated PET surface was more closely packed in layers, with a mean roughness of

Ra=119 nm and a maximal roughness of 522 nm. Due to this kind of aggregation, the

FN conjugated surface maintained higher FN density. Neither of the modification

61

processes changed surface area significantly (scanning area: 1um2, conjugation:

1.06um2, adsorption: 1.13 um2).

6.2 Biological characterization of conjugated and adsorbed FN

6.2.1 ELISA test for RGD domain

Although the FN surface density was higher in the FN-conjugated surface

according to the micro-BCA result, the ELISA test for active RGD domains showed

that FN bioactivity in the access to active RGD was lower in FN conjugated surface.

(Figure 22).

0

200

400

600

800

1000

1200

Conjugation Adsorption

FN d

ensi

ty n

g/cm

2

ELISAMicro-BCA

Figure 22 ELISA measurement of active RGD domains in immobilized FN compared with Micro –BCA result. (FN input concentration of 30ng/ml: conjugated FN surface density: 917.6±133.8 ng/cm2; adsorbed FN surface density: 433.1±45.8 ng/cm2 by micro-BCA) Significant difference existed between two methods *p<0.05 (n>4).

There was little difference between the micro-BCA and ELISA measurements

for the surface density of adsorbed FN, 433.1±45.8 ng/cm2 versus 410.8±13.7 ng/cm2,

respectively, at the FN input concentration of 30ug/ml. In contrast, the corresponding

values for the conjugation method were 917.6±133.8 ng/cm2 and 220.3 ± 10.1 ng/cm2,

which indicated a loss of 76% of the active RGD domains.

62

6.2.2 Ex vivo cell experiment

To further assess the bioactivity of FN adsorbed surface and FN conjugated

surface, we compared the measured cell behaviors as a function of surface. The cells

viability over 24 hours on the two FN-immobilized PET surfaces did not show any

significant difference according to the MTT assay. However, the adhesion of BHK21

cells at the 2 hours time point was quite different (Figure 23). The FN-adsorbed PET

surface supported cell adhesion (from 53.6±10.2% to 70.3±9.4%) with the increase of

adsorbed FN surface density (from 0 to 433.1±45.8 ng/cm2). The FN-conjugated

surface on the other hand suppressed cell adhesion (from 53.6±10.2% to 12.2±6.0%)

with the increase of FN density (from 0 to 917.6±133.8 ng/cm2).

0

20

40

60

80

100

0 200 400 600 800 1000FN surface density ng/cm2

Aad

here

nt c

ells

perc

enta

ges %

Conjugation Adsorption

Figure 23 Cell adhesion on FN immobilized PET surface (n>4)

6.3 Discussion

6.3.1 FN conformation and its bioactivity

Many studies have shown that the conformation flexibility influences the

biological properties of FN. Certain activities of FN are latent in the intact molecule

and become fully expressed in its proteolytic fragments [Muir and Manthorpe (1992)].

For example, 29 and 40 kDa heparin-binding proteolytic fragments of FN are potent

63

inhibitors of endothelial growth ex vivo, while native FN is not [Homandberg et al

(1985)]. Generally it is believed that FN molecules is secreted by cells and folded into

globular domains specialized for particular functions, such as binding to integrins,

collagen, heparin sulfate, hyaluronic acid, the change of their conformation would

affect their bioactivity. [Morla and Ruoslahti (1992)]

6.3.2 FN adsorption

Langmuir isotherm was often used for the rudimentary description of protein

adsorption phenomena: the near-linear dependence of adsorbed molecules on the bulk

concentration at low concentration regimens and the saturation effect at higher

adsorbent concentrations [Horbett (1994); Mientus and Knippel (1995)], which was

quite consistent with our result. Saturation level suggested the formation of monolayer

of FN on surface, so the extra FN unable to form stable bonding onto the FN

monolayer, was washed off. Monolayer coverage for proteins theoretically depends

on the shape and size of the protein molecules as well as on its mode of adsorption.

Varying monolayer coverage of protein surface density has been reported to range

between 200ng/cm2 to 500ng/cm2 [Rezania and Healy (1999); Lhoest et al (1998)]. In

this experiment, we reported a saturation surface density around 400 ng/cm2.

Although we could not compare the adsorbed FN with soluble FN by ELISA or cell

adhesion, the AFM photos showed that the adsorbed FN kept its global compact

conformation, uniformly distributed on the surface, having less structure alteration

compared with conjugated FN.

6.3.3 FN conjugation and fibrillogenesis

64

In solution, the compact conformation of FN is maintained by only a small

number of electrostatic associations between distant segments of the molecule: III12–

14/ III2–3, and III12–14/ amino-terminal Heparin I domains [Johnson et al (1999)].

However the electrostatic associations can be destroyed in a regulated stepwise

process, fibrillogenesis, in which soluble FN is converted into insoluble fibrillar

network. In initiation process: integrins at cells surface recognize the cell-binding

domains of FN, usually α5β1 binding to RGD domains or α4β1 binding to CS domains.

