controlling cell density by micropatterning regulates smad ... · this report demonstrates ......

112
Controlling Cell Density by Micropatterning Regulates Smad Signalling and Mesendoderm Differentiation of Human Embryonic Stem Cells by Lawrence Lee A thesis submitted in conformity with the requirements for the degree of Master of Applied Science Graduate Department of the Institute of Biomaterials and Biomedical Engineering University of Toronto © Copyright by Lawrence Lee 2008

Upload: dinhdan

Post on 07-Apr-2018

219 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

Controlling Cell Density by Micropatterning Regulates Smad Signalling and Mesendoderm Differentiation of

Human Embryonic Stem Cells

by

Lawrence Lee

A thesis submitted in conformity with the requirements for the degree of Master of Applied Science

Graduate Department of the Institute of Biomaterials and Biomedical Engineering

University of Toronto

© Copyright by Lawrence Lee 2008

Page 2: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

ii

Controlling Cell Density by Micropatterning Regulates Smad Signalling

and Mesendoderm Differentiation of Human Embryonic Stem Cells

Lawrence Lee

Master of Applied Science

Graduate Department of the Institute of Biomaterials and Biomedical Engineering

University of Toronto

2008

Abstract

Human embryonic stem cells (hESC) present a potentially unlimited supply of hematopoietic

progenitors for cell-based therapies. However, current protocols for generating these progenitors

typically also generate undesired cell types due to imprecise control of the hESC

microenvironment and poor understanding of the signalling networks regulating mesoderm

differentiation (the germ layer from which hematopoietic cells emerge). This report demonstrates

that activation of the downstream effectors of Activin/Nodal and bone morphogenetic protein

(BMP) signalling (Smad2 (composite of Sma (smaller) and Mad (mothers against

decapentaplegic) and Smad1, respectively) are both required for mesoderm differentiation. It is

further shown that microcontact printing-mediated control of hESC colony size creates local

microenvironments that guide differentiation, via a Smad1-dependent mechanism, preferentially

towards the mesoderm lineage. These findings demonstrate the need for precise control of the

microenvironment in order to effectively guide hESC differentiation to produce specific cell

types for potential therapeutic applications.

Page 3: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

iii

Acknowledgements I would like to acknowledge Dr. Chirag Joshi for his invaluable advice and initial training on

generating hematopoietic cells and quantitative gene expression analysis. I would also like to

thank Dr. Mark Ungrin for his help in generating microwells and spinEBs. Special thanks to

Manuel Alvarez and Ting Yin for providing passage after passage of perfectly cultured hESCs. I

also want to acknowledge Weijia Wang for her assistance with the flow cytometry analysis. I

would like to express my gratitude to Simon Smuckler and Margot Arntfield for their advice and

providing key reagents with the endoderm differentiation. I would like to express my gratitude to

Raheem Peerani, but if I did this section would be far too long. To put it simply, he has taught

me everything I know. Finally, I would like to thank my supervisor, Dr. Peter Zandstra, for his

guidance and support, and providing such a great environment to learn – also for making my

thesis defense happen on such short notice.

Page 4: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

iv

Table of Contents

ABSTRACT .................................................................................................................... II

ACKNOWLEDGEMENTS ............................................................................................. III

TABLE OF CONTENTS ................................................................................................ IV

LIST OF TABLES......................................................................................................... VII

LIST OF FIGURES ...................................................................................................... VIII

ABBREVIATIONS ......................................................................................................... XI

MATHEMATICAL MODEL PARAMETERS ................................................................ XIII

1 INTRODUCTION ........................................................................................................ 1

1.1 Motivation .............................................................................................................................. 1

1.2 Background............................................................................................................................ 2 1.2.1 Key Developmental Signalling Pathways........................................................................ 2

1.2.1.1 Transforming Growth Factor-β Signalling .............................................................. 3 1.2.1.2 Wnt Signalling ......................................................................................................... 4 1.2.1.3 Fibroblast Growth Factor Signalling........................................................................ 5

1.2.2 Gastrulation, Hematopoietic and Pancreatic Development in vivo................................ 5 1.2.2.1 Gastrulation.............................................................................................................. 6 1.2.2.2 Hematopoiesis .......................................................................................................... 7 1.2.2.3 Pancreatic Development .......................................................................................... 9

1.2.3 Embryonic Stem Cells.................................................................................................... 10 1.2.3.1 Primitive Streak-like Development from Embryonic Stem Cells.......................... 11

1.2.3.1.1 Mesendoderm- and Primitive Streak-like Precursors ..................................... 11 1.2.3.2 Mechanisms Governing Mesoderm and Endoderm Differentiation ...................... 12

1.2.3.2.1 Foundations in the Xenopus Model ................................................................ 12 1.2.3.2.2 Extending into the Mouse Embryonic Stem Cell ........................................... 13 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells.............................................. 14

1.2.3.3 Other Mechanisms Regulating Embryonic Stem Cell Differentiation .................. 15 1.2.3.3.1 Cell-cell and Extra-cellular Matrix Mediated Interactions ............................. 15 1.2.3.3.2 Niche-mediated Control of Embryonic Stem Cell Differentiation ................. 16

1.3 Research Objectives ............................................................................................................ 17

1.4 Hypothesis ............................................................................................................................ 17

2 SPECIFIC AIM #1: INDUCTION OF MESENDODERM/PRIMITIVE STREAK-LIKE DEVELOPMENT FROM HESC .................................................................................... 19

Page 5: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

v

2.1 Overview .............................................................................................................................. 19

2.2 Materials and Methods ....................................................................................................... 19 2.2.1 Human Embryonic Stem Cell Culture and Differentiation............................................ 19 2.2.2 Immunofluorescence ...................................................................................................... 21 2.2.3 Quantitative Immunofluorescence and ‘Neighbours Analysis’ ..................................... 21 2.2.4 Mathematical Modeling................................................................................................. 23 2.2.5 Quantitative Gene Expression Analysis......................................................................... 24

2.3 Results .................................................................................................................................. 26 2.3.1 Brachyury Expressing Mesoderm Cells Have High Smad2 Signalling......................... 26 2.3.2 Activin A and BMP2 Synergize to Maximize Mesoderm Differentiation ...................... 28 2.3.3 The Combination of BMP2 and Activin A Induces Both Mesoderm and Endoderm

Precursors but Not Other Lineages............................................................................... 34 2.3.4 Wnt Signalling Does Not Significantly Affect Mesoderm Differentiation in the Presence

of Activin/Nodal and BMP Signalling ........................................................................... 39 2.3.5 Local Cell Density Correlates with Smad1 Signalling and Mesendoderm Differentiation

40

2.4 Discussion............................................................................................................................. 45

3 SPECIFIC AIM #2: MAXIMIZING MESODERM DEVELOPMENT FROM MIXED MESENDODERM POPULATION................................................................................. 50

3.1 Overview .............................................................................................................................. 50

3.2 Materials and Methods ....................................................................................................... 51 3.2.1 Creation of Micropatterned Substrate........................................................................... 51 3.2.2 Seeding of hESC onto Micropatterned Substrate .......................................................... 52 3.2.3 Flow Cytometry Analysis............................................................................................... 53

3.3 Results .................................................................................................................................. 53 3.3.1 Higher Cell Densities Cause Increased Brachyury Expression .................................... 53 3.3.2 Larger Colonies Have Greater Smad1 Signalling......................................................... 56 3.3.3 Smaller Colonies Express Greater Levels of BMP Antagonists.................................... 57

3.4 Discussion............................................................................................................................. 58

4 SPECIFIC AIM #3: DIFFERENTIATION OF MESODERM AND ENDODERM TO HEMATOPOIETIC PROGENITORS AND PRIMITIVE GUT TUBE.............................. 61

4.1 Overview .............................................................................................................................. 61

4.2 Materials and Methods ....................................................................................................... 62 4.2.1 Fabrication of Microwells ............................................................................................. 62 4.2.2 Generation of Forced Aggregate Embryoid Bodies ...................................................... 63 4.2.3 Flow Cytometry ............................................................................................................. 64 4.2.4 Hematopoietic Colony Forming Assay.......................................................................... 64 4.2.5 Primitive Gut Endoderm Differentiation ....................................................................... 65

Page 6: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

vi

4.3 Results .................................................................................................................................. 66 4.3.1 Larger, Mesoderm Enriched Colonies Demonstrate Greater Hematopoietic Potential66 4.3.2 Smaller, Endoderm Enriched Colonies Demonstrate Greater Primitive Gut Endoderm

Potential......................................................................................................................... 69

4.4 Discussion............................................................................................................................. 73

5 CONCLUSIONS ....................................................................................................... 77

6 FUTURE WORK....................................................................................................... 79

7 REFERENCES ......................................................................................................... 82

8 APPENDIX ............................................................................................................... 95

8.1 Other Signalling Pathways Possibly Regulating Mesendoderm Differentiation........... 95 8.1.1 Effect of Rho Kinase Inhibitor Y-27632 on Mesendoderm Differentiation ................... 95 8.1.2 Effect of Fibroblast Growth Factor Inhibition on Mesendoderm Differentiation......... 96

Page 7: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

vii

List of Tables Table 2-1 List of primer sequences used for quantitative gene expression analysis.................... 25 Table 2-2 Table of parameter estimates and associated errors and statistical significance for

quadratic model (Eq. 2-1) ..................................................................................................... 32 Table 2-3 Table of parameter estimates and associated errors and significance for simplified

model (Eq. 2-2) ..................................................................................................................... 33

Page 8: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

viii

List of Figures Figure 1-1 Schematic diagram of day 6 embryo showing emergence of primitive streak and

signalling gradient (adapted from (Pfister et al. 2007)).......................................................... 7 Figure 2-1 Example of Target Activation Assay algorithm. Mask is drawn around the nucleus of

each cell, and the mask is applied to all other channels to calculate average nuclear fluorescent intensity of desired channel................................................................................ 22

Figure 2-2 The nuclear mask (Figure 2-1) is used to calculate the (x, y) coordinates of the centroid of each cell. For every cell in a well, the Neighbours Analysis algorithm iteratively calculates the number of cells within a 200 µm radius for that cell...................................... 23

Figure 2-3 Immunofluorescent images showing Bry expression and Smad2 nuclear localization. Panel a) BMP2 induces the expression of Bry but Bry expressing cells also show nuclear localization of Smad2. Panel b) Inhibition of Smad2 signalling using SB431532 (TGFbi) blocks nuclear localization of Smad2 and Bry expression. Panel c) Activin A alone induces nuclear localization of Smad2 but no Bry expression. Original magnification is 40×, scale bar represents 100 µm........................................................................................................... 26

Figure 2-4 Quantitative immunofluorescence shows that Bry expressing (+ve) cells are characterized by greater levels of nuclear Smad2................................................................. 27

Figure 2-5 Quantitative gene expression analysis reveals that treatment with BMP2 induces the expression of Nodal. ............................................................................................................. 28

Figure 2-6 Dose-response relationship between BMP2 and Activin A concentrations on Bry expression. ............................................................................................................................ 30

Figure 2-7 Analysis of Bry protein (a) and gene expression (b) at various time points following treatment with BMP2 and Activin A showing that Bry expression peaks at 48 hours......... 31

Figure 2-8 Plot of experimental values and predicted values of Bry expression using the simplified quadratic model (Eq. 2-2) .................................................................................... 33

Figure 2-9 Plot of residuals as a function of predicted values for simplified quadratic model (Eq. 2-2)........................................................................................................................................ 34

Figure 2-10 Immunofluorescent images of Sox17 and Bry expression. Original magnification is 40×, scale bar represents 100 µm.......................................................................................... 36

Figure 2-11 Quantitative gene expression analysis showing expression of mesoderm- and endoderm-associated transcripts. .......................................................................................... 36

Figure 2-12 Quantitative immunofluorescent scatter plot showing that neither Bry or Sox17 are coexpressed following 48 hours of treatment with BMP2 and Activin A............................ 37

Figure 2-13 Oct4 and Sox2 flow cytometry analysis of undifferentiated hESC and cultures treated with BMP2 and Activin A for 48 hours. Cells treated with secondary antibodies only were used as negative control. .............................................................................................. 38

Figure 2-14 Quantitative gene expression analysis shows minimal expression of trophectoderm- (CDX2), neural- (Sox2), or extraembryonic endoderm- (Sox7) associated transcripts......... 39

Figure 2-15 Frequency of Bry expression following 48 hours of treatment with BMP2 and Activin, supplemented with Wnt3a, or BIO. ........................................................................ 40

Figure 2-16 Plot of Bry and Sox17 expression frequency as a function of local cell density. (*) indicates significance (p<0.05) over second adjacent bin. ................................................... 41

Figure 2-17 Chloropleth map showing spatial location of Sox17 and Bry expressing cells. Orange/red colour indicates protein expression, blue indicates no protein expression. ....... 42

Figure 2-18 Plot of nuclear pSmad1 intensity as a function of local cell density. (*) indicates p < 0.05........................................................................................................................................ 43

Page 9: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

ix

Figure 2-19 Chloropleth map of nuclear pSmad1 intensity showing higher intensity in areas of greater local cell density. Orange/red colour indicates high intensity, blue indicates low intensity................................................................................................................................. 44

Figure 2-20 Smad1 and Smad2 activation kinetics following treatment with BMP2 and Activin A............................................................................................................................................ 44

Figure 2-21 Quantitative immunofluorescence shows that Bry expressing cells are characterized by higher levels of nuclear pSmad1. (*) indicates p<0.05.................................................... 45

Figure 3-1 Overview of microcontact printing. A) PDMS stamps are inked with Matrigel™; b) Excess Matrigel™ is rinsed away and dried; c) Matrigel™ coated stamp is pressed onto tissue-culture substrate; d) Stamp and substrate are incubated at 85% humidity then stamp is removed transferring Matrigel™ protein onto substrate; e) Single cell suspension is placed on micropatterned substrate and cultured overnight; f) Non-attached cells are rinsed away................................................................................................................................................ 52

Figure 3-2 Phase contrast image of micropatterned colonies of indicated diameter. .................. 54 Figure 3-3 Histogram of local cell density for 200 and 1200 µm micropatterned cultures......... 55 Figure 3-4 Quantitative gene expression analysis of micropatterned cultures following 48 hours

of treatment with BMP2 and Activin A. (*) indicates significance (p<0.05) compared to 200 µm colonies........................................................................................................................... 56

Figure 3-5 Flow cytometry analysis of Bry and KDR expression of micropatterned cultures. (*) indicates significance (p<0.05) compared to 200 µm colonies. ........................................... 56

Figure 3-6 Quantitative immunofluorescence of nuclear pSmad1 intensity of micropatterned cultures. (*) indicates p<0.05................................................................................................ 57

Figure 4-1 Schematic diagram of microwell fabrication. ............................................................ 63 Figure 4-2 Schematic diagram of microwell spinEB formation. a) Micropatterned cultures are

enzymatically dissociated into single cells and pipetted onto microwells; b) Microwells are centrifuged to pellet cells into bottom of microwells and cultured overnight to form embryoid bodies (EB); c) EBs are then removed from the microwells and cultured in suspension. ............................................................................................................................ 64

Figure 4-3 Flow cytometry analysis shows that EB cultures derived from 1200 µm colonies express markers of hematopoietic differentiation at a greater frequency compared to 200 µm colonies. (*) indicates significance (p<0.05). ....................................................................... 67

Figure 4-4 Hematopoietic colony forming assay shows that EB cultures derived from 1200 µm colonies have a greater frequency of colony forming units. (*) indicates significance (p<0.05)................................................................................................................................. 68

Figure 4-5 Representative phase contrast images of hematopoietic colonies showing CFU-GM (a), CFU-M (b) and CFU-GEMM (c, d). Original magnification is 10× and 4×, scale bar represents 200 µm................................................................................................................. 69

Figure 4-6 Flow cytometry analysis of CXCR4 expression of attached cultures derived from 200 and 1200 µm colonies following 2 days of culture in Activin A. Red histogram is of isotype control, green histogram represents sample. (*) indicates p<0.05........................................ 71

Figure 4-7 Fluorescent images of cultures derived from 200 and 1200 µm colonies following endoderm differentiaiton assay showing HNF1β, FoxA2, and HNF4apha staining. Nuclei are shown in blue. Scale bar represents 100 µm; original magnification is 20× and 40×. ... 72

Figure 4-8 Quantitative gene expression analysis of primitive gut tube and pancreatic endoderm cultures derived from micropatterned cultures. a) Cultures derived from smaller 200 µm colonies show greater expression of primitive gut tube-associated genes when compared to 1200 µm colonies; b) Expression of pancreatic endoderm-associated genes is also greater in cultures derived from 200 µm colonies, but considerably less when compared to human

Page 10: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

x

pancreatic tissue. (*) and (**) indicate significance (p<0.05 and p<0.01, respectively) when comparing 200 and 1200 µm colony derived cultures.......................................................... 73

Figure 5-1 Proposed scheme for the regulation of mesendoderm differentiation by local cell density and Smad signalling ................................................................................................. 78

Figure 8-1 Representative immunofluorescent images of Bry expression of micropatterned colonies following 48 hours of treatment with BMP2 and Activin A with or without Y-27632 ROCK inhibitor. Original magnification is 10×, scale bar indicates 500 µm. .......... 96

Figure 8-2 Representative immunofluorescent images of Bry expression following 48 hours of treatment with BMP2, Activin A, FGFR inhibitor PD173074 (20 nM), or MAP kinase inhibitor PD184352 (500 nM) as indicated. Original magnfication is 10×, scale bar indicates 500 µm. ................................................................................................................................. 97

Figure 8-3 Quantitative gene expression analysis of micropatterned colonies following 48 hours of treatment with BMP2, Activin A, and FGFR inhibitor PD173074 (20 nM) as indicated. (*) indicates p<0.05. ............................................................................................................. 98

Figure 8-4 Quantitative immunofluorescent analysis of pSmad1 intensity of micropatterned colonies following 90 minute treatment with BMP2, Activin A, and MAP kinase inhibitor PD184352 (500 nM) as indicated. (*) indicates p<0.05. ...................................................... 99

Figure 8-5 Frequency of hematopoietic colonies following hematopoietic differentiation assay of 1200 µm cultures with or without FGFR1 inhibition. .......................................................... 99

Page 11: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

xi

Abbreviations ActRIIB Activin receptor type II B AGM Aorta-gonad-mesonephros ALK Activin receptor-like kinase APC Allophycocyanin bFGF Basic fibroblast growth factor BIO Bromoindirubin oxime BME β-mercaptoethanol BMP Bone morphogenetic protein BMPRII BMP receptor type II Bry Brachyury BSA Bovine serum albumin cDNA Complementary deoxyribonucleic acid CDX2 Caudal type homeobox 2 Cer1 Cerberus 1 CFU Colony forming unit CFU-GEMM CFU-granylocyte, erythroid, macrophage, megakaryocyte CFU-GM CFU-granulocyte, macrophage CFU-M CFU-macrophage Co-Smad Common Smad CXCR4 Chemokine (C-X-C motif) receptor 4 DKK1 Dickkopf 1 DMEM Dubelcco’s modified Eagles’s medium E Embryonic day EB Embryoid body ECM Extra-cellular matrix EDTA Ethylene diamine tetra-acetic acid ESC Embryonic stem cell ExE Extraembryonic Endoderm FAST-1 Forkhead Activin signal transducer-1 FBS Fetal bovine serum FGF Fibroblast growth factor FITC Flluorescent Isothiocyanate Flk1 Fetal liver kinase 1 FoxA2 Forkhead box A2 FoxH1 Forkhead box H1 G-CSF Granulocytic colony stimulating factor GDF Growth and differentiation factor GFR Growth factor reduced GSC Goosecoid GSK3 Glycogen synthase kinase 3 GUSB Glucuronidase beta hESC Human embryonic stem cell HNF1β Hepatocyte nuclear factor 1β HNF3β Hepatocyte nuclear factor 3β HNF4α Hepatocyte nuclear factor 4α HSC Hematopoietic stem cell ICM Inner cell mass

Page 12: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

xii

IL-3 Interleukin-3 IL-6 Interleukin-6 IPF1 Insulin promoter factor-1 KDR Kinase insert domain receptor KGF Keratinocyte growth factor KO Knockout LEF1 Lymphoid enhancer binding factor 1 Lefty1 Left-right determination factor 1 LTC-IC Long-term culture-initiating cell MAP Mitogen-activated protein mEF Mouse embryonic feeders mESC Mouse embryonic stem cell Mesp1 Mesoderm posterior 1 MIXL1 Mix 1 homeobox-like 1 MNX1 Motor neuron and pancreas homeobox-1 NEAA Nonessential amino acids NOD/SCID Nonobese diabetic/sever combined immunodeficiency OAZ Olf-1/EBF associated zing finger Oct4 Octamer-4 Pax6 Paired box 6 PBS Phosphate buffered solution PDGFRa1 Platelet derived growth factor receptor alpha 1 PDMS Poly(dimethylsiloxane) PDX1 Pancreas/duodenum homeobox-1 PE Phycoerythrin PECAM1 Platelet endoethelial cell adhesion molecule 1 pSmad Phosphorylated Smad RA Retinoic Acid rh Recombinant human RNA Ribonucleic Acid ROCK Rho kinase R-Smad Receptor smad SCF Stem cell factor Smad Composite of Sma (smaller, Caenorhabditis Elegans) and Mad (mothers

against decapentaplegic, Drosophila Melanogaster) Sox17 SRY-box containing gene 17 Sox2 SRY-box containing gene 2 Sox7 SRY-box containing gene 7 SR Serum replacement TCF T-cell factor TGFβ Transforming growth factor β TβRI TGFβ receptor type I TβRII TGFβ receptor type II UV Ultraviolet VEGF Vascular endothelial growth factor VEGFR2 Vascular endothelial growth factor receptor 2 Wnt Wingless-related mouse mammary tumour virus integration site XTC-MIF Xenopus cell line XTC mesoderm inducing factor

Page 13: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

xiii

Mathematical Model Parameters y Percent of cells expressing Brachyury c1 Intercept term c2 BMP2 main effect coefficient c3 Activin A main effect coefficient c4 BMP2 quadratic coefficient c5 Activin A quadratic coefficient c6 Second order interaction coefficient

Page 14: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

1

1 Introduction

1.1 Motivation Currently, a variety of blood and immune disorders can be treated through transplantation of

hematopoietic stem cells (HSC) from a healthy donor. Such HSC can be extracted from the bone

marrow (Thomas et al. 1970), peripheral blood (Korbling et al. 1981), or umbilical cord blood

(Gluckman et al. 1989; Goldberg et al. 2007) obtained immediately following childbirth. These

treatments, however, are restricted by the limited supply of donors and the requirement of human

leukocyte antigen matching. Alternatively, human embryonic stem cells (hESC) have the

potential to form all cell types present in an adult body, and therefore present a potentially

unlimited supply of HSC that can be used for transplantation. Additionally, recent studies have

shown the ability to induce pluripotency from human somatic cells (Takahashi et al. 2007; Yu et

al. 2007). These induced pluripotent stem (iPS) cells offer a patient specific cell source for cell

based therapies and would obviate concerns of immune rejection. However, much remains to be

studied in the embryonic stem cell system before further advances can be made. Current

protocols for generating hESC derived HSC include attached coculture of hESC with various

hemogenic stromal cell lines or three-dimensional embryoid body differentiation in the presence

of serum or a cocktail of cytokines (Kaufman et al. 2001; Chadwick et al. 2003; Vodyanik et al.