It induces FN unfolding that exposes FN binding sites and promotes intermolecular

interactions with other FN. Finally the integrins clustering and binding of additional

FN leads to fibril formation [Bentley et al (1985); Patynowski and Schwarzbauer

(2003)].

According to the protein immobilization result and morphology observed in

AFM, we hypothesize a cell-free fibrillogenesis model in the conjugation process

(Figure 24). At first the amino groups of the protein were randomly immobilized to

the surface, which helped FN to unfold. While the positive charged amino groups

were strongly immobilized to the substrate, the negative charged domains of the

protein were driven away from the surface, forming a negative charge concentrated

“surface”. This negatively charged “surface” could immobilize the amino groups of

the FN by electrostatic association, help the FN unfold, and form new adsorption

“surface” repeatedly. Thus it formed the multiple layers of FN onto the surface, and

initiated the fibrillogenesis process. In summary, this proposed model showed that

the orientation of the FN molecules on amino groups conjugated surface and

subsequent unfolding of the molecule may be the key to the induction of

fibrillogenesis. At high surface charge densities, the created electric field would repel

the negative domains and strongly attract the positive domains, leaving open and

65

accessible the domains implicated in the fibrillogenesis process. By this model we can

explain how conjugation method stably immobilized multiple layers of FN, and how

fibrils like structure formed on surface. Pernodet also reported similar cell-free

fibrillogenesis on surfonated polystyrene surface after 130 hours of 100ug/ml FN

[Pernodet et al (2003)]. Compared his result, our conjugation method showed more

efficiency in initiating fibrillogenesis.

A

B

C

D

Type I Repeat Variable Sequence Type II Repeat Type III Repeat

Figure 24 Hypothetical schemes of FN fibrillogenesis in conjugation process. A: Compact conformation of FN in solution is stabilized by III12-14/III2-3 and III12-14/I domains. B: FN unfolds when the amino groups of the FN are immobilized to the surface. C: The unfolded FN exposes new layer of negatively charged “surface” for extra FN adsorption. D: FN adsorbes and partially unfoldes on the FN layer through electronic association, exposes new “surface” for FN adsorption.

It is increasingly apparent that FN matrix can have profound effects on cell

functions. Data from structural modeling of repeat III10 indicate that the unfolding of

the FN and the following fibrillogenesis process might control the accessibility of the

RGD sequence, and enable cells to retract from FN matrices in order to migrate and

proliferate [Krammer et al (1999)]. Data from endothelial cells adhering assays, also

66

reported that after fibrillogenesis on endothelial cells surface, the FN matrix prevent

the extra cells adhering [Dekker et al (1993)]. Our experiment also showed that the

conjugation induced fibrillogenesis suppressed the accessibility of RGD domains, also

suppressed cells adhesion, which was quite consistent with the literature mentioned

above.

In summary this project studied the regulation of FN involved cell-material

interaction at the level of FN immobilization methods. In the adsorption method, FN

adsorbed as isolated molecules or as a monolayer of protein, and preserved higher

bioactivity in maintaining more RGD domains and favoring cells adhesion. The

conjugation method induced FN fibrillogenesis, which maintained less RGD domains

and suppressed cells adhesion. The combination of these two methods may offer us

the ability to readily control the degree of surface bioactivity, which has particular

significance to the biomaterials applications like microenvironment reconstruction. It

has been noted that it is a relatively easy task to generate a highly adhesive substrate

by direct adsorption of cells adhesion molecules like FN. However a difficulty often

encountered in application is the inability of cells, once attached to an overly adhesive

bridging substrate, to leave the material surface for the comparatively less adhesive

surrounding [Schwab and Bartholdi (1996)]. In this regard, promoting cell migration

away from the device surface probably will require that the surface become less

adhesive in comparison with surrounding tissue, which often is not recognized in the

development of bioactive surfaces.

67

CHAPTER 7

CONCLUSIONS

7.1 Conclusions

The ultimate goal of this thesis project was to develop a HSC ex vivo

expansion system that would be able to produce sufficient and functional HSCs for

the allogeneic transplantation of CB. To fulfill this goal, we aimed to design an

artificial microenvironment that mimicked the hematopoiesis process in vivo, which

can efficiently expanded the cell number and still maintained the “stemness” of the

cells.

We chose MSC, non hematopoietic, well-characterized skeletal and

connective tissue progenitor cells within the bone marrow, as stroma to support HSC

expansion ex vivo. First, MSC is relatively easy to harvest and culture ex vivo. MSC

provides a rich environment of signals, including cytokines, ECM proteins, adhesion

molecules, and cell-cell interactions, controlling the proliferation, survival, and

differentiation of early lympho- hematopoietic stem cells [Haynesworth et al (1996),

Majumdar et al (2000)]. What is more, MSCs, like other stem cells, are poor antigen

presenting and relatively immunologically compatible.

In this study we clearly demonstrated the benefit of MSC inclusion in HSC

expansion ex vivo. We tested the co culture result under different conditions: fresh

stroma & irradiated stroma co-culture; stroma exchanged & non stroma exchanged

co-culture; feeder layer & pellet co-culture; contact & non-contact co-culture; 2D &

3D co-culture. The results showed that MSC can survive in the serum free StemSpan

medium supplemented with growth factors and LDL. MSC’s metabolic level affected

68

the expansion result. Neither too low nor too high metabolic level was favorable for

CB CD34+ cells expansion.