2005; Zambidis et al. 2005). These approaches are limited by their undefined culture medium or

underlying mechanism. Other challenges include the purity of cell types that are produced from

hESC and the sheer number of HSC that would be required for a therapeutic application in

humans.

Page 15: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

2

In order to overcome these challenges, a greater understanding of the mechanisms underlying

hESC fate decisions is required. A key player in these mechanisms is the interaction of hESC

and their microenvironment, or ‘niche.’ This stem cell niche includes cell-cell contact, secreted

and/or exogenously added cytokines and extracellular protein interactions, all of which play

pivotal roles in determining hESC cell fate. In order to precisely control hESC differentiation so

that the desired cell type with sufficiently high purity can be obtained, the hESC niche must also

be controlled during the early stages of differentiation. To achieve this goal, a combination of

exogenous cytokines will be added to hESC cultures while simultaneously controlling hESC

colony size. This will allow for the robust control of cell-cell contact and signalling in order to

guide hESC differentiation towards the hematopoietic lineage.

1.2 Background

1.2.1 Key Developmental Signalling Pathways

In order to develop an full understanding of embryonic stem cells and how to best make use of

their potential, it is often useful to first discuss the biology of a developing embryo and fetus.

During the first several days of an implanting embryo, roughly 4 and 7 days following

fertilization in mice and humans, respectively, a handful of developmental signalling pathways

are responsible for its proper development and tissue patterning. The most important pathways

include transforming growth factor-β (TGFβ) signalling, Wnt signalling, and fibroblast growth

factor (FGF) signalling, the basic elements of which will be briefly discussed here prior to

discussing embryogenesis and embryonic stem cells.

Page 16: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

3

1.2.1.1 Transforming Growth Factor-β Signalling

Transforming growth factor-β (TGFβ) signalling has been shown to be important in early

mammalian embryogenesis and patterning, as well as key regulators of embryonic stem cell

(ESC) self-renewal and differentiation (Chu et al. 2004; Peerani et al. 2007; Smith et al. 2008).

The TGFβ superfamily can be divided into two subclasses of ligands. The first includes the

TGFβs, Activins and Nodals while the second class includes the BMP (bone morphogenetic

protein) and GDF (growth and differentiation factor) ligands. The variety of TGFβ ligands (the

human genome encodes for at least 29 different ligands (Feng et al. 2005)) is accommodated by

various permutations of the TGFβ signalling complex, which consists of two type II and two

type I receptors. In the absence of ligands, type I and type II receptors are found as homodimers

on the cell surface. However, when ligand is present homodimers of each type I and type II

receptors form a complex with the ligand which allows for phosphorylation of a conserved

Glycine/Serine-rich sequence in the type II receptor and leads to autophosphorylation of the type

I receptor (Derynck et al. 2003; Shi et al. 2003). TGFβ1 and Activins/Nodals bind to TβRII

(TGFβ receptor type II) and ActRIIB (Activin receptor type II B) type II receptors, respectively,

and type I receptors TβRI (also known as ALK5, Activin receptor-like kinase 5) and ALK4/7,

respectively. This leads to phosphorylation of the receptor Smads (R-Smads, composite of Sma

(smaller, Caenorhabditis Elegans) and Mad (mothers against decapentaplegic, Drosophila

Melanogaster)) 2 and 3 by the type I receptor. BMPs, on the other hand, such as BMP2, 4 and 7

bind to BMPRII (BMP receptor type II) type II receptor and ALK3/6 type I receptor that leads to

phosphorylation of R-Smads 1, 5 or 8. In either case, the phosphorylated R-Smads form a

complex with a common Smad (co-Smad), Smad4 in vertebrates, which translocates to the

nucleus where it interacts with transcription factors to initiate or suppress transcription.

Reviewed in Feng et al. (2005).

Page 17: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

4

1.2.1.2 Wnt Signalling

Wnt (wingless-related mouse mammary tumour virus integration site) signalling also plays a

crucial role in early embryo axis formation and mice with Wnt3 null mutations fail to form early

germ layers (Liu et al. 1999). As many as 19 Wnt mammalian homologues have been identified

and play key developmental roles in gastrulation and further tissue patterning. Wnt genes encode

secreted cysteine-rich signalling proteins that regulate cell processes such as proliferation,

differentiation and movement. The most studied Wnt signalling pathway is the canonical

pathway that acts primarily through the intracellular signalling molecule β-catenin. In the

absence of Wnt ligand, β-catenin is complexed with Axin and is phosphorylated by casein kinase

Iα (CKIα) and glycogen synthase kinase 3β (GSK3β) destining β-catenin for ubiquitination and

degradation. However, when Wnt ligand is present the Wnt protein forms a signalling complex

with the Frizzled (Fz) family of receptors and LRP (low density lipoprotein receptor-related

protein) 5 and LRP6 coreceptors. This leads to the association of Dishevelled (Dvl) with the

Frizzled receptor and ultimately inhibits the phosphorylation of β-catenin by GSK3β. This allows

for the nuclear accumulation of β-catenin where it associates with the TCF/LEF (T-cell

factor/lymphoid enhancer factor) family of transcription factors leading to transcriptional

activation. Reviewed in Katoh (2007) and Nusse (2008).

The Wnt pathway also interacts with TGFβ through GSK3β. It has been shown that the receptor

Smads contain a region, termed the linker region, that is susceptible to phosphorylation by

activated GSK3β. This prevents its nuclear accumulation and targets it for degradation

(Fuentealba et al. 2007). Through activation of the Wnt receptors and inhibition of GSK3β, Wnt

pathway activation is able to prolong BMP signalling and Smad1 activation.

Page 18: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

5

1.2.1.3 Fibroblast Growth Factor Signalling

Fibroblast growth factor (FGF) signalling has also been shown to play a central role in regulating

embryogenesis. The cells of a pre- and post-implantation embryo express FGF4 (Niswander et

al. 1992) and FGF2 is commonly used to maintain human embryonic stem cells in an

undifferentiated state (Thomson et al. 1998; Xu et al. 2005). Fibroblast growth factor are soluble

proteins that signal through transmembrane receptor tyrosine kinases (RTK). Most RTKs are

monomeric but binding of the ligand to the extracellular domain induces dimerization of the

receptor-ligand complex. In the unbound, unstimulated state, the intrinsic kinase activity of the

RTK is very low, however, once dimerized the RTK is activated causing phosphorylation of the

cytosolic tyrosine residues. The phosphorylation of these residues leads to the activation of a

membrane-bound protein called Ras. Activated Ras then leads to a cascade of protein kinases

causing the activation of the kinase MAP (mitogen activated protein). Activated MAP kinases

then form dimers which translocate to the nucleus to activate various transcription factors

(Lodish et al. 2004). Additionally, it has been shown the FGF signalling pathway interacts with

the TGFβ pathway through MAP kinase in a very similar fashion as GSK3β mentioned above.

MAP kinases are able to phosphorylate receptor Smads in the linker region which prevents their

nuclear accumulation and transcriptional activity (Kretzschmar et al. 1997). In this way, FGF

signalling is able to regulate the TGFβ pathway to control its activity.

1.2.2 Gastrulation, Hematopoietic and Pancreatic Development in vivo

With an introduction to the signalling pathways responsible embryogenesis and proper tissue

patterning, the early stages of development of the mammalian embryo will now be discussed

with a particular emphasis on the tissues that give rise to the hematopoietic system and

pancreatic organs.

Page 19: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

6

1.2.2.1 Gastrulation

In the post-implantation embryo, gastrulation marks one of the earliest morphological changes

when tissue formation and germ layer development begins. Starting at roughly embryonic day 6

(E6) of the developing mouse embryo the three primary germ layers (ectoderm, endoderm and

mesoderm), from which all somatic cell types are derived, begin to emerge. The ectoderm gives

rise to neural tissues such as the central nervous system as well as the outer epithelium of the

body. The endoderm gives rise to the gut such as the liver, lungs and pancreas, while the

mesoderm gives rise to skeletal muscle, bones, and the hematopoietic system. This tissue

patterning is the result of morphogenetic signals secreted by the epiblast and the surrounding

extraembryonic tissue. The epiblast secretes Nodal, while the proximal extraembryonic

endoderm (ExE) secretes bone morphogenetic proteins (BMP), and Wnt ligands. In contrast, the

distal ExE, and later the anterior ExE, secretes antagonists to these morphogens including

Cerberus1, Lefty1 (left-right determination factor 1), and DKK1 (dickkopf 1) (Pfister et al.

2007). This creates a signalling gradient along the proximal/distal axis that leads to the

development and distal expansion of the primitive streak starting at the proximal-posterior region

of the embryonic cup (Brennan et al. 2001; Lowe et al. 2001; Vincent et al. 2003). It is in the

primitive streak where both the definitive endoderm and mesoderm emerge and this region is

marked by the expression of T-Brachyury (T-Bry), Goosecoid (GSC) and Mixl1 (Mix 1 like

homeobox 1). Reviewed in Pfister et al. (2007). At roughly E7.5 when the primitive streak

reaches the distal tip of the egg cylinder, a distinct morphological object, known as the node,

becomes apparent. This structure has a crucial role in organizing and patterning the midline axis

of the embryo (Nagy 2003).

Page 20: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

7

Figure 1-1 Schematic diagram of day 6 embryo showing emergence of primitive streak and signalling gradient (adapted from (Pfister et al. 2007))

1.2.2.2 Hematopoiesis

During mouse development hematopoiesis is initiated independently at two distinct sites: 1) the

extraembryonic yolk sac; and 2) the aorta-gonad-mesonephros (AGM) region in the embryo

proper. Extraembryonic, also known as primitive, hematopoiesis first emerges from clusters of

primitive mesoderm in the extraembryonic area at E8. These clusters, known as yolk sac blood

islands, give rise to both endothelial and hematopoietic cells leading researchers to postulate the

existence of a bipotential ‘hemangioblast’ giving rise to both endothelial and hematopoietic

lineages (Sabin 1920; Murray 1932). This has since been confirmed using both mouse and

human ESC models (Choi et al. 1998; Nishikawa et al. 1998; Kennedy et al. 2007; Zambidis et

al. 2007). This extraembryonic hematopoiesis, however, is characterized by predominantly

nucleated erythroblasts expressing strictly embryonic and fetal hemoglobins (Barker 1968;

Brotherton et al. 1979), the lack of lymphoid progenitors, and the inability to repopulate adult

mice. Reviewed in Tavian et al. (2005) and Zambidis et al. (2006). This transient wave of

Page 21: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

8

hematopoiesis is thought to provide a temporary hematopoietic system for the developing

embryo.

The appearance of HSC with the ability to generate complex hematopoietic progenitors

(myeloid, lymphoid, erythroid) and to repopulate adult mice first appears in the AGM region of

the E9 mouse embryo proper (Medvinsky et al. 1993; Tavian et al. 2001). During gastrulation,

mesoderm cells in the posterior region of the primitive streak migrate laterally out of the

primitive streak to form the splanchnopleura and dorsal aorta. It is in this region, the AGM,

where embryonic hematopoietic cells first emerge. It has been shown that this embryonic source

of hematopoiesis, known as definitive hematopoiesis, also has its origins in a bipotential

hemogenic endothelium lining the wall of the dorsal aorta (Dieterlen-Lievre et al. 1994; Jaffredo

et al. 1998; Nishikawa et al. 1998).

Signalling gradients and morphogens have also been described in the Xenopus embryo. A

Xenopus homologue of Activin A, XTC-MIF (Xenopus cell line XTC mesoderm inducing

factor) (Savage et al. 1989; Green et al. 1990; Green et al. 1990; Smith et al. 1990), has been

shown to induce ventral mesoderm, dorsal mesoderm, and neural tissue at low, moderate and

high concentrations, respectively suggesting an instructive action of XTC-MIF. Similar Xenopus

studies have also shown a role for BMP4 to induce hemogenic mesoderm from early mesodermal

precursors during gastrulation (Dale et al. 1992; Maeno et al. 1996; Huber et al. 1998). These

studies show that BMP4 acts following gastrulation to preferentially induce ventral hemogenic

mesoderm rather than axial tissue. The hemogenic role of BMP4 appears to be conserved in

humans where it has been found that BMP4 is expressed at high levels in the AGM, specifically

in regions of densely packed cells underlying intra-aortic hematopoietic clusters suggesting its

direct role in specifying hematopoietic cells from mesodermal precursors (Marshall et al. 2000).

Page 22: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

9

Other signalling proteins have also been identified as inducers of hemogenic mesoderm. Mouse

embryos with homozygous mutations for Flk1 (fetal liver kinase 1), which encodes for a receptor

of vascular endothelial growth factor (VEGF) (Millauer et al. 1993), undergo gastrulation but die

at E9 due to defects in primitive hematopoietic and endothelial differentiation. Moreover, when

Flk1-/- mESC are made into chimeric embryos, they are unable to contribute to definitive

hematopoiesis demonstrating that the gene is required for both types of hematopoiesis (Shalaby

et al. 1995), suggesting its importance in not only vascularization but also hematopoiesis.

A variety of other hemogenic cytokines have also been identified as maintaining or expanding

HSC in culture including Flt3-ligand, stem cell factor (SCF), granulocytic colony stimulating

factor (G-CSF), interleukin-3 (IL-3) and IL-6 (Bhatia et al. 1997; Conneally et al. 1997; Brugger

et al. 2000; Zheng et al. 2003). These factors expand the number of long-term culture-initiating

cells (LTC-IC) and improve their engraftment ability in subletheally irradiated nonobese

diabetic/severe combined immunodeficiency (NOD/SCID) mice models. Due to their positive

effects on somatic HSC expansion, they have also been applied to deriving hematopoietic

lineages from hESC (Chadwick et al. 2003) but a later report has shown that the addition of G-

SCF, IL-6, and IL-3 are not required for hemogenic induction from hESC (Pick et al. 2007).

1.2.2.3 Pancreatic Development

During gastrulation, the definitive (or embryonic) endoderm originates from the anterior region

of the primitive streak between E6 and E6.5 (Lawson et al. 1986). The endoderm precursors

migrate through the primitive streak and intercalate with the overlying extraembryonic

endoderm. Following gastrulation, gut endoderm appears as a sheet of cells stretching from the

primitive streak to the anterior region of the embryonic cup. This flat sheet then elongates and

Page 23: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

10

forms a primitive gut tube along which the different endoderm lineages are derived. The liver

emerges from the most anterior portion of the primitive gut tube while the most posterior region

forms the hindgut from which with the large intestine and colon are formed. In between the

anterior and posterior regions lies the primitive foregut and midgut, which give rise to the

stomach, duodenum, and pancreas. Reviewed in Wells et al. (1999). The entire primitive gut tube

is marked by the expression of hepatocyte nuclear factor-1 β (HNF1β) (Coffinier et al. 1999),

FoxA2 (forkhead box A2, or HNF3β) (Sasaki et al. 1993), and HNF4α (Duncan et al. 1994). At

this time pancreatic development requires retinoic acid signalling as well as the inhibition of

hedgehog signalling (Kim et al. 1998; Hebrok et al. 2000; Stafford et al. 2004). The developing

pancreatic epithelium expresses hepatocyte nuclear factor-6 (HNF6) (Rausa et al. 1997), insulin

promoter factor-1 (IPF1, or pancreas/duodenum homeobox-1, PDX1) (Wilson et al. 2003), and

motor neuron and pancreas homeobox 1 (MNX1) (Wilson et al. 2003), which are important

regulators of pancreatic development.

1.2.3 Embryonic Stem Cells

Embryonic stem cells (ESC) are the in vitro equivalent of the inner cell mass (ICM) of a

blastocyst stage embryo and were first derived from mice (Evans et al. 1981; Martin 1981) and

researchers have since isolated ESC from other vertebrates including humans (Thomson et al.

1998) and other primates. Although ESC are removed from their native embryonic environment,

they nonetheless retain the capacity to develop into all cell types found in an adult, as

demonstrated by chimera (Bradley et al. 1984) and tetraploid embryo complementation (Nagy et

al. 1993) experiments where ESC are reintroduced into a developing embryo. This unique ability

coupled with their capacity to expand and self-renew indefinitely gives ESC great potential for

cell-based regenerative medicine applications. The commitment of embryonic stem cells from a

Page 24: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

11

pluripotent state to more restricted cell types, a process known as differentiation, will now be

discussed as it relates to the in vivo model discussed above.

1.2.3.1 Primitive Streak-like Development from Embryonic Stem Cells

1.2.3.1.1 Mesendoderm- and Primitive Streak-like Precursors

A bipotential mesendoderm precursor capable of giving rise to both endoderm and mesoderm

lineages has been well characterized in the nematode worm, Caenorhabditis Elegans (Sulston et

al. 1983), as well as the frog, Xenopus Laevis (Rodaway et al. 2001), but its existence in

mammalian species is more contentious. Several attempts have been made using mouse

embryonic stem cells but have fallen short of definitively proving the existence of a

developmentally restricted single cell capable of giving rise to both endoderm and mesoderm

lineages. Tada and colleagues inserted a green fluorescent protein (GFP) reporter into the

Goosecoid (GSC) gene locus, an early marker of mesoderm and endoderm differentiation (Norris

et al. 2002), to monitor mesendoderm differentiation (Tada et al. 2005). Using Activin A, a

protein closely related to Nodal that signals through the same set of receptors and downstream

effectors, they were able to induce GSC expression and show that a population expressing GFP-

goosecoid, epithelial-cadherin, and platelet-derived growth factor receptor (PDGFR) α1 is

capable of giving rise to mesoderm and endoderm lineages. Clonogenic single cell assays,

however, were limited to the measurement of early transcription factor and cell surface marker

expression and no functional output. Another report also attempted to generate developmentally

restricted mesendodermal clonal cell lines through sustained activation of the Wnt pathway

(Bakre et al. 2007). The functional assays shown in the study, however, are restricted to the

mesoderm lineage and do not show production of definitive endoderm. These results

Page 25: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

12

demonstrate that although a bipotential mesendoderm precursor has been shown to exist in lower

order species there is no conclusive evidence thus far to prove its existence in mammals.

1.2.3.2 Mechanisms Governing Mesoderm and Endoderm Differentiation

Regardless of the existence of a mesendoderm precursor, the molecular mechanism that govern

whether a pluripotent, or restricted multipotent, cell will become endoderm or mesoderm fated

remains poorly understood. However, what is known in several in vivo and in vitro embryonic

stem cell models will be discussed here.

1.2.3.2.1 Foundations in the Xenopus Model

It has been shown that the direct action of Activin/Nodal signalling on mesoderm and endoderm

differentiation is the result of an Activin responsive element contained in the promoter region of

the Mix family of homeobox genes in the Xenopus embryo (Huang et al. 1995; Chen et al. 1996).

It was determined that the downstream effector of Activin signalling, XSmad2 (Xenopus

homologue of Smad2), forms a complex with a transcriptional a transcriptional partner named

forkhead Activin signal transducer-1 (FAST-1), and later renamed FoxH1 (forkhead box H1). Its

homologue was subsequently identified in mammalian species and further shown that in mice

that Smad2, along with Smad4, forms a complex with FoxH1 that binds directly to the MIXL1

(Mix1 homeobox-like 1) promoter region and induces its transcription (Hart et al. 2005).

However, FoxH1 can also act as a negative regulator of MIXL1 expression by recruiting

Goosecoid to the FoxH1/Smad2/Smad4 complex which suppresses the expression of MIXL1 to

act as a negative feedback loop (Izzi et al. 2007). Other genes required for mesoderm and

endoderm differentiation such as Goosecoid and Lim1 also contain Activin responsive elements

further demonstrating the role of Activin/Nodal signalling (Labbe et al. 1998; Watanabe et al.

2002).

Page 26: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

13

Although Smad1 and Smad2 are structurally very similar, the region within Smad2 responsible

for binding with FoxH1 is different in Smad1 such that it does not interact with FoxH1 nor does

it induce MIXL1 expression (Attisano et al. 2001; Hart et al. 2005). Despite this, Smad1

activation through BMP signalling is required for primitive streak development and mesoderm

differentiation (Winnier et al. 1995; Fujiwara et al. 2002; Rivera-Perez et al. 2005). This can be

explained by reports describing an analogous BMP responsive element contained in the promoter

regions of genes required for ventral patterning and mesoderm formation in the Xenopus embryo.

Moreover, BMP signalling involves a transcriptional coactivator termed Olf-1/EBF associated

zing finger (OAZ) that interacts with XSmad1, but not XSmad2, to initiate transcription (Hata et

al. 2000). Direct targets of Smad1 signalling include Xvent-2b, a homeobox gene involved in

ventral patterning of the Xenopus embryo (Henningfeld et al. 2000) as well as the Xenopus

Brachyury homologue, Xbra. These results demonstrate the role of Smad1 activation through

BMP signalling in ventral patterning and mesoderm formation in the Xenopus embryo.

1.2.3.2.2 Extending into the Mouse Embryonic Stem Cell

TGFβ and Smad signalling have also been shown to induce endoderm and mesoderm

differentiation in mammalian species. Using mouse embryonic stem cells is found that high

levels of Activin/Nodal signalling specifies definitive endoderm differentiation while low levels

of Activin/Nodal or BMP signalling specifies mesoderm differentiation (Johansson et al. 1995;

Kubo et al. 2004; Ng et al. 2005; Tada et al. 2005; Gadue et al. 2006; Nostro et al. 2008; Shiraki

et al. 2008). This is similar to the Xenopus model described above where Smad2 activation

targets primarily endoderm-related genes while Smad1 activation targets mesoderm genes. This

suggests a balance between Smad2 and Smad1 activation where Smad2 induces anterior cell

Page 27: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

14

fates while Smad1 has the effect of posteriorizing primitive streak-like cells. This model is

supported by the observation that mESC derived definitive endoderm precursors are

characterized by high levels of phosphorylated (p)Smad2 and low levels of pSmad1 while

mesoderm precursors are characterized by high levels of pSmad1 (Shiraki et al. 2008).

The involvement of Wnt signalling, as well as Smad1 and Smad2 activation, in endoderm and

mesoderm development has also been suggested through studies using mouse embryonic stem

cells. Several studies have shown that Wnt signalling is required for the induction of a primitive

streak-like phenotype (Gadue et al. 2006; Nostro et al. 2008). The implication of Wnt signalling

in primitive streak development is supported by studies showing that the β-catenin-LEF-1/TCF

transcription factor complex promotes Nodal expression during gastrulation in the Xenopus

embryo and in mESC (McKendry et al. 1997; Gadue et al. 2006). However, inhibition of Wnt

signalling in the presence of Activin/Nodal signalling blocks mesoderm and endoderm

differentiation suggesting key targets of Wnt signalling in primitive streak development. This is

supported by studies using mouse embryos and mESC models showing that T-Bry is a direct

target of the Wnt-β-catenin signalling pathway (Yamaguchi et al. 1999; Arnold et al. 2000).

Moreoever, it has been shown that high concentrations of Wnt3a alone is able to induce

mesoderm differentiation, but little to no endoderm (Gadue et al. 2006) – an effect that can be

explained by the deactivation of GSK3β, a known antagonist of Smad1 signalling (Fuentealba et

al. 2007).

1.2.3.2.3 From Mouse to Human Embryonic Stem Cells

Despite that mesoderm and endoderm differentiation is mouse embryonic stem and other animal

models is fairly well described, there is a significant lack of understanding when it comes to

human embryonic stem cells. A detailed analysis of Smad signalling in hESC with respect to

Page 28: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

15

mesoderm and endoderm differentiation has yet to be performed. Moreover, despite that many

developmental mechanisms are conserved amongst mouse and human, an important distinction

has been observed whereby Activin/Nodal signalling alone is insufficient to induce mesoderm

differentiation from hESC. Sustained treatment with Activin A, regardless of concentration does

not induce mesoderm differentiation and primarily results in endoderm differentiation at higher

concentrations (D'Amour et al. 2005; D'Amour et al. 2006; McLean et al. 2007; Kroon et al.

2008). This suggests that a greater activation of Smad1 is required in hESC for mesoderm

differentiation, or greater basal activation of Smad1 is present in mESC. Also, although Wnt

signalling has been found to play key roles in mesoderm development of mESC, there are no

such reports to date implicating its role in hESC. An improved understanding of not only Smad

signalling, but also other developmental signalling pathways is required in order to fully exploit

the potential of human embryonic stem cells. By understanding these mechanisms, embryonic

stem cell differentiation can be more precisely controlled in order to generate specific cell types

and avoid the generation of undesired cell types which would otherwise need to be removed

through cell sorting techniques such as fluorescent or magnetic activated cell sorting.

1.2.3.3 Other Mechanisms Regulating Embryonic Stem Cell Differentiation

1.2.3.3.1 Cell-cell and Extra-cellular Matrix Mediated Interactions

Factors other than soluble ligands have also been implicated in ESC derived hematopoiesis. It

has been shown that hematopoietic development from mESC, without the support of a stromal

layer, is completely inhibited when cultured as an attached colony during the first 3 days of

differentiation but is not impaired if cultured initially in suspension and then reattached (Dang et

al. 2002). This suggests a cell-cell contact or extra-cellular matrix- (ECM) adhesion mediated

process required only for early hemogenic mesoderm induction and not for subsequent

Page 29: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

16

hematopoietic specification and expansion. This also suggests that stromal coculture

differentiation acts in part by fulfilling this cell-contact/adhesion step.

1.2.3.3.2 Niche-mediated Control of Embryonic Stem Cell Differentiation

The importance of the stem cell niche on self-renewal and differentiation of both mESC and

hESC has been previously reported (Davey et al. 2006; Peerani et al. 2007). These reports

describe how the radial organization of ESC coupled with auto/paracrine signalling loops affect

cell fate in the absence or withdrawal of growth factors. Cells near the centre of colonies and at a

higher local cell density show increased pluripotency due to the secretion of self-renewal factors

while cells at lower densities secrete excess differentiating factors leading to loss of

pluripotency. It is also shown that the balance of self-renewal and differentiation can be

controlled through the use of micropatterning techniques to independently control colony size

(Peerani et al. 2007; Peerani In Press). Further data (Peerani, unpublished) also shows that

control of colony size can be used to dictate the fate of differentiation hESC where larger

colonies assume a neural fate while smaller colonies predominantly differentiate into

extraembryonic endoderm but a small proportion show primitive streak-like phenotype.

Moreover, it has also been shown that spatial patterns can affect the proliferation of endothelial

cells through a process mediated by mechanical stresses (Nelson et al. 2005). These reports

highlight the importance of spatial organization and cell density in the stem cell niche as well as

the ability to control differentiation by manipulating the niche in the absence of exogenous

growth factors.

Page 30: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

17

1.3 Research Objectives

The overall objective of this project is to optimize conditions allowing for hematopoietic

differentiation of hESC. However, the differentiation of ES cell to hematopoietic progenitor is a

multifaceted process involving complex signalling interactions governing sequential cell lineage

specification. Therefore, this project has been subdivided into three specific aims:

• Specific Aim 1: Induction of mesendoderm/primitive streak-like development from

hESC using a combination of BMP2 and Activin A

• Specific Aim 2: Optimization of conditions allowing for mesoderm specification from

the mixed mesendoderm precursors

• Specific Aim 3: Differentiation of mesoderm precursors towards the hematopoietic

lineage

The deconstruction of the complex process of hematopoiesis from hESC allows for identifying

signalling process involved during the step-wise differentiation of hESC similar to the ontogeny

of hematopoietic stem cells in vivo.

1.4 Hypothesis

It is hypothesized that both exogenous and endogenous signalling factors can influence the

differentiation of hESC towards mesoderm and hemogenic mesoderm lineages. Specifically,

Activin/Nodal and BMP signalling have been shown to be crucial in the development of

mesoderm lineages in not only in vivo but also in vitro ESC models. Due to the observation that

signalling gradients are responsible for patterning early mouse embryo, it is anticipated that

different concentrations of exogenously added Activin/Nodal and BMP signalling ligands will

lead to different cell fates. Moreover, it is also believed that the endogenous secretion of agonists

or antagonists to the TGFβ pathway, and possibly others, in response to the added cytokines will

also play a central role in fate decision. By controlling the colony size of hESC, one can dictate

Page 31: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

18

the level of not only exogenous but also endogenous signalling to optimize the proportion of

mesoderm fated cells. In addition, controlling colony size decreases heterogeneity by

maintaining constant local cell density and endogenous signalling effects within a given sample.

Furthermore, it is hypothesized that this mesoderm fated population can be differentiated further

towards hemogenic mesoderm through the addition of hemogenic cytokines through three-

dimensional embryoid body differentiation. It is anticipated that this optimal hemogenic

mesoderm population will correlate with a maximum yield of hematopoietic progenitors.

Page 32: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

19

2 Specific Aim #1: Induction of Mesendoderm/Primitive

Streak-like Development from hESC

2.1 Overview

Using embryogenesis as a model, this objective takes the first step in developing definitive

hematopoiesis from human embryonic stem cells. Embryonic stem cells represent the in vitro

analog of the inner cell mass of the blastocyst stage embryo. Prior to mesoderm formation in the

mouse embryo, the primitive ectoderm, or epiblast, migrates through the primitive streak under

the influence of an Activin/Nodal, BMP and Wnt signalling gradient. With this model as a guide,

this objective is to use a combination of BMP2 and Activin A to induce primitive streak-like

development from hESC. Although studies from mouse embryos have shown the activity of

BMP4 in primitive streak induction, BMP2 and BMP4 have been found to be functionally

interchangeable (Massague 1990; Sampath et al. 1993; Lyons et al. 1995; Zhang et al. 1996).

The endpoint of this objective is to determine the optimal concentration of both Activin A and

BMP2 to maximize primitive streak-like development.

2.2 Materials and Methods

2.2.1 Human Embryonic Stem Cell Culture and Differentiation Human embryonic stem cell lines H9 (Technion – Israeli Institute of Technology), HES2

(WiCell) and I6 (Technion – Israeli Institute of Technology) were used for all experiments

described in this thesis. H9, HES2, and I6 human ES cell lines were maintained on irradiated

mouse embryonic feeder (mEF) cells plated at a density of 20,000 cells per cm2. H9 and I6

human ES cell lines were maintained in serum-free medium consisting of knockout (KO)-

Dubelcco’s modified Eagle’s medium (DMEM, Invitrogen) and 20% KO-serum replacement

Page 33: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

20

(SR, Invitrogen) supplemented with 4ng/mL recombinant human (rh) basic fibroblast growth

factor (bFGF, PeproTech). Cells were dissociated into clumps using 0.1 % collagenase IV

(Invitrogen) and passaged at a ratio of 1:6 every 5 to 6 days. The HES2 human embryonic cell

line was maintained in serum-free medium consisting of DMEM-F12 (Invitrogen) supplemented

with 2 mM L-Glutamine (Invitrogen), 50 u/mL Penicillin/Streptomycin (Invitrogen), 20% KO-

SR, 1 × nonessential amino acids (NEAA, Invitrogen), 0.1 µM β-mercaptoethanol (BME,

Sigma), 5% mEF conditioned medium, and 20 ng/mL bFGF. HES2 cells were dissociated into

single cells using 0.25% Trypsin-EDTA (Ethylene diamine tetra-acetic acide, Cellgro) and

passaged at a ratio of 1:6 every 4 to 5 days.

In order to remove mEFs prior to differentiation cells were passaged as clumps onto tissue-

culture treated plastic coated with growth factor reduced Matrigel™ (GFR-MG, 1:30, BD

Biosciences) in defined serum-free medium (XV-FAT) consisting of X-VIVO10™ medium

(Cambrex) supplemented with 2 mM GlutaMAX™ (Invitrogen), 1× nonessential amino acids

(NEAA, Invitrogen), 0.1 µM β-mercaptoethanol (BME, Sigma), 40 ng/mL bFGF, 0.1 ng/mL rh-

transforming growth factor β-1 (TGFβ1, R&D Systems), and 5 ng/mL rh-Activin A (R&D

Systems). Following overnight seeding in XV-FAT maintenance medium, medium was replaced

with X-VIVO10™ medium supplemented with 2 mM GlutaMAX™ (Invitrogen), 1×

nonessential amino acids (NEAA, Invitrogen), 0.1 µM β-mercaptoethanol (BME, Sigma), and

indicated cytokines. Recombinant human bone morphogenetic protein-2 (BMP2, R&D Systems)

and rhActivin A (R&D Systems) and rhWnt3a (R&D Systems) were used at 50 ng/mL unless

otherwise indicated. TGFβ inhibitor SB431542 (Sigma) was used at a concentration of 10 nM.

Page 34: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

21

2.2.2 Immunofluorescence Cells were fixed in 3.7% para-formaldehyde in phosphate buffered solution (PBS, Invitrogen) for

15 minutes at room temperature. Following fixation, cells were blocked and permeabilized for 45

minutes at room temperature with PBS containing 0.1% Triton X-100, 10% donkey serum

(Jackson ImmunoResearch) and 1% BSA (bovine serum albumin, Sigma). Cells were then rinsed

in PBS three times and incubated overnight at 4°C with primary antibody at indicated dilution in

PBS containing 10% fetal bovine serum (FBS, Gibco). Following primary antibody incubation,

cells were rinsed three times with PBS and incubated at room temperature for 90 minutes with

the appropriate secondary antibody and Hoechst 33342 nuclear dye (1 µg/mL, Invitrogen). Goat

anti-human Brachyury antibody (R&D Systems) was used at 0.5 µg/mL, rabbit anti-phospho-

Smad1/5/8 antibody (Cell Signaling Technologies) was used at 1:200 dilution, mouse anti

Smad2/3 antibody (BD Biosciences) was used at 1:200 dilution, mouse anti-human Sox17 (R&D

Systems) was used at 10 µg/mL, rabbit anti-FoxA2 (Abcam) was used at 1:4000 dilution, goat

anti-HNF1β (Santa Cruz Biotechnology) was used at a dilution of 1:100, goat anti-HNF4α (Santa

Cruz Biotechnology) was used at a dilution of 1:100. Primary antibodies were counterstained

with Alexa Fluor® 488 donkey anti-goat IgG secondary antibody (Invitrogen, 1:200),

AlexaFluor® 647 donkey anti-rabbit IgG secondary antibody (1:200), or AlexaFluor® 555

donkey anti-mouse IgG secondary antibody (1:200). Fluorescent images were taken with an

AxioCam MRm (Zeiss) digital fluorescent camera connected to an Axio Observer.D1 (Zeiss)

fluorescent microscope.

2.2.3 Quantitative Immunofluorescence and ‘Neighbours Analysis’ Quantitative image analysis was performed using the ArrayScan® VTI imaging platform and the

Target Activation assay algorithm. The imaging platform captures successive fluorescent images

Page 35: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

22

in order to image an entire area of a well of a 96-well plate. The Target Activation assay

algorithm uses the Hoechst nuclear stain to identify and draw a mask surrounding the nuclei of

each cell. The average fluorescent intensity of each channel is then calculated for the area

encompassed by the nuclear mask (Figure 2-1). The spatial coordinates (x, y) of the centroid of

each nucleus are also recorded (Figure 2-2). Positive gating for cells defined as expressing a

given protein is established based on negative controls stained with secondary antibody only.

Figure 2-1 Example of Target Activation Assay algorithm. Mask is drawn around the nucleus of each cell, and the mask is applied to all other channels to calculate average nuclear fluorescent intensity of desired channel

Page 36: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

23

x Coordinate (µm)

0 100 200 300 400 500

y C

oord

inat

e (µ

m)

0

100

200

300

400

500

r = 200 µm

Figure 2-2 The nuclear mask (Figure 2-1) is used to calculate the (x, y) coordinates of the centroid of each cell. For every cell in a well, the Neighbours Analysis algorithm iteratively calculates the number of cells within a 200 µm radius for that cell

An algorithm, termed ‘Neighbours Analysis’ was performed using the spatial information and

fluorescent intensities gathered from quantitative immunofluorescence and is originally

described by Peerani and colleagues (Peerani et al. 2007). The algorithm iteratively calculates for

each cell within a given well the total number of neighbouring cells within a 200 µm radius

(Figure 2-2). A radius of 200 µm was chosen because it is the largest radius for which a

significant trend in Smad signalling or Brachyury expression is observed.

2.2.4 Mathematical Modeling Fitting the empirical data of Bry expression frequency as a function of BMP2 and Activin A

concentration was performed using JMP IN software (version 3.2.6, http://www.jmp.com). The

Page 37: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

24

data was initially fitted using a quadratic model, which includes all main-effect coefficients,

quadratic coefficients, and second-order interaction coefficients. All terms that were found to be

insignificant (p>0.05) were removed from the model, the data was fitted again to the revised

model, and the process was repeated until only significant terms remained.

2.2.5 Quantitative Gene Expression Analysis Total ribonucleic acid (RNA) was extracted from cells using TRIzol® reagent (Invitrogen) and

purified using the RNeasy® Mini RNA isolation kit (Qiagen). Complementary deoxyribonucleic

acid (cDNA) was synthesized from purified RNA using the SuperScript™ III Reverse

Transcriptase kit. cDNA synthesized without reverse transcriptase was used as a negative

control. Real time quantification of transcripts using polymerase chain reaction was performed

using Power SYBR® Green PCR Master Mix (Applied Biosystems) and a 7900HT Fast Real-

Time PCR System (Applied Biosystems). Both β-actin and GUSB (glucuronidase beta) were

used as endogenous controls and yielded very similar results, but the transcript levels presented

here were calculated as 2Δ(ΔCt) normalized to β-actin. The list of primer sequences used is listed

in Table 2-1. Total RNA isolated from human fetal lung (Biochain), human fetal brain

(Biochain), and Caco-2 human colorectal adenocarcinoma cell line (American Type Culture

Collection) were used as positive controls.

Transcript Name Sequence β-Actin – forward 5’-CATGTACGTTGCTATCCAGGC β-Actin – reverse 5’-CTCCTTAATGTCACGCACGAT GUSB – forward 5’-GTCTGCGGCATTTTGTCGG GUSB – reverse 5’-ATGGCGATAGTGATTCGGAGC Oct4 – forward 5’-TGGGCTCGAGAAGGATGTG Oct4 – reverse 5’-GCATAGTCGCTGCTTGATCG T-Bry – forward 5’-TGCTTCCCTGAGACCCAGTT T-Bry – reverse 5’-GATCACTTCTTTCCTTTGCATCAAG MIXL1 – forward 5’-CCGAGTCCAGGATCCAGGTA

Page 38: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

25

MIXL1 – reverse 5’-CTCTGACGCCGAGACTTGG GSC – forward 5’-GAGGAGAAAGTGGAGGTCTGGTT GSC – reverse 5’-CTCTGATGAGGACCGCTTCTG KDR – forward 5’-ACTTTGGAAGACAGAACCAAATTATCTC KDR – reverse 5’-TGGGCACCATTCCACCA Nodal – forward 5’-CCGAGGGCAGACATCATCC Nodal – reverse 5’-CCATCCACTGCCACATCTTCT Wnt3 – forward 5’-CCACAACACGAGGACGGAGAAG Wnt3 – reverse 5’-TCCACAGGCGAGTTGGGTCTG Cer1 – forward 5’-ACAGTGCCCTTCAGCCAGACT Cer1 – reverse 5’-ACAACTACTTTTTCACAGCCTTCGT Sox17 – forward 5’-GGCGCAGCAGAATCCAGA Sox17 – reverse 5’-CCACGACTTGCCCAGCAT CDX2 – forward 5’-GGGCTCTCTGAGAGGCAGGT CDX2 – reverse 5’-CCTTTGCTCTGCGGTTCTG FoxA2 – forward 5’-GGGAGCGGTGAAGATGGA FoxA2 – reverse 5’-TCATGTTGCTCACGGAGGAGTA PDGFRa1 – forward 5’-AAATTGTGTCCACCGTGATCTG PDGFRa1 - reverse 5’-CGACACATAGTTCGAATCATGCA Lefty1 – forward 5’-GTGAGCCCTGAATTTGCTTC Lefty1 – reverse 5’-TGGCCAAAGATTCTCATTCC Mesp1 – forward 5’-GCTCTGTTGGAGACCTGGAT Mesp1 – reverse 5’-GTCTGCCAAGGAACCACTTC Pax6 – forward 5’-ATGTGTGAGTAAAATTCTGGGCA Pax6 – reverse 5’-GCTTACAACTTCTGGAGTCGCTA Sox7 – forward 5’-ACGCCGAGCTCAGCAAGAT Sox7 – reverse 5’-TCCACGTACGGCCTCTTCTG HNF1β – forward 5’-CAAGGGCACCCCTATGAAGA HNF1β – reverse 5’-CGGAGGATCTCTCGTTGCTT HNF4α – forward 5’-AGGCCACAATCTCCTCTCAC HNF4α – reverse 5’-TTGCTGGGGATTATGGTGGGA IPF1 – forward 5’-CTGGATTGGCGTTGTTTGTG IPF1 – reverse 5’-TCCCAAGGTGGAGTGCTGTAG MNX1 – forward 5’-ACCAGTTCAAGCTCAACAAGTACCT MNX1 – reverse 5’-TGCTGCGTTTCCATTTCATC HNF6 – forward 5’-CTGCGCAACCCCAAACC HNF6 – reverse 5’-TCCACATCCTCCGGAAGGT

Table 2-1 List of primer sequences used for quantitative gene expression analysis.

Page 39: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

26

2.3 Results

2.3.1 Brachyury Expressing Mesoderm Cells Have High Smad2 Signalling As BMP4 is well known to induce mesoderm differentiation of mouse and human ESC, the

effect of BMP2 was investigated to examine if the mesoderm inducing effect is specific to

BMP4. It was found that 48 hours of treatment with 50 ng/mL of BMP2 is effective at inducing

the expression of Bry (Figure 2-3a), an established marker of early mesoderm differentiation

(Rivera-Perez et al. 2005), suggesting that their common effector, Smad1, is responsible for

mesoderm differentiation. Interestingly it is found that Bry expressing cells are also

characterized by high levels of Smad2 nuclear localization indicating active Smad2 signalling

(Figure 2-3a and Figure 2-4).

Figure 2-3 Immunofluorescent images showing Bry expression and Smad2 nuclear localization. Panel a) BMP2 induces the expression of Bry but Bry expressing cells also show nuclear localization of Smad2. Panel b) Inhibition of Smad2 signalling using SB431532 (TGFbi) blocks nuclear localization of Smad2 and Bry expression. Panel c) Activin A alone induces nuclear localization of Smad2 but no Bry expression. Original magnification is 40×, scale bar represents 100 µm.

Page 40: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

27

Bry(-ve

)

Bry(+v

e)R

elat

ive

Fluo

resc

ent I

nten

sity

0.6

0.8

1.0

1.2

1.4

1.6NuclearSmad2

*

Figure 2-4 Quantitative immunofluorescence shows that Bry expressing (+ve) cells are characterized by greater levels of nuclear Smad2.

Although BMP2 is not believed to directly activate Smad2 signalling, treatment with BMP2

induces the expression of Nodal, an endogenously secreted ligand that is an activating ligand of

Smad2 (Figure 2-5). This suggests that Smad2 is activated indirectly through the endogenous

expression of Nodal which in turn activates ALK4/7 and triggers nuclear translocation of Smad2.