Compared with non-contact co-culture in transwell system, contact co-culture

increased higher expansion of total cells (545.4 vs.316.5 fold; p<0.05), CD34+ cells

(47.9 vs. 21.1 fold; p<0.05), CD34+CD38- cells (100.4 vs. 52.8 fold; p<0.05). Similarly,

the pluripotency of cells expanded in contact co-culture as evaluated by CFC assay was

also superior compared to those expanded in non-contact co-culture (CFU-GEMM:

84.1 vs. 76.0 fold; total CFU 150.1 vs. 116.5 fold; p<0.05). The animal engraftment

assays also showed that cells expanded in contact co-culture system appeared to be

more competent in reconstructing short term hematopoiesis. These results suggested

that cell-cell contact was required for the further expansion of stem cells; only growth

factors in conditioned medium could not fully replace the function of stroma. The long

term tracing staining- CMFDA experiment showed that about 1.6% of the expanded

cells had communicated with stroma in culture, which were stained with CMFDA. And

63.9% of these CMFDA+ cells expressed CD34 surface marker, which was about 8

times higher than CMFDA- suspension cells. The results highly suggested that cell-cell

communication existed between MSCs and CB cells in contact co-culture, and that

kind of cell-cell communication helped to reserve stem cells primitivity and prevent

them from differentiation.

The conformation of the co-culture system also significantly affected stem

cells expansion. The result showed that the direct contact between MSC and HSC in

3D cultures led to statistically-significant higher expansion of CB CD34+ cells

compared to 2D co-cultures: 891- versus 545-fold increase in total cells, 96- versus

48-fold increase of CD34+ cells, and 230- versus 150-fold increase in CFC.

NOD/SCID mouse engraftment assays also indicated a high success rate of

69

hematopoiesis reconstruction with these expanded cells. Cytochemical staining

revealed that culture of MSCs on 3D mesh led to increased proliferation which may

contribute to the increase in beneficial effects on CB expansion. More importantly, we

observed high expression of ECM fibrils on MSCs grown in 3D culture, especially

FN, a feature not seen for cells grown on 2D culture. This 3D scaffold induced fibrous

structure and excessive expression of ECM protein facilitated and regulated CB

CD34+ cells expansion, which probably explained the enhanced result.

Inspired by the finding that FN was the mainly expressed ECM protein by

MSCs, and FN’s influence in HSC cell cycle [Dao et al (1998)], we hypothesize that

the immobilization of ECM protein FN onto the surface of culture systems might

represent another stroma free expansion strategy. A key problem in FN

immobilization is the change of bioactivity during the immobilization process. We

studied FN bioactivity after two common immobilization pathways: conjugation and

adsorption, we found a way to control the function of the immobilized protein on the

level of immobilization methods, which can be helpful for the construction of an

artificial niche in the future. The adsorption method maintained a normal

conformation of FN, leading to less change of its bioactivity in the access of RGD

domains and cell adhesion ability. The covalent conjugation process, on the other

hand, induced FN unfolding and fibrillogenesis ex vivo, which blocked the access of

active RGD domains and suppressed fibroblast cells adhesion. The HSC expansion on

these FN immobilized scaffolds also showed very promising results [Q. Feng et al,

unpublished data].

Taken together, we designed a 3D bio-artificial “niche” coupled with elevated

deposition of ECM by MSC, which more closely resembled the natural bone marrow

microenvironment; therefore, it favored CD34+ cell proliferation and maintenance of

70

pluripotency. We also studied FN bioactivity after immobilization, which helped us to

construct another stroma free system supporting CD34+ cells expansion.

7.2 Future prospect

In summary, a great deal of experiences over the past 15 years in terms of the

ability to isolate and culture primitive hematopoietic stem and progenitor cells has

been attempted. Especially the research in growth factors and other soluble factors of

stem cell proliferation have provided great insight into the biology of stem cells, but

we do not yet understand how to organize these molecules in a physiologically

relevant manner. The perpetual discoveries in molecular biology and developmental

biology help this process cooperatively. For example, recently Lodish group isolated a

new kind of HSC-supportive cells: mouse fetal liver CD3+ cells. Subsequently, they

identified two growth factors: angiopoietin like 2 and angiopoietin like 3 specially

expressed by these HSC-supportive cells by microarray expression analysis. The 10

days expansion in the presence of these two factors, induced up to 30 net expansion of

long-term HSC by reconstitution analysis, which was the highest stem cell expansion

that had ever been reported [Zhang et al 2006]. These researches pave the way to the

establishment of the procedure for clinical application and build up hope for these

patients.

However, there is still a long way to translate these research results into clinics.

To be widely accepted, procedures for ex vivo stem cell expansion will have to meet

the requirements of Federal Good Manufacturing Practices (GMPs). The key concepts

of GMPs are:

o Preventing the transmission of communicable disease.