To investigate if the mesoderm inducing effect of BMPs is dependent on Smad2 signalling,

hESC were treated with 50 ng/mL of BMP2 and SB431532 (TGFbi), a specific inhibitor of

ALK4, ALK5, and ALK7, but has no effect on the BMP receptors ALK1, ALK2, ALK3, ALK6

(Inman et al. 2002; Laping et al. 2002). Treatment with TGFbi completely blocks nuclear

accumulation of Smad2 as well as Bry expression (Figure 2-3b). This demonstrates that Smad2

signalling is required for mesoderm differentiation. To examine if Smad2 signalling alone is

sufficient to induce mesoderm differentiation, hESC were treated with 0, 5, 25 or 50 ng/mL

Activin A, a Smad2 activator, for 48 hours. A range of concentrations was tested due to previous

observations that moderate concentrations are required for mesoderm differentiation of mESC.

Despite the fact that Activin A alone effectively induces nuclear accumulation of Smad2, it is not

Page 41: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

28

sufficient to promote Bry expression (Figure 2-3c). Together, these findings show that both

Smad1 and Smad2 activation are required for mesoderm differentiation.

hESC

No Cyto

kines

BMP2

Fold

Incr

ease

Exp

ress

ion

0

1

2

3

4

5

6

7

Nodal

Figure 2-5 Quantitative gene expression analysis reveals that treatment with BMP2 induces the expression of Nodal.

2.3.2 Activin A and BMP2 Synergize to Maximize Mesoderm Differentiation Due to the finding that both Smad1 and Smad2 activation are required for mesoderm

differentiation, combinations of different concentrations of BMP2 and Activin A were tested to

determine if supplementing endogenously expressed Nodal with Activin A would further

increase mesoderm differentiation. An initial dose-response using BMP2 and Activin A on the

effect of Brachyury expression was determined using a high-throughput 96-well platform. The

concentration of Activin A and BMP2 were independently varied from 0 to 50 ng/mL in X-

VIVO 10TM serum-free medium. Brachyury protein expression was measured through

quantitative immunofluorescence using the CellomicsTM ArrayScanV platform and Target

Activation algorithm. This method allows for automated high-throughput analysis of different

Page 42: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

29

samples using standard culture-ware. An alternative to quantitative immunofluorescence would

be flow-cytometry, but would typically require 10 times the number of cells for a reliable

measurement. An additional advantage of the ArrayScanV platform is that the spatial coordinates

of each cell are also measured. This allows for the computation of geometric relationships

between various cell types and the effects of local cell density (Davey et al. 2006; Peerani et al.

2007), which would be otherwise lost in a flow-cytometric analysis.

It was found that the frequency of Brachyury expressing cells increases with Activin A and

BMP2 concentration in a dose dependent manner following 48 hours of treatment. The highest

concentrations of Activin A and BMP2 tested (both 50 ng/mL) yielded the highest percentage of

Brachyury expressing cells, 25.6 ± 0.9 % (Figure 2-6). As described above, it is also found that

treatment with Activin A alone, regardless of concentration, resulted in minimal Brachyury

expression, confirming previous reports (D'Amour et al. 2005; McLean et al. 2007) and

demonstrating that Activin/Nodal signalling alone is not sufficient for mesoderm differentiation

of hESC. Instead it is found that BMP signalling is also required and that it synergizes with

Activin/Nodal to induce mesoderm differentiation. This is in agreement with the mouse embryo

model where the primitive streak emerges from the proximal-posterior region of the embryonic

cup, which also corresponds to the highest level of Nodal and BMP4 expression.

Page 43: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

30

0

5

10

15

20

25

30

0 5

25 50

0 10

25 50

% B

ry P

ositi

ve

[Activin A] (ng/mL)[BMP2] (ng/mL)

Figure 2-6 Dose-response relationship between BMP2 and Activin A concentrations on Bry expression.

Further analysis was also performed to determine if the 48-hour time point chosen is the optimal

time to measure mesoderm differentiation. Quantitative immunofluorescence and quantitative

gene expression analysis was used to measure Bry expression following 12, 24, 48, 72 and 120

hours of treatment with Activin A and BMP2. These experiments showed that Bry expression is

first detected at 12 hours, peaks at 48 hours, and declines thereafter (Figure 2-7) in agreement

with previous studies (Gadue et al. 2006; McLean et al. 2007). This confirms that, under the

conditions tested here, the 48 hours is an appropriate time point for analyzing early mesoderm

differentiation.

Page 44: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

31

12 hr

s24

hrs48

hrs

% B

ry p

ositi

ve

0

5

10

15

20

25

30

hESC

48 hr

s72

hrs

120 h

rs

Rel

ativ

e E

xpre

ssio

n

0

5

10

15

20

25

30Bry

a b

Figure 2-7 Analysis of Bry protein (a) and gene expression (b) at various time points following treatment with BMP2 and Activin A showing that Bry expression peaks at 48 hours.

In order to quantitatively confirm if BMP2 and Activin A synergize, the numerical data were

fitted to a quadratic model (Audet et al. 2002). The initial model is as follows:

]AActivin []BMP2[]AActivin [

]BMP2[]AActivin []BMP2[

62

5

24321

××+×

+×+×+×+=

cc

ccccy Eq. 2-1

where y represents the percent of cells expressing Brachyury, c1 is the intercept term, c2 and c3

are the main effect coefficients of BMP2 and Activin A concentrations, respectively, c4 and c5

are the quadratic coefficients of BMP2 and Activin A concentrations, respectively, and c6 is the

second-order interaction coefficient of BMP2 with Activin A. An initial fit of the expression data

with Eq. 2-1 shows that only the intercept term, BMP2 main effect coefficient, and second-order

interaction coefficient are significant (p<0.05, Table 2-2)

Page 45: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

32

Term Estimate Error pValue C1 3.28 0.91 0.0049 C2 0.17 0.07 0.0488 C3 0.062 0.075 0.4266 C4 0.00070 0.0013 0.6114 C5 -0.0018 0.0014 0.236 C6 0.0054 0.0010 0.0002 R2 Adjusted 0.955447

Table 2-2 Table of parameter estimates and associated errors and statistical significance for quadratic model (Eq. 2-1)

The result that the quadratic terms are not significant suggests that the concentrations of BMP2

and Activin A have not reached saturation and that greater concentrations will induce even

greater expression of Bry. However, the cost of high concentrations of cytokines limits the

regular use of even greater concentrations. Once the insignificant terms were removed the data

was fitted again to Eq. 2-1, but with the terms c3, c4 and c5 removed:

]AActivin []BMP2[]BMP2[ 621 ××+×+= cccy Eq. 2-2

All the terms of the revised model (Eq. 2-2) are significant and both the BMP2 main effect

coefficient and the second-order interaction coefficient are positive demonstrating that Bry

expression increases with increasing BMP2 concentration and that BMP2 and Activin A do in

fact quantitatively synergize. A negative value would be indicative of an inhibitory relationship.

The result that Bry expression increases with BMP2 concentration suggests that increasing levels

of Smad1 signalling increases Bry expression. Moreover, the R2 value does not decrease using

the second model equation despite the removal of parameters. A plot of the predicted values

versus the experimental value shows good agreement between predicted and experimental values

(Figure 2-8). Additionally, a plot of the residuals as a function of the predicted value shows that

the residuals are evenly distributed and the model does not show significant bias (Figure 2-9).

Page 46: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

33

Term Estimate Error pValue C1 2.93 0.53 0.0001 C2 0.22 0.02 <.0001 C6 0.0047 0.0006 <.0001 R2 0.96

Table 2-3 Table of parameter estimates and associated errors and significance for simplified model (Eq. 2-2)

% Bry expressing (Experimental)

0 5 10 15 20 25 30

% B

ry e

xpre

ssin

g (P

redi

cted

)

0

5

10

15

20

25

30

Figure 2-8 Plot of experimental values and predicted values of Bry expression using the simplified quadratic model (Eq. 2-2)

Page 47: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

34

% Bry expressing (Predicted)

0 5 10 15 20 25 30

Res

idua

l

-4

-3

-2

-1

0

1

2

3

Figure 2-9 Plot of residuals as a function of predicted values for simplified quadratic model (Eq. 2-2)

2.3.3 The Combination of BMP2 and Activin A Induces Both Mesoderm and Endoderm Precursors but Not Other Lineages

Although the combination of BMP2 and Activin A efficiently induces mesoderm differentiation,

endoderm precursors also emerge as evidenced by expression of Sox17, a marker of definitive

endoderm precursors (Figure 2-10). Quantitative gene expression analysis was performed to

confirm the immunofluorescent results and additionally characterize the cell population. It was

found that in addition to T-Bry (Brachyury), the expression of other early mesoderm-associated

genes were also upregulated including KDR (kinase insert domain receptor, human homologue

of Flk1, or vascular endothelial growth factor receptor 2, VEGFR2) (Kennedy et al. 2007),

PDGFRa1 (platelet derived growth factor receptor alpha 1) (Davis et al. 2008), and Mesp1

(mesoderm posterior 1) (Saga et al. 1996) (Figure 2-11). However, the expression of endoderm-

Page 48: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

35

associated genes were also upregulated including MIXL1 (Mix1 homeobox-like 1) (Izumi et al.

2007), GSC (Goosecoid) (Perea-Gomez et al. 1999), and Sox17 (SRY-box containing gene 17)

(Kanai-Azuma et al. 2002) (Figure 2-11). The expression of both mesoderm- and endoderm-

associated transcripts suggests the emergence of a mixed population including both mesoderm

and endoderm precursors, reminiscent of the primitive streak, but there is no evidence of a

bipotential mesendoderm precursors. Immunofluorescence and quantitative immunofluorescence

shows that neither Bry nor Sox17 are found to be coexpressed following 48 hours of treatment

with BMP2 and Activin A (Figure 2-10 and Figure 2-12), suggesting that lineage commitment to

either mesoderm or endoderm has taken place. A previous report, however, has shown

coexpression of Bry and Sox17 following 48 hours of treatment with Activin A and low

concentrations of FBS (D'Amour et al. 2005). In the model proposed by D’Amour and

colleagues, Activin is shown to induce Bry expression and the removal of Activin permits

mesoderm differentiation. However, this is in the presence of serum, which is known to contain

BMPs. In order to definitively prove the existence of a mesendoderm precursor, sorting cell

populations and performing clonogenic assays would be required to show that a single cell can

give rise to both endoderm and mesoderm but not endoderm lineages.

Page 49: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

36

Figure 2-10 Immunofluorescent images of Sox17 and Bry expression. Original magnification is 40×, scale bar represents 100 µm.

T-Bry

MIXL1KDR

PDGFRa1

Mesp1

Wnt3 GSC

Sox17

Rel

ativ

e E

xpre

ssio

n

1

10

100

1000hESCNo CytokinesActivin ABMP2BMP2 + Activin A

Figure 2-11 Quantitative gene expression analysis showing expression of mesoderm- and endoderm-associated transcripts.

Page 50: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

37

Figure 2-12 Quantitative immunofluorescent scatter plot showing that neither Bry or Sox17 are coexpressed following 48 hours of treatment with BMP2 and Activin A

The population was further characterized using flow cytometry to assess Oct4 (Octamer-4, or

POU5F1, POU domain class 5 transcription factor 1) and Sox2 (SRY-box containing gene 2)

expression, transcription factors associated with pluripotency (Nichols et al. 1998; Avilion et al.

2003). As expected, cultures of undifferentiated hESC mostly contain cells coexpressing Oct4

and Sox2, indicative of pluripotency (Figure 2-13). However, following treatment with BMP2

and Activin A, Sox2 expression is highly downregulated while Oct4 expression remains high

(Figure 2-13). This suggests that the cells are no longer pluripotent, due to loss of Sox2

expression, but are slowly undergoing differentiation with retention of Oct4. This accounts for

the large proportion of cells that do not express either Bry or Sox17 (Figure 2-12), suggesting

that these cells have lost pluripotency but have not yet expressed markers of mesoderm or

endoderm differentiation.

Page 51: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

38

Figure 2-13 Oct4 and Sox2 flow cytometry analysis of undifferentiated hESC and cultures treated with BMP2 and Activin A for 48 hours. Cells treated with secondary antibodies only were used as negative control.

Quantitative gene expression analysis was also used to assess the differentiation of other

lineages. Minimal differentiation towards neural, extraembryonic endoderm, or trophectoderm

lineages was found as evidenced by low expression of Sox2 (Ellis et al. 2004), Sox7 (Kanai-

Azuma et al. 2002), and CDX2 (caudal type homeobox 2) (Beck et al. 1995), respectively

(Figure 2-14), demonstrating that BMP2 and Activin A together preferentially drives mesoderm

and endoderm differentiation.

Page 52: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

39

hESC

No Cyto

kines

Activin

BMP2

BMP2 + A

ctivin

Caco-2

Rel

ativ

e E

xpre

ssio

n

0

20

40

600

CDX2

hESC

No Cyto

kines

Activin

BMP2

BMP2 + A

ctivin

0.0

0.2

0.4

0.6

0.8

1.0

1.2

1.4

1.6

Sox2

hESC

No Cyto

kines

Activin

BMP2

BMP2 + A

ctivin

Human

Fetal L

ung

0

5

40

50 Sox7

Figure 2-14 Quantitative gene expression analysis shows minimal expression of trophectoderm- (CDX2), neural- (Sox2), or extraembryonic endoderm- (Sox7) associated transcripts.

2.3.4 Wnt Signalling Does Not Significantly Affect Mesoderm Differentiation in the Presence of Activin/Nodal and BMP Signalling

In addition to Nodal the expression of Wnt3 also increased following treatment with BMP2

(Figure 2-11) as previously reported (Nostro et al. 2008; Zhang et al. 2008). However, unlike

Activin A, the addition of exogenous Wnt ligand (Wnt3a, 50 ng/mL) does not significantly

increase Bry expression, nor does the addition of BIO (bromoindirubin oxime, 500 nM) a soluble

GSK3β (glycogen synthase kinase 3β) inhibitor in the presence of Activin A and BMP2 (Figure

2-15). A previous report has also demonstrated that blocking the Wnt pathway using high

concentrations of Dkk1, an inhibitor of Wnt signalling, in the presence of BMP4 does not affect

mesoderm differentiation suggesting that Wnt signalling is dispensable for mesoderm induction

from hESC in the presence of active BMP signalling, contrary to evidence found for mESC

(Nostro et al. 2008).

Page 53: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

40

BMP2+Acti

vin A

+Wnt3

a+B

IO

% P

ositi

ve

0

10

20

30

40

Bry

Figure 2-15 Frequency of Bry expression following 48 hours of treatment with BMP2 and Activin, supplemented with Wnt3a, or BIO.

2.3.5 Local Cell Density Correlates with Smad1 Signalling and Mesendoderm Differentiation

In addition to Bry, Sox17 (a transcription factor marking definitive endoderm precursors (Kanai-

Azuma et al. 2002)) expressing cells are also detected following 48 hours of treatment with

Activin A and BMP2. This demonstrates that both mesoderm and endoderm precursors are

arising in this condition indicative of mesendoderm/primitive streak-like development. From the

analysis of immunofluorescent images, it was observed that cells expressing Bry are typically

found in areas of high cell density while cells expressing Sox17 are typically found in areas of

low cell density. To quantify this observation a previously developed algorithm, termed

‘Neighbours Analysis’ (Peerani et al. 2007), was used in conjunction with the ArrayScan® VTI

(Cellomics) imaging platform to measure the frequency of Bry expressing cells as a function of

Page 54: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

41

the number of cells within a 200 µm radius. The number of neighbouring cells is used a measure

of local cell density. Each cell within a well of a 96-well microplate, was binned according to the

number of cells within a 200 µm radius of that cell (Figure 2-2). The frequency of Bry and

Sox17 expressing cells was calculated for each bin and plotted. This algorithm quantitatively

shows that Bry expressing cells are enriched in areas of high local cell density while Sox17

expressing cells are enriched in areas of low cell density (Figure 2-16). This relationship is

visually confirmed through chloropleth maps with dots representing spatial location of cells and

colour representing Bry or Sox17 protein level. It can be seen that expression of Sox17 is

predominant at lower cell densities at the periphery of colonies while Bry expressing cells are

enriched in areas of higher cell density towards the centre of colonies (Figure 2-17).

# Cells within 200 µm radius

0-50

50-10

0

100-1

50

150-2

00

200-2

50

250-3

00

300-3

50

350-4

00

400-4

50

% P

ositi

ve

010203040506070

BrySox17

**

*

Figure 2-16 Plot of Bry and Sox17 expression frequency as a function of local cell density. (*) indicates significance (p<0.05) over second adjacent bin.

Page 55: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

42

Figure 2-17 Chloropleth map showing spatial location of Sox17 and Bry expressing cells. Orange/red colour indicates protein expression, blue indicates no protein expression.

To investigate a possible mechanism governing this effect a similar algorithm was employed to

measure Smad1 signalling, measured by levels of nuclear phosphorylated Smad1 (pSmad1), as

function of local cell density. Following a 90 minutes pulse with Activin A and BMP2 it was

found that pSmad1 increases with local cell density (Figure 2-18) and is visually confirmed

through a chloropleth map showing higher nuclear pSmad1 levels in areas of high local cell

density (Figure 2-19). The 90 minute time point was chosen because Smad1 and Smad2

signalling peaks after 90 minutes of treatment before decaying to near or below baseline after 48

hours of treatment with BMP2 and Activin A (Figure 2-20). This suggests that a high Smad1

signalling environment is required for mesoderm differentiation whereas low Smad1 is required

for definitive endoderm differentiation. The effect of Smad signalling on mesendoderm

differentiation has been previously described in mESC where Foxa2 expressing definitive

endoderm precursors are characterized by low levels of pSmad1 while Bry expressing mesoderm

precursors are characterized by high levels of pSmad1 (Shiraki et al. 2008). A similar

observation is made here. Although the average nuclear pSmad1 level for all cells has decreased

Page 56: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

43

to near baseline after 48 hours, it is found that Bry positive cells maintain significantly greater

levels of nuclear pSmad1 when compared to the Bry negative population (Figure 2-21)

demonstrating a direct correlation between Smad1 signalling and Bry expression.

# Cells within 200 µm radius

0-50

50-10

0

100-1

50

150-2

00

200-2

50

250-3

00

300-3

50

350-4

00

400-4

50

Rel

ativ

e Fl

uore

scen

t Int

ensi

ty

0.8

1.0

1.2

1.4

1.6

pSmad1

*

*

Figure 2-18 Plot of nuclear pSmad1 intensity as a function of local cell density. (*) indicates p < 0.05.

Page 57: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

44

Figure 2-19 Chloropleth map of nuclear pSmad1 intensity showing higher intensity in areas of greater local cell density. Orange/red colour indicates high intensity, blue indicates low intensity.

Time after pulse (hours)

0 10 20 30 40 50 60

Rel

ativ

e Fl

uore

scen

t Int

ensi

ty

0.6

0.8

1.0

1.2

1.4

1.6

1.8Nuclear pSmad1Nuclear Smad2No treatment

Figure 2-20 Smad1 and Smad2 activation kinetics following treatment with BMP2 and Activin A.

Page 58: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

45

Bry(-ve

)

Bry(+v

e)

Rel

ativ

e Fl

uore

scen

t Int

ensi

ty

0.6

0.8

1.0

1.2

1.4Nuclear pSmad1

*

Figure 2-21 Quantitative immunofluorescence shows that Bry expressing cells are characterized by higher levels of nuclear pSmad1. (*) indicates p<0.05.

2.4 Discussion The results presented here is the first detailed report of the synergistic effects of BMP and

Activin/Nodal signalling in a dose-dependent manner on mesoderm differentiation from human

embryonic stem cells. Previous reports has shown that Activin A alone is able to induce both

mesoderm and endoderm differentiation of mESC, with high concentrations of Activin A

promoting endoderm and medium to low concentrations promoting mesoderm differentiation

(Smith et al. 1990; Johansson et al. 1995; Robertson et al. 2003; Ang et al. 2004; Gadue et al.

2006; McKiernan et al. 2007). Moreover, the combinatorial effect of Activin A and BMP4 on

mesoderm development has also been shown in mESC (Huber et al. 1998; Nostro et al. 2008)

and recently in hESC (Laflamme et al. 2007; Yang et al. 2008). However, this is the first detailed

analysis of the downstream effectors of Activin/Nodal and BMP signalling on mesoderm and

endoderm differentiation in hESC. It is shown here that not only BMP signalling but also Smad2

activation are required for mesoderm differentiation, as evidenced by Bry expression. Moreover,

Page 59: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

46

the addition of an exogenous Activin/Nodal agonist, such as Activin A, increases the mesoderm

inductive effects of BMP signalling. It is also shown that Bry expressing cells are characterized

by greater levels of pSmad1 consistent with the results of Shiraki and colleagues who found

that, in mESC derived populations, Bry expressing cells show high levels of pSmad1 while

FoxA2 expressing endoderm precursors show low levels of pSmad1 (Shiraki et al. 2008). It is

demonstrated here that this mechanism is conserved in hESC. Despite the fact that the average

nuclear pSmad1 level has decayed to near baseline levels after 48 hours of treatment with

Activin A and BMP2, Bry expressing cells maintain a relatively high level of Smad1 signalling

suggesting that duration of BMP signalling also plays a role in mesoderm specification.

On the other hand, in hESC the role of Smad2 activation has been overlooked in the signalling

pathway regulating mesoderm differentiation. Although several groups have identified that

TGFβ signalling through ALK4, 5, and 7 is required for mesoderm and endoderm differentiation

(Gadue et al. 2006; Nostro et al. 2008; Zhang et al. 2008), this is the first report showing that Bry

expressing mesoderm precursors show high levels of Smad2 activation and that the addition of

exogenous Smad2 activating ligands, such as Activin A, increases Bry expression. Similar to

Smad1, Smad2 signalling remains active in Bry expressing cells despite the decay in Smad2

signalling in the remaining cells after 48 hours. This suggests that, in addition to Smad1,

sustained Smad2 signalling is also required for mesoderm differentiation. Despite the previous

associations of high Activin A concentrations with endoderm differentiation protocols of mouse

and human ESC we find that, in the presence of BMP signalling, Activin A increases mesoderm

differentiation. It is believed that Smad1 signalling has the effect of posteriorizing cell fates of

hESC, driving differentiation of hESC away from anterior endoderm towards a mesoderm fate, a

phenomenon that has been observed in mESC (Nostro et al. 2008). Without sufficiently high

levels of Smad1 activation hESC in the presence of Activin A become endoderm fated.