71

o Processing controls, for example, prevent contamination and preserve integrity

and function.

o Assuring clinical safety and effectiveness, especially with presence of

allogeneic materials, for example the allogeneic MSC in this study

That is to say products (for infusion) must be produced in a manner that ensures that

they are safe and (reproducibly) fit for their intended use.

So far, we seem to only have safety data. Studies Allogeneic MSC expanded

in culture could be successfully co-infused with T-cell depleted peripheral blood

hematopoietic progenitors into a haploidentical receipient and it is currently under

further clinical evaluation [Lee et al (2002)]. And a short term standard lymphocytes

mixture test is recommended before transplantation, to make sure expanded cells with

MSCs will not induce immunological reaction.

The uniformity and effectiveness of expanded products is still under the

developmental stages. As mentioned before, the main limit of CB allogeneic

transplantation is insufficient cells number. In transplantation CB nucleated cells,

about 2,500,000 CD34+ cells per kg body weight is required [Koller et al 1973].

Therefore, for a patient of 70 kg body weight, 175,000,000 CD34+ cells are required.

Based on the 3D co-culture system mentioned above, 96-fold expansion of CD34+

cells can be achieved. about 1,820,000 CD34+ cells are required to start expansion.

More than 3000 scaffolds are needed, which is quite unpractical. A clinical-scale

bioreactor must be developed to accommodate large number of cells and scaffolds

with high cell densities. With this developed culture system of sufficient simplicity,

flexibility and economic efficacy, the true potential of ex vivo expansion of HSCs that

mimic the in vivo conditions will finally be realized.

72

REFERENCES

Ahmed SA, Gogal JRM, Walsh JE. 1994. A new rapid and simple non radio-active assay to monitor and determine the proliferation of leukocytes: an alternative to H3-thymidine incorporation assay. J Immunol Method 170: 211-224

Andrade JD, Hlady V. 1986. Protein adsorption and materials biocompatibility: a tutorial review and suggested hypotheses. Adv Polym Sci 79:1–63

Balduini A, Braun SE, Cornetta K et al. 1998. Comparative effects of retroviral-mediated gene transfer into primary human stromal cells of Flt3-ligand, interleukin 3 and GM-CSF on production of cord blood progenitor cells in long-term culture. Stem Cells 16 Suppl 1:37-49

Banu N, Rosenzweig M, Kim H, et al. 2001. Cytokine-augmented culture of haematopoietic progenitor cells in a novel three-dimensional cell growth matrix. Cytokines 13: 349-358

Baum CM, Weissman IL, Tsukamoto AS, et al. 1992. Isolation of a candidate human hematopoietic stem-cell population. Proc Natl Acad Sci 89:2804–2808

Bentley KL, Klebe RJ, Hurst RE, Horowitz PM. 1985. Haparin binding is necessary, but not sufficient, for fibronectin aggregation. A florescence polarization study. J Biol Chem 260:7250-7256

Bentley SA, Tralka TS 1983 Fibronectin-mediated attachment of hematopoietic cells to stromal elements in continuous bone marrow culture. Exp Hematol 11:129-138

Bhatia M, Bonnet D, Wu D, et al. 1999. Bone morphogenetic proteins regulate the developmental program of human hematopoietic stem cells. J Exp Med 189:1139–1148

Bianco P, Riminucci M,Gronthos S, Robey PG. 2001. Bone marrow stromal stem cells: nature, biology, and potential applications. Stem Cells 19:180-192

Biran R, Webb K, Noble MD, Tresco PA. 2001. Surfactant-immobilized fibronectin enhances bioactivity and regulates sensory neurite outgrowth. J Biomed Mater Res 55(1):1-12

Breems DA, Blokland EA, Siebel KE et al. 1998. Stroma-contact prevents loss of hematopoietic stem cell quality during ex vivo expansion of CD34+ mobilized peripheral blood stem cells. Blood 91(1):111-117

Brockbank KGM, de Jong JP, Piersma AH,Voerman JSA 1986 Hemopoiesis on purified bone-marrow-derived reticular fibroblast in vitro. Exp Hematol 14:386-394

Broudy VC. 1997. Stem cell factor and hematopoiesis. Blood 90:1345–1364

Calvi LM, Adams GB, Weibrecht KW at al. 2003. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature 425:841-846

Campbell AD, Long MW, Wicha MS. 1987. Haemonectin, a bone marrow adhesion protein specific for cells of granulocyte lineage Nature 329:744-746

Cashman JD, Lapidot T, Wang JC, et al. 1997. Kinetic evidence of the regeneration of multilineage hematopoiesis from primitive cells in normal human bone marrow transplanted into immunodeficient mice. Blood 89:4307–4316

73

Coulombel L,Vullet MH, Leroy C, Tehernia G. 1988. Lineage- and stage-specific adhesion of human hematopoietic progenitor cells to extracellular matrices from marrow fibroblasts. Blood 71:329-334

Crocker PR, Gordon S. 1987. In: Rich IN (ed) Molecular and cellular aspects of erythropoietin and erythropoiesis. Springer, Berlin Heidelberg New York, p 259

Dao MA, Hashino K, Kato I, et al. 1998. Adhesion to Fibronectin Maintains Regenerative Capacity During Ex Vivo Culture and Transduction of Human Hematopoietic Stem and Progenitor Cells. Blood 92: 4612-4621

Dekker A, Beugeling T, Poot A, et al. 1993. The role of cellular fibronectin in the interaction of human Endothelial cells with polymers. Clinic Materials 13: 101-107

Devine SM, Bartholomew AM, Mahmud N, et al. 2001. Mesenchymal stem cells are capable of homing to the bone marrow of non-human primates following systemic infusion. Exp Hematol 129: 244-255

Dexter TM, Allen TD, Lajtha LG, et al. 1973. Stimulation of differentiation and proliferation of haemopoietic cells in vitro. Cell Physiol 82: 461-473

Dexter TM. 1989. Regulation of hemopoietic cell growth and development: experimental and clinical studies Leukemia 3:469-474

Gartner S, Kaplan HS. 1980. Long-term culture of human bone marrow cells. Proc Natl Acad Sci 77:4756-4759

George KT, Russell JC, Christopher SA, Irving MS, Paul D.2004. Fibronectin adsorption on surface-activated poly(dimethylsiloxane) and its effect on cellular function. J Biomed Mater Res A 71(3):449-461

Gluckman E, Broxmeyer HA, Auerbach AD, et al. 1989. Hematopoietic reconstitution in a patient with Fanconi’s anemia by means of umbilical-cord blood from an HLA-identical sibling. N. Engl. J. Med. 321: 1174–1178.

Gluckman, E. 2001. Hematopoietic stem-cell transplants using umbilical-cord blood. N. Engl. J. Med. 344: 1860–1861.

Gordon MY, Riley GP, Clarke D. 1988. Heparan sulfate is necessary for adhesive interactions between human early hemopoietic progenitor cells and the extracellular matrix of the marrow microenvironment. Leukemia 2:804-809

Grayson W.L, Ma T., Bunnell B. 2004. Human Mesenchymal Stem Cells Tissue Development in 3D PET Matrices. Biotechnol. Prog. 20: 905-912

Greenberger JS. 1978. Sensitivity of corticosteroid-dependent insulin-resistant lipogenesis in marrow preadipocytes of obese-diabetic (db/db) mice. Nature 275:752-754

Grinnell F, Feld MK. 1982. Fibronectin adsorption on hydrophilic and hydrophobic surfaces detected by antibody binding and analyzed during cell adhesion in serum-containing medium. J Biol Chem 257:4888–4893

Gupta P, Oegema Jr TR, Brazil JJ et al. 2000. Human LTC-IC can be maintained for at least 5 weeks in vitro when interleukin-3 and a single chemokine are combined with O-sulfated

74

heparan sulfates: requirement for optimal binding interactions of heparin sulfate with early-acting cytokines and matrix proteins. Blood 95:147-155

Haylock DN, Horsfall MJ, Dowse TL, et al. 1997. Increased recruitment of hematopoietic progenitor cells underlies the ex vivo expansion potential of FLT3 ligand. Blood 90:2260–2272

Haynesworth SE, Baber MA, Caplan AI. 1992. Cell surface antigens on human marrow-derived mesenchymal cells are detected by monoclonal antibodies. Bone 13:69-80

Haynesworth SE, Baber MA, Caplan AI. 1996. Cytokine expression by human marrow-derived mesenchymal progenitor cells in vitro: effects of dexamethasone and IL-1 alpha. J Cell Physiol 166:585-592

Highfill JG, Haley S and Kompala DS. 1996. Large-scale production of murine bone marrow cells in an airlift packed bed bioreactor. Biotechnol Bioeng 50: 514-520

Hogge DE, Lansdorp PM, Reid D, et al. 1996. Enhanced detection, maintenance, and differentiation of primitive human hematopoietic cells in cultures containing murine fibroblasts engineered to produce human steel factor, interleukin-2, and granulocyte colonystimulating factor. Blood 88:3765–3773

Holyoake TL, Nicolini FE, Eaves CJ. 1999. Functional differences between transplantable human hematopoietic stem cells from fetal liver, cord blood, and adult marrow. Exp Hematol 27:1418-1427

Homandberg G, Williams JE, Grant D et al. 1985. Heparin-binding fragments of fibronectin are potent inhibitors of endothelial cell growth. Am J Pathol 120: 327-332

Horbett TA. 1994. The role of adsorbed proteins in animal cell adhesion. Colloid Surfaces B Biomater 2:225–240

Iscove NN, Shaw AR, Keller G. 1989. Net increase of pluripotential hematopoietic precursors in suspension culture in response to IL-1 and IL-3. Immunol 142: 2332-2337

Johnson KJ, Sage H, Briscoe G, Erickson HP. 1999. The compact conformation of Fibronectin is determined by intramolecular ionic interactions. J Biol Chem 73:440-443