Page 60: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

47

The effect of Wnt signalling was also investigated here. A canonical Wnt ligand, Wnt3a, was

added with BMP2 and Activin to determine if it would further increase mesoderm

differentiation. It was found, however, that this is not the case. Wnt3a had no significant effect

on Bry expression, neither did BIO, a synthetic activator of the Wnt pathway. This suggests that

either Wnt signalling is not required for mesoderm induction from hESC or that endogenous

secretion of Wnt ligand is sufficient for mesoderm specification in the presence of BMP2 and

Activin A.

This relationship between Smad1 signalling and mesoderm differentiation is strengthened by

observation that both nuclear pSmad1 intensity and Bry expression increases with local cell

density. In these regions of higher local cell density and higher Smad1 signalling, Bry expressing

mesoderm precursors emerge whereas in regions of lower local cell density and lower Smad1

signalling, Sox17 expressing endoderm precursors emerge. Smad1 signalling was also

investigated but the opposite relationship between Smad1 activation and local cell density is

observed when compared to the report by Peerani and colleagues (Peerani et al. 2007). They find

that Smad1 signalling decreases with local cell density and show that opposing signals from

endogenous secretion of BMP2 and GDF3 (growth and differentiation factor 3), an antagonist of

BMP signalling, is responsible for this phenomenon. However, their experiments were conducted

in serum-free medium containing no cytokines, whereas the experiments presented here are

conducted in the presence of high concentrations of Activin A and BMP2. These two signalling

environments are drastically different considering that Activin A and BMP2 elicit the expression

of signalling proteins including Wnt3 and Nodal, as demonstrated here, and expression of BMP2

and BMP4 as described previously (Pera et al. 2004; Nostro et al. 2008).

Page 61: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

48

Although Bry expression is used here as a marker of mesoderm differentiation, there is

considerable evidence suggesting that Bry is also expressed on endoderm precursors. Using an

EGFP-Bry/CD4-FoxA2 reporting mouse embryonic stem cell line it has been shown that

coexpression of Bry and FoxA2 delineates endoderm fated cells, while Bry positive FoxA2

negative cells are mesoderm precursors (Gadue et al. 2006; Nostro et al. 2008). Due to the fact

GFP has been shown to have a half-life of roughly 24 hours (Fukumura et al. 1998), it is possible

that Bry and FoxA2 were not coexpressed but that Bry expression had been downregulated prior

to FoxA2 expression. Nonetheless, it does demonstrate that the endoderm fated cells were

derived from a Bry expressing precursor. Moreover, reports have shown that Bry and Sox17 are

coexpressed on hESC derived definitive endoderm precursors following 48 hours of treatment

with Activin A and low concentrations of FBS but that Bry expression is subsequently

downregulated while Sox17 expression persists at the 72 hour time point (D'Amour et al. 2005;

McLean et al. 2007). However, in the conditions described here using high concentrations of

BMP2 and Activin A, it is found that Bry and Sox17 are not coexpressed, but it does not rule out

the possibility that the Bry expressing cells observed here are also capable of giving rise to

endoderm lineages. Further investigation into the developmental potential of the Bry expressing

population through cell sorting techniques as well as temporal analysis of Bry and Sox17

expression are required to confirm the assumption that the Bry expressing cells obtained here are

in fact mesoderm and not endoderm precursors.

The use of a quadratic model to fit the experimental data provides the first reported quantitative

evidence demonstrating that Activin A synergizes with BMP2 with respect to Bry expression.

This is a simple yet powerful tool that allows one to extract quantitative relationships between

BMP2 and Activin A concentrations with mesoderm differentiation. The insignificance of the

quadratic terms for both Activin A and BMP2 concentrations suggests that the response of these

Page 62: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

49

ligands has not reached saturation. If that were the case, the quadratic terms would have a

negative value. Also, the insignificance of the Activin A main effect coefficient demonstrates

that Activin A alone has no effect on Bry expression, regardless of concentration, showing that

Smad2 signalling alone is insufficient for mesoderm differentiation. However, the second-order

interaction coefficient is significant and greater than zero, demonstrating that Activin A

synergizes with BMP2. Had the second-order interaction coefficient been negative, this would

suggest an inhibitory effect of Activin A.

The role of Smad1 regulating the differentiation axis between endoderm and mesoderm of ESC

is supported by the correlation between nuclear pSmad1 levels and local cell density. It is shown

that cells in a higher local cell density microenvironment show higher activation of Smad1 and it

is in these regions of greater local cell density where Bry expressing mesoderm precursors are

most highly enriched. The activity of Smad1 signalling specifically in mesoderm precursors is

supported by a previous report showing that Smad1 transcript levels are greater in the Bry

expressing population compared to the Bry negative population in differentiating mESC (Zafonte

et al. 2007).

Page 63: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

50

3 Specific Aim #2: Maximizing Mesoderm Development from Mixed Mesendoderm Population

3.1 Overview Considering that BMP2 together with Activin A induces both mesoderm and endoderm

precursors, further control of the stem cell microenvironment is required for hESC differentiation

such that mesoderm rather than endoderm precursors emerge. As described previously, local cell

density and colony size have a significant effect on hESC self-renewal and differentiation

mediated by endogenous signalling. This objective is to use techniques developed by Peerani and

colleagues (Peerani In Press) to precisely control hESC colony size in order to manipulate local

cell density and modulate endogenous signalling effects in the presence of Activin A and BMP2.

Activin/Nodal signalling is autoregulated through feedforward promotion of its own signalling

cascade as well as the expression of inhibitors, namely Lefty1 (left-right determination factor-1),

and Cer1 (Cerberus1) in the overlaying visceral endoderm (Brennan et al. 2001; Ulloa et al.

2001; Norris et al. 2002; Perea-Gomez et al. 2002; Robertson et al. 2003). TGFβ signalling has

also been shown to induce the expression of Wnt ligands (Zhou et al. 2004) and that Wnt and

TGFβ signalling act cooperatively to activate the transcription of developmentally regulated

genes in the Xenopus embryo through physical interactions between LEF-1/TCF and Smad3, a

downstream signalling protein in the TGFβ pathway (Labbe et al. 2000). The spatially controlled

expression of these ligands that interact with TGFβ signalling is essential for correct tissue

patterning and specifying the fate of cells migrating through the primitive streak. It is believed

that by controlling the colony size of differentiating hESC in the presence of Activin A and

BMP2, one can concurrently control the relative signalling effects of other ligands that are

known to interact with the TGFβ superfamily in specifying cell fates. Considering the results of

Specific Aim 1, it is anticipated that hESC differentiated as larger colonies, which mimic the

Page 64: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

51

microenvironment of high local cell density, will result in selective specification of the

mesoderm lineage, while smaller colonies, which mimic the microenvironment of low local cell

density, will result in selective specification of the endoderm lineage. It is believed that this will

allow for uniform differentiation of hESC towards a single lineage when compared to random

seeding of hESC clumps.

3.2 Materials and Methods

3.2.1 Creation of Micropatterned Substrate Microfabricated poly(dimethylsiloxane) (PDMS) stamps were created using standard soft

lithography techniques (Xia et al. 1998) and as described in (Peerani In Press). Three-inch <100>

silicon wafers (Silicon Sence Inc.) were spin-coated with SU-8 25 photoresist (Microchem Corp)

and exposed to ultraviolet (UV) radiation through a mask with opaque features. Areas exposed to

UV radiation are crosslinked and resistant to dissolution by developing solvent. Areas not

exposed were dissolved in SU-8 developer (Microchem Corp) leaving behind an etched layer of

photoresist polymer between 40 to 60 µm in thickness. This creates a mould from which

Poly(dymethylsiloxane) (PDMS) stamps were cast. PDMS prepolymer was mixed with a

crosslinking agent (Sylgard® 184 Silicone Elastomer Kit) and poured onto the microfabricated

silicon wafer. The PDMS was cured at 70°C for 4 hours then peeled away from silicon wafer

creating a stamp with raised features corresponding to the areas exposed by UV radiation. The

stamps were ultrasonically cleaned and sterilized in 70% ethanol for 24 hours. PDMS stamps

were inked with growth factor reduced- (GFR) Matrigel™ diluted 1:30 with pH 5 PBS for at

least 1 hour at 4°C. Excess GFR-Matrigel™ was rinsed away with deionized water and quickly

dried using pressurized nitrogen gas. Dried stamps were then pressed onto 3”×1” tissue-culture

treated microscope slides (Nunc) or 60 mm tissue-culture treated dishes (BD) then incubated at

Page 65: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

52

room temperature in a humidity controlled chamber (85% relative humidity) for 10 minutes to

allow rehydration of Matrigel™ protein and transfer to culture substrate. The stamps are

removed and the surface is passivated using 5% (w/v) Pluronic F-127 (Sigma) for at least 1 hour.

Pluronic acid surfactant coats areas not printed with protein to prevent non-specific attachment

of cells (Figure 3-1).

3.2.2 Seeding of hESC onto Micropatterned Substrate Human ESC maintained on mEFs were dissociated into single cells using TrypLE™ Express for

4 minutes. TrypLE™ was inactivated and diluted using culture media saved from hESC culture

and then the single cell suspension was strained through a 40 µm cell strainer (BD Falcon) to

remove cell clumps and large debris. hESC were resuspended in XV-FAT maintenance medium

and seeded onto micropatterned Matrigel™ substrates at a concentration of 1500 cells/mm2.

Following overnight incubation, non-attached cells were rinsed away three times with X-

VIVO10™ medium, then cultured with X-VIVO™ supplemented with specified cytokines.

Figure 3-1 Overview of microcontact printing. A) PDMS stamps are inked with Matrigel™; b) Excess Matrigel™ is rinsed away and dried; c) Matrigel™ coated stamp is pressed onto tissue-culture substrate; d) Stamp and substrate are incubated at 85% humidity then stamp is removed transferring Matrigel™ protein onto substrate; e) Single cell suspension is placed on micropatterned substrate and cultured overnight; f) Non-attached cells are rinsed away.

Page 66: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

53

3.2.3 Flow Cytometry Analysis All flow cytometry analysis was conducted using a FACSCanto flow cytometer (BD

Biosciences). Micropatterned colonies and embryoid bodies were dissociated into single cells

using TrypLE™ Express for 4 and 10 minutes, respectively. For intracellular staining of Bry,

cells were fixed in 3.7 % formaldehyde for 15 minutes then permeabilized with methanol on ice

for 2 minutes. Goat anti-human Bry antibody (R&D Systems) was used at 1 µg/mL, and

counterstained with Alexa Fluor® 488 donkey anti-goat IgG secondary antibody (1:100). Cells

stained with secondary antibody only were used as a negative control. Allophycocyanin- (APC)

conjugated mouse anti-human KDR antibody (R&D Systems), fluorescent isothiocyanate-

(FITC) conjugated mouse anti-human CD31 antibody (BD Pharmingen), Phycoerythrin- (PE)

conjugated mouse anti-human CD34 antibody (Beckman Coulter) were used at 1:10 dilution.

PE-conjugated mouse anti-human CD117 (also known as c-kit, BD Biosciences) and APC-

conjugated mouse anti-human CD184 (also known as CXCR4, BD Pharmingen) were used at a

1:50 dilution. FITC-, APC-, and PE-conjugated mouse IgG isotype controls (Beckman Coulter)

were used as controls at a dilution of 1:10.

3.3 Results

3.3.1 Higher Cell Densities Cause Increased Brachyury Expression Although high concentrations of both BMP2 and Activin A maximize mesoderm differentiation,

endoderm precursors also emerge in this condition as evidenced by expression of MIXL1, Sox17,

and GSC. In light of the observation that the frequency of Bry expressing mesoderm precursors

is greatest in areas of high cell density while Sox17 expressing endoderm precursors are enriched

in areas of low local cell density, it is hypothesized that directly manipulating cell density can

control mesoderm versus endoderm differentiation. To test this hypothesis, microcontact printing

Page 67: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

54

technology was used to create hESC colonies of controlled size and geometry. Circular colonies

of 200, 400, 800, and 1200 µm in diameter were successfully produced. The ratio of the diameter

to the pitch (spacing between colonies) was kept constant so that the fraction of the total area

covered by hESC was also held constant to minimize the effects of macroscopic cell density (i.e.

the total number of cells per well) (Figure 3-2). The larger colonies are designed to mimic the

microenvironment of areas of high local cell density while smaller colonies represent areas of

low cell density. The Neighbours Analysis algorithm was applied to micropatterned cultures to

confirm that cells cultured as small 200 µm colonies are all found to have low local cell density

while the majority of cells cultured as large 1200 µm colonies are found to have high local cell

density (Figure 3-3).

Figure 3-2 Phase contrast image of micropatterned colonies of indicated diameter.

Page 68: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

55

# Cells within 200 µm radius

0-50

50-10

0

100-1

50

150-2

00

200-2

50

250-3

00

300-3

50

350-4

00

400-4

50

% o

f Tot

al

010203040506070

200 µm1200 µm

Figure 3-3 Histogram of local cell density for 200 and 1200 µm micropatterned cultures

The micropatterned colonies were treated in an identical manner as the hESC that were seeded

randomly as clumps described in Specific Aim 1. Following 24 hours of seeding in serum-free

maintenance medium, the micropatterned colonies were treated with 50 ng/mL of Activin A and

BMP2 for 48 hours and analyzed for mesoderm and endoderm differentiation. Confirming the

initial hypothesis, the expression of T-Bry and KDR increases with increasing colony size

indicating greater mesoderm differentiation in larger, higher density colonies (Figure 3-4).

Conversely, the expression of the endoderm-associated transcripts MIXL1, GSC, Sox17 increases

with decreasing colony size indicating greater endoderm differentiation in smaller, lower density

colonies (Figure 3-4). Gene expression analysis was confirmed through protein level analysis of

Bry and KDR by flow cytometry showing greater frequency of protein expression in larger

colonies (Figure 3-5).

Page 69: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

56

T-Bry

KDRGSC

Sox17

Cer1Le

fty1

Rel

ativ

e E

xpre

ssio

n

0.0

0.5

1.0

1.5

2.0

2.5200 µm400 µm800 µm1200 µm

****

*

*

*

Figure 3-4 Quantitative gene expression analysis of micropatterned cultures following 48 hours of treatment with BMP2 and Activin A. (*) indicates significance (p<0.05) compared to 200 µm colonies.

Figure 3-5 Flow cytometry analysis of Bry and KDR expression of micropatterned cultures. (*) indicates significance (p<0.05) compared to 200 µm colonies.

3.3.2 Larger Colonies Have Greater Smad1 Signalling As described in Specific Aim 1 (Chapter 2), it was shown that differential levels of Smad1

signalling might explain the increased mesoderm differentiation in areas of high local cell

Page 70: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

57

density. Nuclear pSmad1 levels increased as a function of local cell density leading to mesoderm

differentiation at high local cell density and endoderm differentiation at low local cell density.

This relationship is once again evident in the micropatterned colonies where cells in the large

1200 µm colonies are characterized by higher levels of nuclear pSmad1 when compared to the

smaller 200 µm colonies following a 90 minute treatment with BMP2 and Activin A (Figure

3-6). This reinforces the relationship between local cell density, pSmad1 and Bry expression.

Independently controlling colony size and local cell density demonstrates that effect of local cell

density on Bry expression and Smad1 signalling is in fact causative and not merely correlative.

Relative Fluorescent Intensity

0.0 0.2 0.4 0.6 0.8 1.0 1.2 1.4 1.6 1.8

1200 µm A+B

200 µm A+BNuclearpSmad1

*

Figure 3-6 Quantitative immunofluorescence of nuclear pSmad1 intensity of micropatterned cultures. (*) indicates p<0.05

3.3.3 Smaller Colonies Express Greater Levels of BMP Antagonists To investigate a possible mechanism governing the different levels of Smad1 signalling in large

and small colonies, the expression of BMP signalling antagonists was measured. Quantitative

gene expression analysis shows that expression of Cer1 (Cerberus 1) and Lefty1 (left-right

determination factor 1) are greater in smaller colonies (Figure 3-4). Cer1 encodes a secreted

protein that binds to BMP proteins extracellulary preventing its signalling activity (Piccolo et al.

Page 71: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

58

1999). Lefty1, on the other hand, encodes an intracellular protein that inhibits the

heterodimerization of Smad5 with Smad4, preventing its nuclear localization (Ulloa et al. 2001).

Together, it is believed that both these inhibitors of BMP signalling are responsible for

attenuating Smad1 signalling in smaller colonies.

3.4 Discussion The findings discussed in Specific Aim 1 (Chapter 2) show that increased local cell density

positively correlates with mesoderm differentiation and negatively correlates with endoderm

differentiation. It is conceivable that mesoderm and endoderm precursors emerge stochastically

in culture and migrate leading to the observed relationship with local cell density. However,

controlling colony size through microcontact printing shows that this relationship is causative.

Independently controlling colony size shows that a higher local cell density microenvironment is

conducive to mesoderm differentiation while a lower local cell density microenvironment is

conducive to endoderm differentiation. This is not to say, however, that the combination of

Activin A and BMP2 is ideal for endoderm differentiation even if colony size is restricted. This

condition has been identified as inducing optimal mesoderm differentiation but it is also found to

support endoderm differentiation. Typical definitive endoderm differentiation protocols employ

high concentrations (100 ng/mL) of Activin A alone (D'Amour et al. 2005; D'Amour et al. 2006).

Although the combination of Activin A and BMP2 at 50 ng/mL is permissive to both mesoderm

and endoderm differentiation, the control of colony size is able to direct lineage specification.

Furthermore, the relationship between local cell density and Smad1 signalling, as was seen in

Specific Aim 1, again holds true in the micropatterned scenario. Independently controlling

colony size is able to manipulate Smad1 signalling in the presence of BMP2 and Activin A. The

Page 72: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

59

trend of increasing Smad1 activation with greater local cell density is again demonstrated in the

micropatterned colonies whereby larger 1200 µm colonies are characterized by higher levels of

pSmad1 compared to the smaller 200 µm colonies demonstrating that local cell density causes

differences in Smad1 signalling. As mentioned in Specific Aim 1, this trend is opposite to that

described by Peerani and colleagues (Peerani et al. 2007) where they report increasing Smad1

activation in smaller colony sizes. Again, this discrepancy is due to the fact that their assays were

conducted in the absence of any exogenous cytokines. In this assay, high levels of BMP and

Activin/Nodal ligands are present creating a drastically different signalling environment. The

addition of these cytokines induces the expression of a different set of endogenous signalling

proteins including Wnt3, Nodal, Cerberus 1, and Lefty 1.

In the presence of BMP2 and Activin A, it is proposed that the Smad1 signalling gradient is due

to endogenous expression of Cer1 and Lefty1, inhibitors of BMP signalling. Both Lefty1 and

Cer1 are known to be expressed in the overlying visceral endoderm of the early primitive streak-

stage mouse embryo where they serve to restrict primitive streak and mesoderm formation to the

posterior side of the embryo. In the in vitro model using hESC, it is proposed that Lefty1 and

Cer1 are more highly expressed in areas of low cell density and attenuate BMP signalling to

limit mesoderm differentiation and allow endoderm formation. To further confirm this

mechanism small interfering (si)RNA could be used to block Lefty1 or Cerberus expression. In

this proposed model, the addition of siRNA against Lefty1 or Cer1 to small colonies would result

in higher nuclear pSmad1 levels, greater mesoderm, and less endoderm differentiation.

These results demonstrate that micropatterning hESC into defined colony sizes allows one to

probe different microenvironments by specifying a specific local cell density. By doing so, it has

allowed the identification of Lefty1 and Cer1 as potential regulators of mesendoderm

Page 73: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

60

differentiation in hESC. Moreover, it allows for the specification of desired germ layers from

hESC allowing for decreased heterogeneity of differentiation that would otherwise occur in

traditional, randomly seeded hESC cultures.

Page 74: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

61

4 Specific Aim #3: Differentiation of Mesoderm and Endoderm to Hematopoietic Progenitors and Primitive Gut Tube

4.1 Overview Once an optimal mesoderm population has been established, the next step is to further

differentiate mesoderm precursors into hemogenic mesoderm and hematopoietic progenitors.

The mechanisms through which this occurs, however, remains poorly understood. Due to the

increased complexity of the mouse and human embryo post-gastrulation, it is increasingly

difficult to decipher the signalling network responsible for hematopoietic induction. However, as

mentioned earlier, several cytokines have been identified to promote hemogenic mesoderm, but

it remains unclear if these cytokines induce hemogenic mesoderm or simply maintain and

expand hematopoietic progenitors that arise through an unknown mechanism. Moreover, in an

effort to more precisely understand the mechanisms underlying hematopoiesis, a completely

defined culture medium is desired to preclude the effects of undefined factors present in serum or

secreted by hemogenic stromal cells. Additionally, it has been shown that, in a stromal free

system, attached culture inhibits hematopoiesis while promoting endothelial differentiation

(Dang et al. 2002).

With these considerations in mind, a previously established differentiation protocol will be

employed (Chadwick et al. 2003; Ng et al. 2005). This protocol involves an embryoid body (EB)

differentiation system that uses defined culture medium supplemented with several cytokines

shown to support hematopoietic differentiation and expansion. The potential to form endoderm

lineages will also be assessed using a previously reported protocol for specifying primitive gut

tube and pancreatic endoderm (D'Amour et al. 2005; D'Amour et al. 2006; Kroon et al. 2008).

Page 75: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

62

The use of the previously established protocols allows for the unbiased and direct comparison of

the mesoderm and endoderm forming potential of the micropatterned colonies.

4.2 Materials and Methods

4.2.1 Fabrication of Microwells Microwells were fabricated as described by Ungrin and colleagues (Ungrin et al. 2008). Silicon

moulds were generated by the Canadian Microsystems Consortium (http://www.cmc.ca). <100>

silicon wafers were coated with a silicon nitride layer followed by a layer of photoresist. The

photoresist was selectively removed using an opaque mask, UV radiation, and photoresist

developer to produce a protective square grid pattern on top of the silicon nitride layer. Deep

reactive ion etching was used to remove the silicon nitride layer in areas not protected by

photoresist. The remaining photoresist was then chemically dissolved. Wet anisotropic etching

was used to etch square pyramids outlined by the silicon nitride layer producing a silicon master

mould consisting of an array of square pyramids 800 µm in width (Figure 4-1). Reviewed in

Kovacs et al. (1998). The microfabricated silicon wafer was used to cast a PDMS negative

mould. PDMS prepolymer was mixed with a crosslinking agent and poured over the silicon

wafer and cured at 70°C for 4 hours then peeled away from silicon wafer creating an array of

pyramidal peaks. The negative mould was then used to cast a positive mould using the same

process creating a microtextured PDMS sheet containing an array of pyramidal wells of 800 µm

in width. The microtextured PDMS sheets were then cut into circular inserts that fit tightly into

the bottom of 24-well plates resulting in approximately 300 microwells per insert. The circular

inserts were ultrasonically cleaned and sterilized by autoclave. Prior to use, inserts were coated

with 5% (w/v) Pluronic-F127 (Sigma) solution for at least 1 hour to prevent cell attachment to

PDMS inserts.