Kadereit S, Deeds LS, Haynesworth SE et al. 2002. Expansion of LTC-ICs and Maintenance of p21 and BCL-2 Expression in Cord Blood CD34+/CD38– Early Progenitors Cultured over Human MSC as a Feeder Layer. STEM CELLS 20:573-582

Karanu FN, Murdoch B, Gallacher L, et al. 2000. The notch ligand jagged-1 represents a novel growth factor of human hematopoietic stem cells. J Exp Med 192:1365–1372

Kay H. 1965. How many cell-generations? Lancet 11:418-428

Kiger AA, White-Cooper H, Fuller MT. 2000. Somatic support cells restrict germline stem cell self-renewal and promote differentiation. Nature 407:750-754

Koc ON, Gerson SL, Cooper BW, et al. 2000. Rapid hematopoietic recovery after coinfusion of autologous-blood stem cells and culture-expanded marrow mesenchymal stem cells in advanced breast cancer patients receiving high-dose chemotherapy. J Clin Oncol 18: 307-316

75

Kogler G, Nurnberger W, Fischer J et al. 1999. Simultaneous cord blood transplantation of ex vivo expanded together with nonexpanded cells for high risk leukemia. Bone Marrow Transplant 24:397-403

Koller M.R. Emerson S.G. Palsson B.O.1993. Large-scale expansion of human stem and progenitor cells from bone marrow mononuclear cells in continuous perfusion cultures. Blood 82: 378-384

Koller MR, Manchel I, Newsom BS, et al. 1995. Bioreactor expansion of human bone marrow: comparison of unprocessed, density-separated, and CD34-enriched cells. J Hematother. 4: 159-169

Koller M.R., Manchel I, Maher RJ et al.1998. Clinical-scale human umbilical cord blood cell expansion in a novel automated perfusion culture system. Bone Marrow Transplant 21:653-663

Kondo M., Wagers A.J., Manz M.J., Prohaska, S.S., Scherer D.C., Beilhack G.F., Shizuru J.A., Weissman I.L., Biology of hematopoietic stem cells and progenitors: Implications for Clinical Application. Annu. Rev. Immunol. 2003. 21:759–806

Kowalczynska HM, Nowak-Wyrzykowska M, Kolos R, et al. 2005. Fibronectin adsorption and arrangement on copolymer surfaces and their significance in cell adhesion. J Biomed Mater Res A 1;72(2):228-236

Krammer A, Lu H, Isralewitz B, et al. 1999. Forced unfolding of fibronectin type III module reveals a tensile molecular recognition switch. Proc Natl Acad Sci 96:1351-1356

Krause DS, Fackler MJ, Civin CI, May WS. 1996. CD34: structure, biology, and clinical utility. Blood 87:1–13

Lazzari L, Lucchi S, Rebulla P et al. 2001. Long-term expansion and maintenance of cord blood haematopoietic stem cells using thrombopoietin, Flt3-ligand, interleukin (IL)-6 and IL-11 in a serum-free and stroma-free culture system. Br. J. Haematol. 112(2):397-404

Leary AG, Ikebuchi K, Hirai Y, et al. 1988. Synergism between interleukin-6 and interleukin-3 in supporting proliferation of human hematopoietic stem cells: comparison with interleukin-1 alpha. Blood 71: 1759-1763

Lee ST, Jang JH, Cheong JW, et al. 2002. Treatment of high-risk acute myelogenous leukaemia by myeloablative chemoradiotherapy followed by co-infusion of T cell-depleted haematopoietic stem cells and culture-expanded marrow mesenchymal stem cells from a related donor with one fully mismatched human leucocyte antigen haplotype. Br J Haematol 118: 1128-1131

Lhoest J-B, Detrait E, van den Bosch de Aguilar P, Bertrand P. 1998. Fibronectin adsorption, conformation, and orientation on polystyrene substrates studied by radiolabeling, XPS, and ToF SIMS. J Biomed Mater Res 41:95–103

Li Y, Ma T, Douglas A. et al. 2001. Human Cord Cell Hematopoiesis in three-dimensional nonwoven fibrous matrices: In vitro simulation of the marrow microenvironment. Journal of hematotherapy stem cell reseach 10:355–368

Long MW, Dixit VM. 1990. Thrombospondin functions as a cytoadhesion molecule for human hematopoietic progenitor cells. Blood 75:2311-2318

76

Luens KM, Travis MA, Chen BP, et al. 1998. Thrombopoietin, kit ligand, and flk2/flt3 ligand together induce increased numbers of primitive hematopoietic progenitors from human CD341Thy-11Lin2 cells with preserved ability to engraft SCID-hu bone. Blood 91:1206–1215

Lyman SD, Jacobsen SE. 1998. c-kit ligand and Flt3 ligand: stem/progenitor cell factors with overlapping yet distinct activities. Blood 91:1101–1134

Majka M, Janowska-Wieczorek A, Ratajczak J, et al. 2001. Numerous growth factors, cytokines, and chemokines are secreted by human CD34(1) cells, myeloblasts, erythroblasts, and megakaryoblasts and regulate normal hematopoiesis in an autocrine/ paracrine manner. Blood 97:3075–3085

Majumdar MK, Thiede MA, Mosca JD et al. 1998. Phenotypic and functional comparison of cultures of marrow-derived mesenchymal stem cells (MSCs) and stromal cells. J Cell Physiol 176:57-66

Majumdar MK, Thiede MA, Haynesworth SE et al. 2000. Human marrow-derived mesenchymal stem cells (MSCs) express hematopoietic cytokines and support long-term hematopoiesis when differentiated toward stromal and osteogenic lineages. J Hematother Stem Cell Res 9:841-848

Mantalaris A, Keng P, Bourne P, et al. 1998 Engineering a human bone marrow model: A case study on ex vivo erythropoiesis. Biotechnol Progr 14:126–133

Maximow A. 1909. Der Lymphozyt als gemeinsame Stammzelle der verschiedenen Blutelemente in der embryonalen Entwicklung und im postfetalen Leben der Säugetiere. Folia Haematologica VIIII 8:125-134

McNiece I, Harrington J, Turney J, et al. 2004. Ex vivo expansion of cord blood mononuclear cells on mesenchymal stem cells. Cytotherapy l: 1-7

Meyer C, Drexler HG. 1999. FLT3 ligand inhibits apoptosis and promotes survival of myeloid leukemia cell lines. Leuk Lymphoma 32:577–581

Mientus W, Knippel E. 1995. Theoretical modeling of plasma protein adsorption/desorption processes onto solid surfaces. J Biomater Sci Polym Ed 7:401– 414

Morla A, Ruoslahti E. 1992. A fibronectin self-assembly site involved in matrix assembly: reconstruction in a synthetic peptide. J Cell Biol 118: 421–429

Muir D, Manthorpe. 1992. Stromelysin generates a fibronectin fragment that inhibits Schwann cell proliferation. M. J Cell Biol 116: 177-185

Naughton BA, Preti RA, Naughton GK. 1987. Hematopoiesis on nylon mesh templates I. Long-term culture of rat bone marrow cells. J Med 18:219–250

Naughton BA, Jacob L, Naughton GK. 1990. A three dimensional culture system for the growth of hematopoietic cells. Progr Clin Biol Res 333:435–445

Nishioka K, Fujimori Y, Hashimoto-Tamaoki T, et al. 2003. Immortalization of bone marrow-derived human mesenchymal stem cells by removable simian virus 40T antigen gene: analysis of the ability to support expansion of cord blood hematopoietic progenitor cells. Int J Oncol. 23(4):925-932

77

Ohmizono Y, Sakaba H, Kimura T, et al. 1997. Thrombopoietin augments ex vivo expansion of human cord blood-derived hematopoietic progenitors in combination with stem cell factor and flt3 ligand. Leukemia 11:524–530

Osawa M, Hanada K, Hamada H, Nakauchi H.1996. Long-term lymphohematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell. Science 273: 242–245

Patynowski IW, Schwarzbauer JE. 2003. The ins and outs of fibronectin matrix assembly. J Cell Sci 116:3269-3276

Pecora AL, Stiff P, Jennis A et al. 2000. Prompt and durable engraftment in two older adult patients with high risk chronic myelogenous leukemia (CML) using ex vivo expanded and unmanipulated unrelated umbilical cord blood. Bone Marrow Transplant 25:797-799

Pernodet N, Rafailovich M, Sokolov J, et al. 2003. Fibronectin fibrillogenesis on sulfonated polystyrene surfaces. J Biomed Mater Res A. 64(4):684-692

Pittenger MF, Mackay AM, Beck SC et al. 1999. Multilineage potential of adult human mesenchymal stem cells. Science 284:143-147

Quesenberry P, Temeles D, McGrath H, et al. 1991. Long-term marrow cultures: human and murine systems. J Cell Biochem. 45: 273-278

Quito FL, Beh J, Bashayan O, et al. 1996. Effects of fibroblast growth factor-4 (k-FGF) on long-term cultures of human bone marrow cells Blood 87:1282-1291

Ramirez F, Rifkinb DB. 2003. Cell signaling events: a view from the matrix. Matrix Biology 22: 101–107

Rezania A, Healy KE. 1999. Biomimetic peptide surfaces that regulate adhesion and mineralization of osteoblast-like cells. Biotechnol Prog 15:19 –32

Rich IN. 1988. The macrophage as a production site for hematopoietic regulator molecules: sensing and responding to normal and pathophysiological signals. Anticancer Res 8:1015-1040

Roberts R, Gallagher JT, Spooncer E, Allen TD. 1988. Heparan sulphate bound growth factors: a mechanism for stromal cell mediated haemopoiesis. Nature 332:376-378

Rocha V, Wagner JE Jr, Sobocinski KA, et al. 2000. Graft-versus-host disease in children who have received a cord-blood or bone marrow transplant from an HLA-identical sibling. Eurocord and International Bone Marrow Transplant Registry Working Committee on Alternative Donor and Stem Cell Sources. N Engl J Med 342(25):1846-1854

Rosenzweig M, Pykett MJ, Marks DF, et al. 1997. Enhanced maintenance and retroviral transduction of primitive hematopoietic progenitor cells using a novel three-dimensional culture system. Gene Therapy 4:928–936