Page 76: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

63

Figure 4-1 Schematic diagram of microwell fabrication.

4.2.2 Generation of Forced Aggregate Embryoid Bodies Forced aggregate embryoid bodies (spin EBs) were generated as originally described by Ng and

colleagues (Ng et al. 2005) and modified by Ungrin and colleagues (Ungrin et al. 2008).

Following 48 hours of treatment with BMP2 and Activin A, micropatterned hESC colonies were

dissociated into single cells using TrypLE™ Express for 4 minutes then rinsed thoroughly with

X-VIVO10™ medium to remove any residual TrypLE™ Express. Harvested cells were then

resuspended in X-VIVO10™ serum-free medium supplemented with 50 ng/mL rhSCF (R&D

Systems), 150 ng/mL rhFlt3 Ligand (R&D Systems), 10 ng/mL rhVEGF165 (R&D Systems), and

10 µm Y-27632 Rho kinase (ROCK) inhibitor (Calbiochem). ROCK inhibitor is added in order

to improve survival of single cell suspension (Watanabe et al. 2007), though the mechanism

through which it acts remains unclear. The single cell suspension (5,000 cells per microwell) was

pipetted onto microtextured PDMS inserts within a well of a 24-well plate. The entire plate is

centrifuged for 3 minutes at 200g to pellet single cell suspension into the bottom of microwells.

Page 77: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

64

The cells were then cultured overnight at 37°C to allow embryoid body (EB) formation. EBs

were then removed from the microwells by gently pipetting with a 1 mL pipette and rinsed with

PBS to remove ROCK inhibitor, resulting in roughly 300 EBs with approximately 5,000 cells per

EB. EBs were subsequently cultured in ultralow adhesion 6-well plates (Costar) in the same

medium as above without ROCK inhibitor for 9 days (Figure 4-2).

Figure 4-2 Schematic diagram of microwell spinEB formation. a) Micropatterned cultures are enzymatically dissociated into single cells and pipetted onto microwells; b) Microwells are centrifuged to pellet cells into bottom of microwells and cultured overnight to form embryoid bodies (EB); c) EBs are then removed from the microwells and cultured in suspension.

4.2.3 Flow Cytometry Flow cytometry was performed as described in the Materials and Methods section of Specific

Aim 1.

4.2.4 Hematopoietic Colony Forming Assay After 9 days of embryoid body culture, EBs were dissociated into single cells using TrypLE™

Express for 10 minutes and pipetting through a 200 µL micropipette tip. Single cell suspensions

Page 78: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

65

were strained through a 40 µm cell strainer to remove any cell clumps. Single cells were

resuspended in MethoCult® GF+ semi-solid medium (H4435, Stem Cell Technologies) at a

density of 50,000 cells per mL. Hematopoietic colonies were counted following 14 days of

culture. A minimum of 100,000 cells was assayed per condition per replicate.

4.2.5 Primitive Gut Endoderm Differentiation The differentiation protocol implemented here was first described in references (D'Amour et al.

2005; D'Amour et al. 2006; Kroon et al. 2008). Following 48 hours of treatment with BMP2 and

Activin A, 200 and 1200 µm diameter micropatterned cultures were reseeded onto Matrigel™

coated 6-well plates by physical scraping with a cell scraper (Sarstedt). Cells from an entire 60

mm culture dish were seeded into 2 wells of a 6-well plate in X-VIVO10™ medium

supplemented with 100 ng/mL Activin A. Following 2 days of culture, medium was replaced

with RPMI Medium 1640 (Gibco) supplemented with 2% FBS, 2 mM GlutaMAX™

(Invitrogen), 50 U/mL Penicillin/Streptomycin (Invitrogen), and 50 ng/mL fibroblast growth

factor 7 (FGF7, or keratinocytes growth factor, KGF, R&D Systems). Following 3 more days of

culture, medium was replaced with DMEM supplemented with 1% B27 supplement (Gibco), 2

mM GlutaMAX™ (Invitrogen), 50 U/mL Penicillin/Streptomycin (Invitrogen), 50 ng/mL

Noggin (R&D Systems), 250 nM cyclopamine (a kind gift from Dr. Derek van der Kooy,

University of Toronto), and 2 µm retinoic acid (RA). Following 3 more days of differentiation,

cultures were fixed and stained for immunofluorescence or harvested for quantitative gene

expression analysis as described in the Materials and Methods section of Specific Aim 1.

Page 79: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

66

4.3 Results

4.3.1 Larger, Mesoderm Enriched Colonies Demonstrate Greater Hematopoietic Potential

A hematopoietic differentiation assay was used to assess the functional capacity of the mesoderm

precursors described in Specific Aim 2. Following the initial 48 hours of mesoderm induction

with BMP2 and Activin A, the micropatterned colonies were dissociated into single cells and

formed into forced aggregate embryoid bodies as originally described by Ng and colleagues (Ng

et al. 2005) and modified by Ungrin and colleagues (Ungrin et al. 2008). The embryoid bodies

were cultured in suspension for 9 days in X-VIVO10™ serum-free medium supplemented with

hemogenic cytokines VEGF (vascular endothelial growth factor, 10 ng/mL), SCF (stem cell

factor, 50 ng/mL) and Flt-3 ligand (150 ng/mL) (Chadwick et al. 2003). Following 9 days of

culture, the embryoid bodies were dissociated into single cells and hematopoietic potential was

assessed through hematopoietic colony forming assay (Eaves et al. 1999) and flow cytometry

analysis. This hematopoietic differentiation protocol was employed because it allows for the

tight control of embryoid body size and number so that the differentiation of hESC is uniformly

conducted except for the variation of colony size during the initial 48 hours of differentiation.

This allows for the isolation of the effect of local cell density on early mesoderm differentiation.

Flow cytometry analysis shows that hESC differentiated in larger colonies give rise to 2.5-fold

(p<0.05) greater frequency of CD34, CD31 (PECAM1, platelet endothelial cell adhesion

molecule 1) and VEGFR2 expressing cells, markers of hematopoietic progenitors (Tavian et al.

1996; Labastie et al. 1998; Tavian et al. 1999) (Figure 4-3). Hematopoietic colony forming assay

further demonstrates that the larger, mesoderm enriched colonies yield significantly greater

frequencies of colony forming units (CFU), a 10-fold increase (p<0.05) when compared to the

smaller colonies. Such colony forming units include CFU-macrophage (CFU-M), CFU-

Page 80: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

67

granulocytes, macrophage (CFU-GM), and CFU-granulocytes, erythroid, macrophage

megakaryocyte (CFU-GEMM) (Figure 4-4 and Figure 4-5). This demonstrates the functional

output of the mesoderm precursors described in Specific Aim 2 and shows that the larger, higher

local cell density colonies are in fact enriched for mesoderm precursors.

Figure 4-3 Flow cytometry analysis shows that EB cultures derived from 1200 µm colonies express markers of hematopoietic differentiation at a greater frequency compared to 200 µm colonies. (*) indicates significance (p<0.05).

Page 81: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

68

200 µ

m

1200

µm#

Col

onie

s pe

r 105

cel

ls0

5

10

15

20

25

30

35*

Figure 4-4 Hematopoietic colony forming assay shows that EB cultures derived from 1200 µm colonies have a greater frequency of colony forming units. (*) indicates significance (p<0.05).

Page 82: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

69

Figure 4-5 Representative phase contrast images of hematopoietic colonies showing CFU-GM (a), CFU-M (b) and CFU-GEMM (c, d). Original magnification is 10× and 4×, scale bar represents 200 µm.

4.3.2 Smaller, Endoderm Enriched Colonies Demonstrate Greater Primitive Gut Endoderm Potential

Although the hematopoietic differentiation assay described above demonstrates that the larger

colonies have greater potential for mesoderm differentiation when compared to the smaller

colonies, an endoderm differentiation assay is required to support the earlier finding that larger

colonies are deficient in endoderm potential while the smaller colonies are enriched in endoderm

precursors. To accomplish this the micropatterned hESC colonies were further cultured in

conditions supportive of definitive gut tube and pancreatic endoderm differentiation (D'Amour et

al. 2005; D'Amour et al. 2006; Kroon et al. 2008). Following 48 hours of treatment with BMP2

and Activin A, 200 and 1200 µm diameter micropatterned colonies were subsequently treated

a b

c d

Page 83: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

70

with Activin A alone for 2 days, FGF7 for 3 days, followed by RA, cyclopamine and Noggin for

3 more days.

Following 48 hours the treatment with Activin A alone, cultures originating from the 200 µm

colonies express CXCR4 (chemokine (C-X-C motif) receptor 4), a marker of definitive

endoderm (McGrath et al. 1999; D'Amour et al. 2005; Yasunaga et al. 2005; D'Amour et al.

2006) at a significantly greater frequency when compared to cultures originating from the larger

1200 µm colonies as measured by flow cytometry (Figure 4-6). Moreover, after treatment with

FGF7 followed by Noggin, RA, and cyclopamine, cultures originating from 200 µm colonies

express hepatocyte nuclear factor-1 β (HNF1β) (Coffinier et al. 1999), FoxA2 (forkhead box A2,

or HNF3β) (Sasaki et al. 1993), and HNF4α (Duncan et al. 1994), markers of primitive gut tube

differentiation, whereas cultures originating from 1200 µm colonies do not (Figure 4-7).

Quantitative gene expression analysis confirms the protein expression showing that HNF1β,

FoxA2, and HNF4α gene expression is also greater in cultures derived from the smaller 200 µm

colonies (Figure 4-8a). Moreover, it is also found that transcripts associated with pancreatic

epithelium such as hepatocyte nuclear factor-6 (HNF6) (Rausa et al. 1997), insulin promoter

factor-1 (IPF1, or pancreas/duodenum homeobox-1, PDX1) (Wilson et al. 2003), and motor

neuron and pancreas homeobox 1 (MNX1) (Wilson et al. 2003) are also more highly expressed

in cultures derived from the smaller colonies, though the overall expression is considerably less

than human pancreatic tissue (Figure 4-8b). Together, these results demonstrate that the 200 µm

colonies are enriched for definitive endoderm precursors capable of primitive gut tube and

pancreatic endoderm differentiation.

Page 84: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

71

Figure 4-6 Flow cytometry analysis of CXCR4 expression of attached cultures derived from 200 and 1200 µm colonies following 2 days of culture in Activin A. Red histogram is of isotype control, green histogram represents sample. (*) indicates p<0.05.

Page 85: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

72

Figure 4-7 Fluorescent images of cultures derived from 200 and 1200 µm colonies following endoderm differentiaiton assay showing HNF1β, FoxA2, and HNF4apha staining. Nuclei are shown in blue. Scale bar represents 100 µm; original magnification is 20× and 40×.

Page 86: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

73

HNF1beta

HNF4alph

aFox

A2

Rel

ativ

e E

xpre

ssio

n

0

20

40

60

80

hESC200 µm1200 µm

HNF6IP

F1MNX1

0.00

0.05

0.10

1.00

hESC200 µm1200 µmHumanPancreatic Tissue

***

* * *

*

a b

Figure 4-8 Quantitative gene expression analysis of primitive gut tube and pancreatic endoderm cultures derived from micropatterned cultures. a) Cultures derived from smaller 200 µm colonies show greater expression of primitive gut tube-associated genes when compared to 1200 µm colonies; b) Expression of pancreatic endoderm-associated genes is also greater in cultures derived from 200 µm colonies, but considerably less when compared to human pancreatic tissue. (*) and (**) indicate significance (p<0.05 and p<0.01, respectively) when comparing 200 and 1200 µm colony derived cultures.

4.4 Discussion Using markers of early mesoderm and endoderm differentiation such as T-Bry, Sox17, GSC and

MIXL1, it was shown that mesoderm precursors are enriched in areas of higher local cell density

while endoderm precursors are enriched in areas of lower cell density. However, it is still

necessary to demonstrate that these markers of early mesoderm and endoderm differentiation

truly reflect their differentiation potential. It is shown here that using differentiation protocols

Page 87: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

74

supportive of either hematopoietic or primitive gut endoderm differentiation, the larger colonies

represent precursors of the mesoderm and not endoderm lineage while the opposite is true for

small colonies.

A hematopoietic differentiation assay demonstrated the functional output of the mesoderm

progenitors. It was shown that there is a 10-fold increase in the number of hematopoietic colony

forming units in embryoid bodies derived from the larger 1200 µm colonies when compared to

the smaller 200 µm colonies. Although the hematopoietic assay used demonstrates that the larger

colonies give rise to greater frequencies of colony forming units, the overall frequency is

considerably less than other reports. Using a hematopoietic differentiation protocol that includes

a combination of cytokines and serum Chadwick and colleagues report a clonogenic frequency

of hematopoietic precursors of roughly 1:400 (Chadwick et al. 2003), while Ng and colleagues

report a similar frequency of 1:523 (Ng et al. 2005) using serum-free conditions. The frequency

reported here, however, is roughly 1:4300, one tenth of these previous reports. The low

frequency of hematopoietic colonies could be explained by the types of mesoderm precursors

that arise in the micropatterned colonies. Mesoderm differentiation was assessed by Bry

expression, which is a pan-mesodermal marker that is expressed in precursors of all types of

mesoderm, including somitic, cardiac as well as hemogenic mesoderm. Treatment with Activin

A and BMP2 may have skewed mesodermal differentiation towards one of these other lineages

resulting in low hematopoietic frequency. In fact, previous reports have used BMP4 and Activin

A to robustly induce cardiac differentiation from hESC suggesting that this condition is

supportive of cardiac rather than hematopoietic differentiation (Laflamme et al. 2007; Yang et al.

2008). Previous studies have also shown that Wnt signalling acts during later stages of

mesoderm differentiation to specify hematopoietic lineages at the expense of cardiac (Naito et al.

2006; Ueno et al. 2007). In order to more precisely direct hematopoietic differentiation, further

Page 88: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

75

studies could include the addition of exogenous Wnt ligands during embryoid body

differentiation.

To demonstrate that the smaller colonies are enriched from endoderm precursors, a

differentiation assay supportive of primitive gut tube and pancreatic endoderm was employed.

Both protein and gene expression of markers of primitive gut endoderm were shown to be

greater in cultures derived from smaller 200 µm colonies. Moreover, expression of genes

associated with pancreatic epithelium development were also more highly expressed in cultures

derived from smaller colonies, but the overall expression was still considerably less that that of

human pancreatic tissue. This suggests that the pancreatic lineage has not fully matured from the

primitive gut tube endoderm and that further differentiation is required. Nonetheless, these

results demonstrate that the smaller colonies are enriched for endoderm precursors. However, to

further confirm the functional capacity of these endoderm precursors, further differentiation and

functional assays such as insulin secretion and c-peptide release would be required.

The relationship between colony or EB size and mesendoderm differentiation that is observed

here has been previously reported but the mechanism governing this relationship was unknown.

Ng and colleagues have reported that a minimum EB size of roughly 500 cells/EB is required for

hematopoietic differentiation of hESC (Ng et al. 2005) while another report shows that endoderm

differentiation occurs much more efficiently when ESC are seeded at low densities (Cho et al.

2008). It is shown here, however, that the effect of cell density and colony size on mesendoderm

differentiation is mediated by Smad1 signalling where greater Smad1 activation is present at

higher cell densities while lower Smad1 activation is present at lower cell densities. It is

proposed here that this effect of cell density on Smad1 signalling is responsible for controlling

mesoderm versus endoderm differentiation. By manipulating cell density it is now possible to

Page 89: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

76

more finely tune the ESC microenvironment allowing for more tight control over ESC

differentiation.

Page 90: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

77

5 Conclusions In order for stem cell researchers to effectively guide embryonic stem cell differentiation and

obtain pure population of transplantable cells for therapeutic applications, a greater

understanding of the molecular mechanisms controlling ESC differentiation is required. From the

results presented here, it is clear that both Smad1 and Smad2 signalling play pivotal roles in

mesoderm and endoderm differentiation from hESC. Although previous reports have shown that

BMP and Activin/Nodal signalling are required for mesoderm and endoderm differentiation, this

is the first report demonstrating that Smad2 signalling directly targets mesoderm precursors.

Moreover, the use of a quadratic mathematical model has quantitatively confirmed the

synergistic relationship between Activin A and BMP2 in mesoderm differentiation.

Activin/Nodal alone is not sufficient for mesoderm differentiation but it does synergize with

BMP signalling to maximize the frequency of mesoderm precursors.

This is also the first report to elucidate a possible mechanism governing the relationship between

stem cell microenvironment, specifically local cell density, and mesendoderm differentiation

whereby expression of BMP antagonists in areas of low local cell density attenuate Smad1

signalling leading to endoderm differentiation while mesoderm differentiation occurs mainly in

areas of high local cell density. By controlling colony size one can independently manipulate

local cell density, and consequently Smad1 signalling, in order to precisely guide mesendoderm

differentiation of human embryonic stem cells (Figure 5-1).

It is finally shown that this method allows for the control of the differentiation of more mature

cell types including hematopoietic cells and primitive gut endoderm. Using a hematopoietic

differentiation assay, a 10-fold increase in hematopoietic colony forming frequency is observed

Page 91: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

78

in embryoid bodies derived from the larger mesoderm-enriched colonies when compared to the

smaller colonies. On the other hand, an endoderm differentiation protocol reveals that cultures

derived from smaller endoderm enriched colonies show greater protein and gene expression of

markers of primitive gut tube and pancreatic endoderm differentiation. This is a vast

improvement over traditional bulk culture systems where the stem cell microenvironment is not

controlled leading to heterogeneous differentiation. These results highlight the importance of

tightly controlling all aspects of the stem cell niche, including extracellular signalling and cell

density, in order to obtain the desired cell types.

Figure 5-1 Proposed scheme for the regulation of mesendoderm differentiation by local cell density and Smad signalling

Page 92: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

79

6 Future Work

There remain several specific outstanding issues in this thesis that merit further investigation.

Firstly, the role of Cer1 and Lefty1 gene expression needs to be validated through protein level

analysis and loss of function analysis through RNA interference. Demonstrating the effects of

suppressing Cer1 and/or Lefty1 expression through small interfering RNA would definitively

demonstrate the direct effect of Smad1 inhibition on mesendoderm differentiation. Moreover,

other signalling pathways other than Wnt and the TGFβ warrant investigation, such as cell

tension mediated by RhoA activation, or FGF signalling among others. The results of

preliminary experiments looking into the role of Rho kinase and FGF signalling are discussed in

the appendix and they demonstrate the need for further investigation.

Also, as alluded to earlier, the efficiency of hematopoietic differentiation from the mesoderm

precursors is quite low compared to other published reports. This highlights the importance of

not only dissecting the mechanisms governing early germ layer differentiation, but also later

lineage specification. Several reports have identified Wnt signalling as a factor that influences

the differentiation of committed mesoderm precursors to either the cardiac or hematopoietic

lineage (Naito et al. 2006; Ueno et al. 2007). Moreoever, Sukurai and colleagues (Sakurai et al.

2006) have shown using an in vitro mESC model that two populations representing paraxial and

lateral mesoderm have the ability to interconvert between lineages at early stages. The

mechanism underlying this interconversion, however, remains unknown. Also, Wnt3a has been

implicated in the specification of trunk and tail paraxial mesoderm and that graded

concentrations of Wnt3a are required for posterior derivatives of paraxial mesoderm (Takada et

al. 1994; Greco et al. 1996). It is clear that complex interactions between various signalling

Page 93: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

80

pathways regulate the specification of mature cell types from early mesoderm precursors and a

greater understanding of these interactions could explain the seemingly low frequency of

hematopoietic progenitors observed, and also improve the purity and number of desired cell

types, be they hematopoietic, osteogenic, or any other mesoderm-derived cell type.

Thinking more broadly, this project has focused heavily on the differentiation of hESC as an

attached culture on a laminin based substrate. This aspect alone brings into question the

difference of two very distinct differentiation paradigms – attached versus non-attached cultures.

In vivo, embryogenesis occurs in the uterine wall surrounded by extraembryonic tissue, a very

disparate microenvironment than the stiff substrate of a tissue-culture flask. Future work may

involve completely suspension culture systems, such as embryoid bodies, or even semi-solid

medium that more closely resembles the physical properties a developing embryo would

experience in vivo. Similar questions could still be raised, such as the effect of local cell density,

or EB size in this case, on mesendoderm differentiation, or perhaps the role of substrate

mechanical properties on the epithelial-mesenchymal transition (EMT) that characterizes the

development of pluripotent epiblast cells as they migrate through the primitive streak (Moustakas

et al. 2007).

Recently, the ability to generate induced pluripotent stem cells from somatic cells (Takahashi et

al. 2007; Yu et al. 2007) has opened the door to a wide range of possibilities for generating

transplantable hematopoietic stem cells for treating blood disorders. Rather than focusing on

guiding the differentiation of pluripotent cells to hematopoietic progenitors, it is worthwhile to

consider a similar strategy to reprogram somatic cells directly to hematopoietic stem cells

circumventing the need for precise differentiation protocols. This, however, introduces new

challenges such as identifying the minimal combination of factors required for hematopoietic

Page 94: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

81

reprogramming, as well as concerns with tumourogenicity from reintroducing potentially

oncogenic genes or proteins. There are clearly a number of challenges that must be overcome

before a practical and clinically relevant cell-based therapy for treating blood disorders can be

achieved.

Page 95: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

82

7 References Ang, S. L. and D. B. Constam (2004). "A gene network establishing polarity in the early mouse

embryo." Semin Cell Dev Biol 15(5): 555-61.

Arnold, S. J., J. Stappert, A. Bauer, A. Kispert, B. G. Herrmann and R. Kemler (2000). "Brachyury is a target gene of the Wnt/beta-catenin signaling pathway." Mech Dev 91(1-2): 249-58.