Rubinstein P, Dobrila L, Rosenfield RE et al.1995. Processing and cryopreservation of placental/umbilical cord blood for unrelated bone marrow reconstitution. Proc Natl Acad Sci 92:10119-10122

Sandstorm CE, Bender JG, Miller WM, Papoutsakis ET. 1996. Development of novel perfusion chamber to retain nonadherent cells and its use for comparison of human

78

mobilized peripheral blood mononuclear cell cultures with and without irradiated bone marrow stroma. Biotechnol Bioeng 50: 493-504

Sardonini CA, Wu YJ. 1993. Expansion and differentiation of human hematopoietic cells from static cultures through small-scale bioreactors. Biotechnol Prog 9: 131-137

Schwab ME, Bartholdi D. 1996. Degeneration and regeneration of axons in the lesioned spinal cord. Physiol Rev 76:319–370

Schwartz RM, Palsson BO, Emerson SG. 1991. Rapid medium perfusion rate significantly increases the productivity and longevity of human bone marrow cultures. Proc. Natl. Acad. Sci 88: 6760-6764

Shpall EJ, Quinones R, Giller R, et al. 2002. Transplantation of ex vivo expanded cord blood. Biol Blood Marrow Transplant. 8(7):368-376

Staffan J, Gunbjørg S, Krister W, et al. 1997. Fibronectin- integrin interactions. Frontiers in Bioscience 2:126-137

Sutherland HJ, Lansdorp PM, Henkelman DH, et al. 1990. Functional characterization of individual human hematopoietic stem cells cultured at limiting dilution on supportive marrow stromal layers. Proc Natl Acad Sci 87:3584–3588

Tavassoli M, Maniatis A, Crosby WH. 1974. Induction of sustained hemopoiesis in fatty marrow Blood 43:33-38

Till JE, McCullough EA. 1961. A direct measurement of the radiation sensitivity of normal mouse bone marrow cells. Radiat. Res. 14: 213-222

Terstappen LW, Huang S, Safford M, et al. 1991. Sequential generations of hematopoietic colonies derived from single nonlineage-committed CD34+CD38- progenitor cells. Blood 77:1218–1227

Tran J, Brenner TJ, DiNardo S. 2000. Somatic control over the germline stem cell lineage during Drosophila spermatogenesis. Nature 407:754-757

Varnum FB, Xu L, Brashem-Stein C, et al. 2000. Pluripotent, cytokine-dependent, hematopoietic stem cells are immortalized by constitutive Notch 1 signaling. Nature Med 6:1278–1281

Vormoor J, Lapidot T, Pflumio F, et al. 1994. Immature human cord blood progenitors engraft and proliferate to high levels in severe combined immunodeficient mice. Blood 83:2489–2497

Wagner JE, Barker JN, DeFor TE, et al. 2002. Transplantation of unrelated donor umbilical cord blood in 102 patients with malignant and nonmalignant diseases: influence of CD34 cell dose and HLA disparity on treatment-related mortality and survival. Blood 100: 1611–1618.

Wang JF, Wang LJ, Wu YF et al. 2004. Mesenchymal stem/progenitor cells in human umbilical cord blood as support for ex vivo expansion of CD34+ hematopoietic stem cells and for chondrogenic differentiation. Haematologica 89:837-844

Wang TY, Brennan JK, Wu JHD. 1995. Multilineal hematopoiesis in a three-dimensional murine long-term bone marrow culture. Exp Hematol 23:26–32

79

Westerman KA, Leboulch. 1996.Reversible immortalization of mammalian cells mediated by retroviral transfer and site-specific recombination. Proc Natl Acad Sci 93: 8971-8976

Williams EC, Janmey PA, Ferry JD, Mosher DF. 1982. Conformational states of fibronectin. Effects of pH, ionic strength and collagen binding. J Biol Chem 257:14973-14978

Williams WJ, Beutler E, Erslev AJ, Lichtman MA.1990. Hematology 4th edition. New York: McGraw-Hill Inc. neutropenia in baboons. Blood 90(10) (suppl 1):92a

Won JH, Cho SD, Park SK, et al. 2000. Thrombopoietin is synergistic with other cytokines for expansion of cord blood progenitor cells. J Hematother Stem Cell Res 9:465–473

Zandstra PW, Petzer AL, Eaves CJ, Piret JM. 1997. Cellular determinants affecting the rate of cytokine depletion in cultures of human hematopoietic cells. Biotech Bioeng 54:58–66

Zhang CC, Kaba M, Ge G, Xie K, Tong W, Hug C, Lodish HF. Angiopoietin-like proteins stimulate ex vivo expansion of hematopoietic stem cells. Nat Med. 2006 Jan 22; [Epub ahead of print]

Zhang J, Niu C, Ye L at al.2003. Identification of the haematopoietic stem cell niche and control of the niche size. Nature.425:836-841

Zuckerman KS, Wicha MS. 1983. Extracellular matrix production by the adherent cells of long-term murine bone marrow cultures. Blood 61:540-547

80