Attisano, L., C. Silvestri, L. Izzi and E. Labbe (2001). "The transcriptional role of Smads and FAST (FoxH1) in TGFbeta and activin signalling." Mol Cell Endocrinol 180(1-2): 3-11.

Audet, J., C. L. Miller, C. J. Eaves and J. M. Piret (2002). "Common and distinct features of cytokine effects on hematopoietic stem and progenitor cells revealed by dose-response surface analysis." Biotechnol Bioeng 80(4): 393-404.

Avilion, A. A., S. K. Nicolis, L. H. Pevny, L. Perez, N. Vivian and R. Lovell-Badge (2003). "Multipotent cell lineages in early mouse development depend on SOX2 function." Genes Dev 17(1): 126-40.

Bakre, M. M., A. Hoi, J. C. Mong, Y. Y. Koh, K. Y. Wong and L. W. Stanton (2007). "Generation of multipotential mesendodermal progenitors from mouse embryonic stem cells via sustained Wnt pathway activation." J Biol Chem 282(43): 31703-12.

Barker, J. E. (1968). "Development of the mouse hematopoietic system. I. Types of hemoglobin produced in embryonic yolk sac and liver." Dev Biol 18(1): 14-29.

Beck, F., T. Erler, A. Russell and R. James (1995). "Expression of Cdx-2 in the mouse embryo and placenta: possible role in patterning of the extra-embryonic membranes." Dev Dyn 204(3): 219-27.

Bhatia, M., D. Bonnet, U. Kapp, J. C. Wang, B. Murdoch and J. E. Dick (1997). "Quantitative analysis reveals expansion of human hematopoietic repopulating cells after short-term ex vivo culture." J Exp Med 186(4): 619-24.

Bradley, A., M. Evans, M. H. Kaufman and E. Robertson (1984). "Formation of germ-line chimaeras from embryo-derived teratocarcinoma cell lines." Nature 309(5965): 255-6.

Brennan, J., C. C. Lu, D. P. Norris, T. A. Rodriguez, R. S. Beddington and E. J. Robertson (2001). "Nodal signalling in the epiblast patterns the early mouse embryo." Nature 411(6840): 965-9.

Brotherton, T. W., D. H. Chui, J. Gauldie and M. Patterson (1979). "Hemoglobin ontogeny during normal mouse fetal development." Proc Natl Acad Sci U S A 76(6): 2853-7.

Brugger, W., S. Scheding, B. Ziegler, H. J. Buhring and L. Kanz (2000). "Ex vivo manipulation of hematopoietic stem and progenitor cells." Semin Hematol 37(1 Suppl 2): 42-9.

Page 96: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

83

Chadwick, K., L. Wang, L. Li, P. Menendez, B. Murdoch, A. Rouleau and M. Bhatia (2003). "Cytokines and BMP-4 promote hematopoietic differentiation of human embryonic stem cells." Blood 102(3): 906-15.

Chen, X., M. J. Rubock and M. Whitman (1996). "A transcriptional partner for MAD proteins in TGF-beta signalling." Nature 383(6602): 691-6.

Cho, C. H., N. Parashurama, E. Y. Park, K. Suganuma, Y. Nahmias, J. Park, A. W. Tilles, F. Berthiaume and M. L. Yarmush (2008). "Homogeneous differentiation of hepatocyte-like cells from embryonic stem cells: applications for the treatment of liver failure." FASEB J 22(3): 898-909.

Choi, K., M. Kennedy, A. Kazarov, J. C. Papadimitriou and G. Keller (1998). "A common precursor for hematopoietic and endothelial cells." Development 125(4): 725-32.

Chu, G. C., N. R. Dunn, D. C. Anderson, L. Oxburgh and E. J. Robertson (2004). "Differential requirements for Smad4 in TGFbeta-dependent patterning of the early mouse embryo." Development 131(15): 3501-12.

Coffinier, C., J. Barra, C. Babinet and M. Yaniv (1999). "Expression of the vHNF1/HNF1beta homeoprotein gene during mouse organogenesis." Mech Dev 89(1-2): 211-3.

Conneally, E., J. Cashman, A. Petzer and C. Eaves (1997). "Expansion in vitro of transplantable human cord blood stem cells demonstrated using a quantitative assay of their lympho-myeloid repopulating activity in nonobese diabetic-scid/scid mice." Proc Natl Acad Sci U S A 94(18): 9836-41.

D'Amour, K. A., A. D. Agulnick, S. Eliazer, O. G. Kelly, E. Kroon and E. E. Baetge (2005). "Efficient differentiation of human embryonic stem cells to definitive endoderm." Nat Biotechnol 23(12): 1534-41.

D'Amour, K. A., A. G. Bang, S. Eliazer, O. G. Kelly, A. D. Agulnick, N. G. Smart, M. A. Moorman, E. Kroon, M. K. Carpenter and E. E. Baetge (2006). "Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells." Nat Biotechnol 24(11): 1392-401.

Dale, L., G. Howes, B. M. Price and J. C. Smith (1992). "Bone morphogenetic protein 4: a ventralizing factor in early Xenopus development." Development 115(2): 573-85.

Dang, S. M., M. Kyba, R. Perlingeiro, G. Q. Daley and P. W. Zandstra (2002). "Efficiency of embryoid body formation and hematopoietic development from embryonic stem cells in different culture systems." Biotechnol Bioeng 78(4): 442-53.

Davey, R. E. and P. W. Zandstra (2006). "Spatial organization of embryonic stem cell responsiveness to autocrine gp130 ligands reveals an autoregulatory stem cell niche." Stem Cells 24(11): 2538-48.

Davis, R. P., E. S. Ng, M. Costa, A. K. Mossman, K. Sourris, A. G. Elefanty and E. G. Stanley (2008). "Targeting a GFP reporter gene to the MIXL1 locus of human embryonic stem cells identifies human primitive streak-like cells and enables isolation of primitive hematopoietic precursors." Blood 111(4): 1876-84.

Page 97: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

84

Derynck, R. and Y. E. Zhang (2003). "Smad-dependent and Smad-independent pathways in TGF-beta family signalling." Nature 425(6958): 577-84.

Dieterlen-Lievre, F., I. E. Godin, J. A. Garcia-Porrero and M. A. Marcos (1994). "Initiation of hemopoiesis in the mouse embryo." Ann N Y Acad Sci 718: 140-6.

Duncan, S. A., K. Manova, W. S. Chen, P. Hoodless, D. C. Weinstein, R. F. Bachvarova and J. E. Darnell, Jr. (1994). "Expression of transcription factor HNF-4 in the extraembryonic endoderm, gut, and nephrogenic tissue of the developing mouse embryo: HNF-4 is a marker for primary endoderm in the implanting blastocyst." Proc Natl Acad Sci U S A 91(16): 7598-602.

Eaves, C., C. Miller, E. Conneally, J. Audet, R. Oostendorp, J. Cashman, P. Zandstra, S. Rose-John, J. Piret and A. Eaves (1999). "Introduction to stem cell biology in vitro. Threshold to the future." Ann N Y Acad Sci 872: 1-8.

Ellis, P., B. M. Fagan, S. T. Magness, S. Hutton, O. Taranova, S. Hayashi, A. McMahon, M. Rao and L. Pevny (2004). "SOX2, a persistent marker for multipotential neural stem cells derived from embryonic stem cells, the embryo or the adult." Dev Neurosci 26(2-4): 148-65.

Evans, M. J. and M. H. Kaufman (1981). "Establishment in culture of pluripotential cells from mouse embryos." Nature 292(5819): 154-6.

Feng, X. H. and R. Derynck (2005). "Specificity and versatility in tgf-beta signaling through Smads." Annu Rev Cell Dev Biol 21: 659-93.

Fuentealba, L. C., E. Eivers, A. Ikeda, C. Hurtado, H. Kuroda, E. M. Pera and E. M. De Robertis (2007). "Integrating patterning signals: Wnt/GSK3 regulates the duration of the BMP/Smad1 signal." Cell 131(5): 980-93.

Fujiwara, T., D. B. Dehart, K. K. Sulik and B. L. Hogan (2002). "Distinct requirements for extra-embryonic and embryonic bone morphogenetic protein 4 in the formation of the node and primitive streak and coordination of left-right asymmetry in the mouse." Development 129(20): 4685-96.

Fukumura, D., R. Xavier, T. Sugiura, Y. Chen, E. C. Park, N. Lu, M. Selig, G. Nielsen, T. Taksir, R. K. Jain and B. Seed (1998). "Tumor induction of VEGF promoter activity in stromal cells." Cell 94(6): 715-25.

Gadue, P., T. L. Huber, P. J. Paddison and G. M. Keller (2006). "Wnt and TGF-beta signaling are required for the induction of an in vitro model of primitive streak formation using embryonic stem cells." Proc Natl Acad Sci U S A 103(45): 16806-11.

Gluckman, E., H. A. Broxmeyer, A. D. Auerbach, H. S. Friedman, G. W. Douglas, A. Devergie, H. Esperou, D. Thierry, G. Socie, P. Lehn and 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(17): 1174-8.

Goldberg, J. L., M. J. Laughlin and V. J. Pompili (2007). "Umbilical cord blood stem cells: implications for cardiovascular regenerative medicine." J Mol Cell Cardiol 42(5): 912-20.

Page 98: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

85

Greco, T. L., S. Takada, M. M. Newhouse, J. A. McMahon, A. P. McMahon and S. A. Camper (1996). "Analysis of the vestigial tail mutation demonstrates that Wnt-3a gene dosage regulates mouse axial development." Genes Dev 10(3): 313-24.

Green, J. B., G. Howes, K. Symes, J. Cooke and J. C. Smith (1990). "The biological effects of XTC-MIF: quantitative comparison with Xenopus bFGF." Development 108(1): 173-83.

Green, J. B. and J. C. Smith (1990). "Graded changes in dose of a Xenopus activin A homologue elicit stepwise transitions in embryonic cell fate." Nature 347(6291): 391-4.

Hart, A. H., T. A. Willson, M. Wong, K. Parker and L. Robb (2005). "Transcriptional regulation of the homeobox gene Mixl1 by TGF-beta and FoxH1." Biochem Biophys Res Commun 333(4): 1361-9.

Hata, A., J. Seoane, G. Lagna, E. Montalvo, A. Hemmati-Brivanlou and J. Massague (2000). "OAZ uses distinct DNA- and protein-binding zinc fingers in separate BMP-Smad and Olf signaling pathways." Cell 100(2): 229-40.

Hebrok, M., S. K. Kim, B. St Jacques, A. P. McMahon and D. A. Melton (2000). "Regulation of pancreas development by hedgehog signaling." Development 127(22): 4905-13.

Henningfeld, K. A., S. Rastegar, G. Adler and W. Knochel (2000). "Smad1 and Smad4 are components of the bone morphogenetic protein-4 (BMP-4)-induced transcription complex of the Xvent-2B promoter." J Biol Chem 275(29): 21827-35.

Huang, H. C., L. C. Murtaugh, P. D. Vize and M. Whitman (1995). "Identification of a potential regulator of early transcriptional responses to mesoderm inducers in the frog embryo." EMBO J 14(23): 5965-73.

Huber, T. L., Y. Zhou, P. E. Mead and L. I. Zon (1998). "Cooperative effects of growth factors involved in the induction of hematopoietic mesoderm." Blood 92(11): 4128-37.

Inman, G. J., F. J. Nicolas, J. F. Callahan, J. D. Harling, L. M. Gaster, A. D. Reith, N. J. Laping and C. S. Hill (2002). "SB-431542 is a potent and specific inhibitor of transforming growth factor-beta superfamily type I activin receptor-like kinase (ALK) receptors ALK4, ALK5, and ALK7." Mol Pharmacol 62(1): 65-74.

Izumi, N., T. Era, H. Akimaru, M. Yasunaga and S. Nishikawa (2007). "Dissecting the molecular hierarchy for mesendoderm differentiation through a combination of embryonic stem cell culture and RNA interference." Stem Cells 25(7): 1664-74.

Izzi, L., C. Silvestri, I. von Both, E. Labbe, L. Zakin, J. L. Wrana and L. Attisano (2007). "Foxh1 recruits Gsc to negatively regulate Mixl1 expression during early mouse development." EMBO J 26(13): 3132-43.

Jaffredo, T., R. Gautier, A. Eichmann and F. Dieterlen-Lievre (1998). "Intraaortic hemopoietic cells are derived from endothelial cells during ontogeny." Development 125(22): 4575-83.

Page 99: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

86

Johansson, B. M. and M. V. Wiles (1995). "Evidence for involvement of activin A and bone morphogenetic protein 4 in mammalian mesoderm and hematopoietic development." Mol Cell Biol 15(1): 141-51.

Kanai-Azuma, M., Y. Kanai, J. M. Gad, Y. Tajima, C. Taya, M. Kurohmaru, Y. Sanai, H. Yonekawa, K. Yazaki, P. P. Tam and Y. Hayashi (2002). "Depletion of definitive gut endoderm in Sox17-null mutant mice." Development 129(10): 2367-79.

Katoh, M. (2007). "WNT signaling pathway and stem cell signaling network." Clin Cancer Res 13(14): 4042-5.

Kaufman, D. S., E. T. Hanson, R. L. Lewis, R. Auerbach and J. A. Thomson (2001). "Hematopoietic colony-forming cells derived from human embryonic stem cells." Proc Natl Acad Sci U S A 98(19): 10716-21.

Kennedy, M., S. L. D'Souza, M. Lynch-Kattman, S. Schwantz and G. Keller (2007). "Development of the hemangioblast defines the onset of hematopoiesis in human ES cell differentiation cultures." Blood 109(7): 2679-87.

Kim, S. K. and D. A. Melton (1998). "Pancreas development is promoted by cyclopamine, a hedgehog signaling inhibitor." Proc Natl Acad Sci U S A 95(22): 13036-41.

Korbling, M., P. Burke, H. Braine, G. Elfenbein, G. Santos and H. Kaizer (1981). "Successful engraftment of blood derived normal hemopoietic stem cells in chronic myelogenous leukemia." Exp Hematol 9(6): 684-90.

Kovacs, G. T. A., N. I. Maluf and K. E. Petersen (1998). "Bulk Micromachining of Silicon." Proceedings of the IEEE 86: 1536-1551.

Kretzschmar, M., J. Doody and J. Massague (1997). "Opposing BMP and EGF signalling pathways converge on the TGF-beta family mediator Smad1." Nature 389(6651): 618-22.

Kroon, E., L. A. Martinson, K. Kadoya, A. G. Bang, O. G. Kelly, S. Eliazer, H. Young, M. Richardson, N. G. Smart, J. Cunningham, A. D. Agulnick, K. A. D'Amour, M. K. Carpenter and E. E. Baetge (2008). "Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo." Nat Biotechnol 26(4): 443-52.

Kubo, A., K. Shinozaki, J. M. Shannon, V. Kouskoff, M. Kennedy, S. Woo, H. J. Fehling and G. Keller (2004). "Development of definitive endoderm from embryonic stem cells in culture." Development 131(7): 1651-62.

Labastie, M. C., F. Cortes, P. H. Romeo, C. Dulac and B. Peault (1998). "Molecular identity of hematopoietic precursor cells emerging in the human embryo." Blood 92(10): 3624-35.

Labbe, E., A. Letamendia and L. Attisano (2000). "Association of Smads with lymphoid enhancer binding factor 1/T cell-specific factor mediates cooperative signaling by the transforming growth factor-beta and wnt pathways." Proc Natl Acad Sci U S A 97(15): 8358-63.

Page 100: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

87

Labbe, E., C. Silvestri, P. A. Hoodless, J. L. Wrana and L. Attisano (1998). "Smad2 and Smad3 positively and negatively regulate TGF beta-dependent transcription through the forkhead DNA-binding protein FAST2." Mol Cell 2(1): 109-20.

Laflamme, M. A., K. Y. Chen, A. V. Naumova, V. Muskheli, J. A. Fugate, S. K. Dupras, H. Reinecke, C. Xu, M. Hassanipour, S. Police, C. O'Sullivan, L. Collins, Y. Chen, E. Minami, E. A. Gill, S. Ueno, C. Yuan, J. Gold and C. E. Murry (2007). "Cardiomyocytes derived from human embryonic stem cells in pro-survival factors enhance function of infarcted rat hearts." Nat Biotechnol 25(9): 1015-24.

Laping, N. J., E. Grygielko, A. Mathur, S. Butter, J. Bomberger, C. Tweed, W. Martin, J. Fornwald, R. Lehr, J. Harling, L. Gaster, J. F. Callahan and B. A. Olson (2002). "Inhibition of transforming growth factor (TGF)-beta1-induced extracellular matrix with a novel inhibitor of the TGF-beta type I receptor kinase activity: SB-431542." Mol Pharmacol 62(1): 58-64.

Lawson, K. A., J. J. Meneses and R. A. Pedersen (1986). "Cell fate and cell lineage in the endoderm of the presomite mouse embryo, studied with an intracellular tracer." Dev Biol 115(2): 325-39.

Liu, P., M. Wakamiya, M. J. Shea, U. Albrecht, R. R. Behringer and A. Bradley (1999). "Requirement for Wnt3 in vertebrate axis formation." Nat Genet 22(4): 361-5.

Lodish, H., A. Berk, P. Matsudaira, C. Kaiser, M. Krieger, M. Scott, S. Zipursky and J. Darnell (2004). Molecular Cell Biology. New York, Freeman.

Lowe, L. A., S. Yamada and M. R. Kuehn (2001). "Genetic dissection of nodal function in patterning the mouse embryo." Development 128(10): 1831-43.

Lyons, K. M., B. L. Hogan and E. J. Robertson (1995). "Colocalization of BMP 7 and BMP 2 RNAs suggests that these factors cooperatively mediate tissue interactions during murine development." Mech Dev 50(1): 71-83.

Maeno, M., P. E. Mead, C. Kelley, R. H. Xu, H. F. Kung, A. Suzuki, N. Ueno and L. I. Zon (1996). "The role of BMP-4 and GATA-2 in the induction and differentiation of hematopoietic mesoderm in Xenopus laevis." Blood 88(6): 1965-72.

Marshall, C. J., C. Kinnon and A. J. Thrasher (2000). "Polarized expression of bone morphogenetic protein-4 in the human aorta-gonad-mesonephros region." Blood 96(4): 1591-3.

Martin, G. R. (1981). "Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells." Proc Natl Acad Sci U S A 78(12): 7634-8.

Massague, J. (1990). "The transforming growth factor-beta family." Annu Rev Cell Biol 6: 597-641.

McBeath, R., D. M. Pirone, C. M. Nelson, K. Bhadriraju and C. S. Chen (2004). "Cell shape, cytoskeletal tension, and RhoA regulate stem cell lineage commitment." Dev Cell 6(4): 483-95.

Page 101: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

88

McGrath, K. E., A. D. Koniski, K. M. Maltby, J. K. McGann and J. Palis (1999). "Embryonic expression and function of the chemokine SDF-1 and its receptor, CXCR4." Dev Biol 213(2): 442-56.

McKendry, R., S. C. Hsu, R. M. Harland and R. Grosschedl (1997). "LEF-1/TCF proteins mediate wnt-inducible transcription from the Xenopus nodal-related 3 promoter." Dev Biol 192(2): 420-31.

McKiernan, E., L. O'Driscoll, M. Kasper, N. Barron, F. O'Sullivan and M. Clynes (2007). "Directed differentiation of mouse embryonic stem cells into pancreatic-like or neuronal- and glial-like phenotypes." Tissue Eng 13(10): 2419-30.

McLean, A. B., K. A. D'Amour, K. L. Jones, M. Krishnamoorthy, M. J. Kulik, D. M. Reynolds, A. M. Sheppard, H. Liu, Y. Xu, E. E. Baetge and S. Dalton (2007). "Activin a efficiently specifies definitive endoderm from human embryonic stem cells only when phosphatidylinositol 3-kinase signaling is suppressed." Stem Cells 25(1): 29-38.

Medvinsky, A. L., N. L. Samoylina, A. M. Muller and E. A. Dzierzak (1993). "An early pre-liver intraembryonic source of CFU-S in the developing mouse." Nature 364(6432): 64-7.

Millauer, B., S. Wizigmann-Voos, H. Schnurch, R. Martinez, N. P. Moller, W. Risau and A. Ullrich (1993). "High affinity VEGF binding and developmental expression suggest Flk-1 as a major regulator of vasculogenesis and angiogenesis." Cell 72(6): 835-46.

Moustakas, A. and C. H. Heldin (2007). "Signaling networks guiding epithelial-mesenchymal transitions during embryogenesis and cancer progression." Cancer Sci 98(10): 1512-20.

Murray, P. (1932). "The development in vitro of the blood of the early chick embryo." Proc Royal Soc London 11: 497-521.

Nagy, A., Gertsenstein, M., Vintersten, K., Behringer, R. (2003). Manipulating the Mouse Embryo. New York, Cold Spring Harbor Laboratory Press.

Nagy, A., J. Rossant, R. Nagy, W. Abramow-Newerly and J. C. Roder (1993). "Derivation of completely cell culture-derived mice from early-passage embryonic stem cells." Proc Natl Acad Sci U S A 90(18): 8424-8.

Naito, A. T., I. Shiojima, H. Akazawa, K. Hidaka, T. Morisaki, A. Kikuchi and I. Komuro (2006). "Developmental stage-specific biphasic roles of Wnt/beta-catenin signaling in cardiomyogenesis and hematopoiesis." Proc Natl Acad Sci U S A 103(52): 19812-7.

Nelson, C. M., R. P. Jean, J. L. Tan, W. F. Liu, N. J. Sniadecki, A. A. Spector and C. S. Chen (2005). "Emergent patterns of growth controlled by multicellular form and mechanics." Proc Natl Acad Sci U S A 102(33): 11594-9.

Ng, E. S., L. Azzola, K. Sourris, L. Robb, E. G. Stanley and A. G. Elefanty (2005). "The primitive streak gene Mixl1 is required for efficient haematopoiesis and BMP4-induced ventral mesoderm patterning in differentiating ES cells." Development 132(5): 873-84.

Page 102: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

89

Ng, E. S., R. P. Davis, L. Azzola, E. G. Stanley and A. G. Elefanty (2005). "Forced aggregation of defined numbers of human embryonic stem cells into embryoid bodies fosters robust, reproducible hematopoietic differentiation." Blood 106(5): 1601-3.

Nichols, J., B. Zevnik, K. Anastassiadis, H. Niwa, D. Klewe-Nebenius, I. Chambers, H. Scholer and A. Smith (1998). "Formation of pluripotent stem cells in the mammalian embryo depends on the POU transcription factor Oct4." Cell 95(3): 379-91.

Nishikawa, S. I., S. Nishikawa, M. Hirashima, N. Matsuyoshi and H. Kodama (1998). "Progressive lineage analysis by cell sorting and culture identifies FLK1+VE-cadherin+ cells at a diverging point of endothelial and hemopoietic lineages." Development 125(9): 1747-57.

Nishikawa, S. I., S. Nishikawa, H. Kawamoto, H. Yoshida, M. Kizumoto, H. Kataoka and Y. Katsura (1998). "In vitro generation of lymphohematopoietic cells from endothelial cells purified from murine embryos." Immunity 8(6): 761-9.

Niswander, L. and G. R. Martin (1992). "Fgf-4 expression during gastrulation, myogenesis, limb and tooth development in the mouse." Development 114(3): 755-68.

Norris, D. P., J. Brennan, E. K. Bikoff and E. J. Robertson (2002). "The Foxh1-dependent autoregulatory enhancer controls the level of Nodal signals in the mouse embryo." Development 129(14): 3455-68.

Nostro, M. C., X. Cheng, G. M. Keller and P. Gadue (2008). "Wnt, activin, and BMP signaling regulate distinct stages in the developmental pathway from embryonic stem cells to blood." Cell Stem Cell 2(1): 60-71.

Nusse, R. (2008). "Wnt signaling and stem cell control." Cell Res 18(5): 523-7.

Peerani, R., Bauwens, C., Kumacheva, E., Zandstra, P.W. (In Press). Patterning mouse and human embryonic stem cells using micro-contact printing. New Jersey, Humana Press.

Peerani, R., B. M. Rao, C. Bauwens, T. Yin, G. A. Wood, A. Nagy, E. Kumacheva and P. W. Zandstra (2007). "Niche-mediated control of human embryonic stem cell self-renewal and differentiation." EMBO J.

Pera, M. F., J. Andrade, S. Houssami, B. Reubinoff, A. Trounson, E. G. Stanley, D. Ward-van Oostwaard and C. Mummery (2004). "Regulation of human embryonic stem cell differentiation by BMP-2 and its antagonist noggin." J Cell Sci 117(Pt 7): 1269-80.

Perea-Gomez, A., W. Shawlot, H. Sasaki, R. R. Behringer and S. Ang (1999). "HNF3beta and Lim1 interact in the visceral endoderm to regulate primitive streak formation and anterior-posterior polarity in the mouse embryo." Development 126(20): 4499-511.

Perea-Gomez, A., F. D. Vella, W. Shawlot, M. Oulad-Abdelghani, C. Chazaud, C. Meno, V. Pfister, L. Chen, E. Robertson, H. Hamada, R. R. Behringer and S. L. Ang (2002). "Nodal antagonists in the anterior visceral endoderm prevent the formation of multiple primitive streaks." Dev Cell 3(5): 745-56.

Page 103: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

90

Pfister, S., K. A. Steiner and P. P. Tam (2007). "Gene expression pattern and progression of embryogenesis in the immediate post-implantation period of mouse development." Gene Expr Patterns 7(5): 558-73.

Piccolo, S., E. Agius, L. Leyns, S. Bhattacharyya, H. Grunz, T. Bouwmeester and E. M. De Robertis (1999). "The head inducer Cerberus is a multifunctional antagonist of Nodal, BMP and Wnt signals." Nature 397(6721): 707-10.

Pick, M., L. Azzola, A. Mossman, E. G. Stanley and A. G. Elefanty (2007). "Differentiation of human embryonic stem cells in serum-free medium reveals distinct roles for bone morphogenetic protein 4, vascular endothelial growth factor, stem cell factor, and fibroblast growth factor 2 in hematopoiesis." Stem Cells 25(9): 2206-14.

Rausa, F., U. Samadani, H. Ye, L. Lim, C. F. Fletcher, N. A. Jenkins, N. G. Copeland and R. H. Costa (1997). "The cut-homeodomain transcriptional activator HNF-6 is coexpressed with its target gene HNF-3 beta in the developing murine liver and pancreas." Dev Biol 192(2): 228-46.

Rivera-Perez, J. A. and T. Magnuson (2005). "Primitive streak formation in mice is preceded by localized activation of Brachyury and Wnt3." Dev Biol 288(2): 363-71.

Robertson, E. J., D. P. Norris, J. Brennan and E. K. Bikoff (2003). "Control of early anterior-posterior patterning in the mouse embryo by TGF-beta signalling." Philos Trans R Soc Lond B Biol Sci 358(1436): 1351-7; discussion 1357.

Rodaway, A. and R. Patient (2001). "Mesendoderm. an ancient germ layer?" Cell 105(2): 169-72.

Sabin, F. (1920). "Studies on the origin of blood vessels and of red corpuscles as seen in the living blastoderm of the chick during the second day of incubation." Contrib Embryol. 9: 213-262.

Saga, Y., N. Hata, S. Kobayashi, T. Magnuson, M. F. Seldin and M. M. Taketo (1996). "MesP1: a novel basic helix-loop-helix protein expressed in the nascent mesodermal cells during mouse gastrulation." Development 122(9): 2769-78.

Sakurai, H., T. Era, L. M. Jakt, M. Okada, S. Nakai and S. Nishikawa (2006). "In vitro modeling of paraxial and lateral mesoderm differentiation reveals early reversibility." Stem Cells 24(3): 575-86.

Sampath, T. K., K. E. Rashka, J. S. Doctor, R. F. Tucker and F. M. Hoffmann (1993). "Drosophila transforming growth factor beta superfamily proteins induce endochondral bone formation in mammals." Proc Natl Acad Sci U S A 90(13): 6004-8.

Sasaki, H. and B. L. Hogan (1993). "Differential expression of multiple fork head related genes during gastrulation and axial pattern formation in the mouse embryo." Development 118(1): 47-59.

Savage, R. and C. R. Phillips (1989). "Signals from the dorsal blastopore lip region during gastrulation bias the ectoderm toward a nonepidermal pathway of differentiation in Xenopus laevis." Dev Biol 133(1): 157-68.

Page 104: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

91

Shalaby, F., J. Rossant, T. P. Yamaguchi, M. Gertsenstein, X. F. Wu, M. L. Breitman and A. C. Schuh (1995). "Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice." Nature 376(6535): 62-6.

Shi, Y. and J. Massague (2003). "Mechanisms of TGF-beta signaling from cell membrane to the nucleus." Cell 113(6): 685-700.

Shiraki, N., T. Yoshida, K. Araki, A. Umezawa, Y. Higuchi, H. Goto, K. Kume and S. Kume (2008). "Guided Differentiation of ES Cells into Pdx1-Expressing Regional Specific Definitive Endoderm." Stem Cells.

Smith, J. C., B. M. Price, K. Van Nimmen and D. Huylebroeck (1990). "Identification of a potent Xenopus mesoderm-inducing factor as a homologue of activin A." Nature 345(6277): 729-31.

Smith, J. R., L. Vallier, G. Lupo, M. Alexander, W. A. Harris and R. A. Pedersen (2008). "Inhibition of Activin/Nodal signaling promotes specification of human embryonic stem cells into neuroectoderm." Dev Biol 313(1): 107-17.

Stafford, D., A. Hornbruch, P. R. Mueller and V. E. Prince (2004). "A conserved role for retinoid signaling in vertebrate pancreas development." Dev Genes Evol 214(9): 432-41.

Stavridis, M. P., J. S. Lunn, B. J. Collins and K. G. Storey (2007). "A discrete period of FGF-induced Erk1/2 signalling is required for vertebrate neural specification." Development 134(16): 2889-94.

Sulston, J. E., E. Schierenberg, J. G. White and J. N. Thomson (1983). "The embryonic cell lineage of the nematode Caenorhabditis elegans." Dev Biol 100(1): 64-119.

Tada, S., T. Era, C. Furusawa, H. Sakurai, S. Nishikawa, M. Kinoshita, K. Nakao and T. Chiba (2005). "Characterization of mesendoderm: a diverging point of the definitive endoderm and mesoderm in embryonic stem cell differentiation culture." Development 132(19): 4363-74.

Takada, S., K. L. Stark, M. J. Shea, G. Vassileva, J. A. McMahon and A. P. McMahon (1994). "Wnt-3a regulates somite and tailbud formation in the mouse embryo." Genes Dev 8(2): 174-89.

Takahashi, K., K. Tanabe, M. Ohnuki, M. Narita, T. Ichisaka, K. Tomoda and S. Yamanaka (2007). "Induction of pluripotent stem cells from adult human fibroblasts by defined factors." Cell 131(5): 861-72.

Tavian, M., L. Coulombel, D. Luton, H. S. Clemente, F. Dieterlen-Lievre and B. Peault (1996). "Aorta-associated CD34+ hematopoietic cells in the early human embryo." Blood 87(1): 67-72.

Tavian, M., M. F. Hallais and B. Peault (1999). "Emergence of intraembryonic hematopoietic precursors in the pre-liver human embryo." Development 126(4): 793-803.

Tavian, M. and B. Peault (2005). "Embryonic development of the human hematopoietic system." Int J Dev Biol 49(2-3): 243-50.

Page 105: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

92

Tavian, M., C. Robin, L. Coulombel and B. Peault (2001). "The human embryo, but not its yolk sac, generates lympho-myeloid stem cells: mapping multipotent hematopoietic cell fate in intraembryonic mesoderm." Immunity 15(3): 487-95.

Thomas, E. D. and R. Storb (1970). "Technique for human marrow grafting." Blood 36(4): 507-15.

Thomson, J. A., J. Itskovitz-Eldor, S. S. Shapiro, M. A. Waknitz, J. J. Swiergiel, V. S. Marshall and J. M. Jones (1998). "Embryonic stem cell lines derived from human blastocysts." Science 282(5391): 1145-7.

Ueno, S., G. Weidinger, T. Osugi, A. D. Kohn, J. L. Golob, L. Pabon, H. Reinecke, R. T. Moon and C. E. Murry (2007). "Biphasic role for Wnt/beta-catenin signaling in cardiac specification in zebrafish and embryonic stem cells." Proc Natl Acad Sci U S A 104(23): 9685-90.

Ulloa, L. and S. Tabibzadeh (2001). "Lefty inhibits receptor-regulated Smad phosphorylation induced by the activated transforming growth factor-beta receptor." J Biol Chem 276(24): 21397-404.

Ungrin, M. D., C. Joshi, A. Nica, C. Bauwens and P. W. Zandstra (2008). "Reproducible, ultra high-throughput formation of multicellular organization from single cell suspension-derived human embryonic stem cell aggregates." PLoS ONE 3(2): e1565.

Vincent, S. D., N. R. Dunn, S. Hayashi, D. P. Norris and E. J. Robertson (2003). "Cell fate decisions within the mouse organizer are governed by graded Nodal signals." Genes Dev 17(13): 1646-62.

Vodyanik, M. A., J. A. Bork, J. A. Thomson and Slukvin, II (2005). "Human embryonic stem cell-derived CD34+ cells: efficient production in the coculture with OP9 stromal cells and analysis of lymphohematopoietic potential." Blood 105(2): 617-26.

Watanabe, K., M. Ueno, D. Kamiya, A. Nishiyama, M. Matsumura, T. Wataya, J. B. Takahashi, S. Nishikawa, K. Muguruma and Y. Sasai (2007). "A ROCK inhibitor permits survival of dissociated human embryonic stem cells." Nat Biotechnol 25(6): 681-6.

Watanabe, M., M. L. Rebbert, M. Andreazzoli, N. Takahashi, R. Toyama, S. Zimmerman, M. Whitman and I. B. Dawid (2002). "Regulation of the Lim-1 gene is mediated through conserved FAST-1/FoxH1 sites in the first intron." Dev Dyn 225(4): 448-56.

Wells, J. M. and D. A. Melton (1999). "Vertebrate endoderm development." Annu Rev Cell Dev Biol 15: 393-410.

Wilson, M. E., D. Scheel and M. S. German (2003). "Gene expression cascades in pancreatic development." Mech Dev 120(1): 65-80.

Winnier, G., M. Blessing, P. A. Labosky and B. L. Hogan (1995). "Bone morphogenetic protein-4 is required for mesoderm formation and patterning in the mouse." Genes Dev 9(17): 2105-16.

Page 106: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

93

Xia, Y. and G. M. Whiteside (1998). "Soft Lithography." Angewandte Chemie International Edition 37: 550-575.

Xu, R. H., R. M. Peck, D. S. Li, X. Feng, T. Ludwig and J. A. Thomson (2005). "Basic FGF and suppression of BMP signaling sustain undifferentiated proliferation of human ES cells." Nat Methods 2(3): 185-90.

Yamaguchi, T. P., S. Takada, Y. Yoshikawa, N. Wu and A. P. McMahon (1999). "T (Brachyury) is a direct target of Wnt3a during paraxial mesoderm specification." Genes Dev 13(24): 3185-90.

Yang, L., M. H. Soonpaa, E. D. Adler, T. K. Roepke, S. J. Kattman, M. Kennedy, E. Henckaerts, K. Bonham, G. W. Abbott, R. M. Linden, L. J. Field and G. M. Keller (2008). "Human cardiovascular progenitor cells develop from a KDR(+) embryonic-stem-cell-derived population." Nature.

Yasunaga, M., S. Tada, S. Torikai-Nishikawa, Y. Nakano, M. Okada, L. M. Jakt, S. Nishikawa, T. Chiba and T. Era (2005). "Induction and monitoring of definitive and visceral endoderm differentiation of mouse ES cells." Nat Biotechnol 23(12): 1542-50.

Yu, J., M. A. Vodyanik, K. Smuga-Otto, J. Antosiewicz-Bourget, J. L. Frane, S. Tian, J. Nie, G. A. Jonsdottir, V. Ruotti, R. Stewart, Slukvin, II and J. A. Thomson (2007). "Induced pluripotent stem cell lines derived from human somatic cells." Science 318(5858): 1917-20.

Zafonte, B. T., S. Liu, M. Lynch-Kattman, I. Torregroza, L. Benvenuto, M. Kennedy, G. Keller and T. Evans (2007). "Smad1 expands the hemangioblast population within a limited developmental window." Blood 109(2): 516-23.

Zambidis, E. T., E. Oberlin, M. Tavian and B. Peault (2006). "Blood-forming endothelium in human ontogeny: lessons from in utero development and embryonic stem cell culture." Trends Cardiovasc Med 16(3): 95-101.

Zambidis, E. T., B. Peault, T. S. Park, F. Bunz and C. I. Civin (2005). "Hematopoietic differentiation of human embryonic stem cells progresses through sequential hematoendothelial, primitive, and definitive stages resembling human yolk sac development." Blood 106(3): 860-70.

Zambidis, E. T., L. Sinka, M. Tavian, V. Jokubaitis, T. S. Park, P. Simmons and B. Peault (2007). "Emergence of human angiohematopoietic cells in normal development and from cultured embryonic stem cells." Ann N Y Acad Sci 1106: 223-32.

Zhang, H. and A. Bradley (1996). "Mice deficient for BMP2 are nonviable and have defects in amnion/chorion and cardiac development." Development 122(10): 2977-86.

Zhang, P., J. Li, Z. Tan, C. Wang, T. Liu, L. Chen, J. Yong, W. Jiang, X. Sun, L. Du, M. Ding and H. Deng (2008). "Short-term BMP-4 treatment initiates mesoderm induction in human embryonic stem cells." Blood 111(4): 1933-41.

Zheng, Y., N. Watanabe, T. Nagamura-Inoue, K. Igura, H. Nagayama, A. Tojo, R. Tanosaki, Y. Takaue, S. Okamoto and T. A. Takahashi (2003). "Ex vivo manipulation of umbilical

Page 107: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

94

cord blood-derived hematopoietic stem/progenitor cells with recombinant human stem cell factor can up-regulate levels of homing-essential molecules to increase their transmigratory potential." Exp Hematol 31(12): 1237-46.

Zhou, S., K. Eid and J. Glowacki (2004). "Cooperation between TGF-beta and Wnt pathways during chondrocyte and adipocyte differentiation of human marrow stromal cells." J Bone Miner Res 19(3): 463-70.

Page 108: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

95

8 Appendix

8.1 Other Signalling Pathways Possibly Regulating Mesendoderm

Differentiation

As discussed above, it is possible that other signalling pathways besides the TGFβ pathway are

involved in regulating mesendoderm differentiation. During the course of this project, the Rho

kinase and FGF signalling pathway were also investigated but the results from these

investigations were not included in the main body of this thesis because they did not significantly

contribute to the ideas or conclusions presented. However, these results are presented in the

appendix for future consideration.

8.1.1 Effect of Rho Kinase Inhibitor Y-27632 on Mesendoderm

Differentiation

Although it is proposed here that differential Smad1 activation is responsible for specifying

mesoderm differentiation at the expense of endoderm, it is possible that other factors such as cell

spreading or cell-cell contact also mediate this effect. It has been previously shown that cell

shape and cytoskeletal tension regulate the differentiation of human mesenchymal stem cells

towards adipocyte or osteoblast cell types and this process is mediated through RhoA activation

(McBeath et al. 2004). Through constitutive activation or inhibition of the RhoA effector, Rho

kinase, they were able to induce osteogenesis or adipogenesis, respectively, independent of cell

shape. A similar approach can be used here to investigate if cell shape or cytoskeletal tension

processes mediated by RhoA activation play a role in mesendoderm differentiation. Preliminary

studies were conducted by concomitantly treating micropatterned cultures with BMP2, Activin A

and the Rho kinase inhibitor Y-27632 (10 nM). These studies showed that the Rho kinase

Page 109: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

96

inhibitor increases Bry expression in large colonies but has little or no effect on smaller colonies

suggesting that the mechanism governing mesendoderm differentiation may be related to RhoA

activation (Figure 8-1). However, the Rho kinase inhibitor has also been shown to increase

survival (Watanabe et al. 2007), decrease Smad1 activation, and increase self-renewal of hESC

(Peerani et al. 2007). The many known interactions of the Rho signalling pathway with various

other pathways precludes any conclusions to be drawn here as to the mechanism governing Rho

kinase activation and mesoderm differentiation. Further studies are required in order to better

understand the effects of Rho signalling on hESC fate.

Figure 8-1 Representative immunofluorescent images of Bry expression of micropatterned colonies following 48 hours of treatment with BMP2 and Activin A with or without Y-27632 ROCK inhibitor. Original magnification is 10×, scale bar indicates 500 µm.

8.1.2 Effect of Fibroblast Growth Factor Inhibition on Mesendoderm Differentiation

Fibroblast growth factor (FGF) signalling has been previously implicated in maintaining

pluripotency of hESC (Thomson et al. 1998; Xu et al. 2005) as well as ectoderm differentiation

Page 110: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

97

(Stavridis et al. 2007) but its role in mesendoderm differentiation remains unclear. Therefore,

inhibition of FGF signalling through pharmacological inhibition of the FGF receptor (FGFR) 1

and inhibition of MAP kinase was investigated using the small molecules PD173074 (FGFRi, 20

nM) and PD184352 (MAPKi, 500 nM), respectively. It was found that inhibition of FGFR1 or

MAP kinase during the initial 48 hours of differentiation in the presence of BMP2 and Activin A

was able to significantly increase Bry expression in the larger 1200 µm colonies but not the

smaller 200 µm colonies (Figure 8-2). Protein expression levels were further confirmed through

quantitative gene expression analysis (Figure 8-3). As previously mentioned, FGF signalling is

able to modulate Smad signalling through phosphorylation of receptor Smads in the linker region

by MAP kinase which prevents its nuclear accumulation and transcriptional activity.

Figure 8-2 Representative immunofluorescent images of Bry expression following 48 hours of treatment with BMP2, Activin A, FGFR inhibitor PD173074 (20 nM), or MAP kinase inhibitor PD184352 (500 nM) as indicated. Original magnfication is 10×, scale bar indicates 500 µm.

Page 111: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

98

Bry KDR

Rel

ativ

e E

xpre

ssio

n0

1

2

3

4

5

6

7

200 µm B+A200 µm B+A+FGFRi1200 µm B+A1200 µm B+A+FGFRi

*

Figure 8-3 Quantitative gene expression analysis of micropatterned colonies following 48 hours of treatment with BMP2, Activin A, and FGFR inhibitor PD173074 (20 nM) as indicated. (*) indicates p<0.05.

To determine if this was the mechanism through which FGF inhibition acts, the levels of pSmad1

were measured following 90 minutes of treatment with BMP2, Activin A and the MAP kinase

inhibitor PD184352 (500 nM). Quantitative immunofluorescent analysis demonstrates that

pSmad1 activation is greater when MAP kinase is inhibited supporting the role of FGF signalling

attenuating Smad1 activation and decreasing mesoderm differentiation (Figure 8-4). However,

when the hematopoietic assay was applied to FGF inhibited cultures, there was a striking

decrease in hematopoietic colony forming frequency (Figure 8-5). As a result, it is unclear what

role FGF signalling plays in mesoderm or hematopoietic specification. It is possible that FGF

inhibition does not increase mesoderm differentiation, or FGF inhibition only inhibits

hematopoietic development at a very early stage.

Page 112: Controlling Cell Density by Micropatterning Regulates Smad ... · This report demonstrates ... 1.2.3.2.3 From Mouse to Human Embryonic Stem Cells ... 2 SPECIFIC AIM #1:

99

200

µm B

+A

200 µ

m B+A

+MAPKi

1200

µm B

+A

1200

µm B

+A+M

APKi

Rel

ativ

e Fl

uore

scen

t Int

ensi

ty0.0

0.5

1.0

1.5

2.0

2.5

3.0pSmad1

*

Figure 8-4 Quantitative immunofluorescent analysis of pSmad1 intensity of micropatterned colonies following 90 minute treatment with BMP2, Activin A, and MAP kinase inhibitor PD184352 (500 nM) as indicated. (*) indicates p<0.05.

1200

µm

1200

µm +

FGFRi

# C

olon

ies

per 1

05 c

ells

0

5

10

15

20

25

30

Figure 8-5 Frequency of hematopoietic colonies following hematopoietic differentiation assay of 1200 µm cultures with or without FGFR1 inhibition.