reprogramming mouse glioma stem cells with defined · pdf filereprogramming mouse glioma stem...

121
Reprogramming Mouse Glioma Stem Cells with Defined Factors by Julia Alexandra Maria DiLabio A thesis submitted in conformity with the requirements for the degree of Master of Science Laboratory Medicine and Pathobiology University of Toronto © Copyright by Julia A.M. DiLabio 2011

Upload: lythuy

Post on 09-Mar-2018

223 views

Category:

Documents


5 download

TRANSCRIPT

Page 1: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

Reprogramming Mouse Glioma Stem Cells with Defined Factors

by

Julia Alexandra Maria DiLabio

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

Laboratory Medicine and Pathobiology University of Toronto

© Copyright by Julia A.M. DiLabio 2011

Page 2: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

ii

Reprogramming Mouse Glioma Stem Cells with Defined Factors

Julia A.M. DiLabio

Master of Science

Laboratory Medicine and Pathobiology

University of Toronto

2011

Abstract

This thesis shows that p53-deficient mouse glioma brain tumour stem cells (BTSCs), which fail

to express pluripotency factors, can be reprogrammed with specific transcription factors to

generate iPS cell lines (GNS-iPS) expressing endogenous pluripotency factors (Nanog, Oct4, and

Rex1). GNS-iPS cell lines formed embryoid bodies (EBs) in vitro and undifferentiated growths

in vivo that phenotypically did not resemble tumours derived from non-reprogrammed BTSCs.

EBs formed from one GNS-iPS cell line expressed markers of mesoderm, endoderm, and

ectoderm. Tumours produced from GNS-iPS cells had reduced astrocytic marker (GFAP)

expression compared to those generated from control iPS cell lines or non-reprogrammed

BTSCs. Preliminary results suggest that the reprogrammed cells can be re-differentiated into

cells that show neural precursor phenotype. These findings suggest that BTSCs can acquire

aspects of the pluripotent state with a defined set of transcription factors, opening the door for

further exploration of reprogramming strategies to attenuate the cancer phenotype.

Page 3: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

iii

Acknowledgements

I owe a great deal of gratitude to all members of the Dirks Lab and colleagues at the Hospital for

Sick Children for advice and support throughout my MSc candidacy. I am particularly grateful

for Dr. Peter Dirks’ mentorship, unwavering support, and contagious enthusiasm for science and

medicine. I am also thankful for the scientific guidance and support from Dr. Ian Clarke, Dr.

Steven Pollard, and advisory committee members Dr. James Ellis and Dr. Derek van der Kooy.

I would like to acknowledge the work of Dr. Ryan Ward, who established and

characterized the mouse glioma stem cell lines, Lilian Lee, who carried out mouse injections and

helped to process tumours, Dr. Akitsu Hotta, who produced and shared the EOS lentivirus, and

Natalie Farra, who provided experimental guidance in generating and characterizing mouse

induced pluripotent stem cell lines.

I am also eternally grateful for the love and support of family and friends. This body of

work is dedicated to my parents, who have always been there for me every step of the way. They

have helped me to learn that happiness and success are not found at the end of the road, but are

experienced along the way.

Page 4: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

iv

Table of Contents

Acknowledgements ........................................................................................................................ iii

Table of Contents ........................................................................................................................... iv

List of Abbreviations ................................................................................................................... viii

List of Tables ................................................................................................................................ xii

List of Figures .............................................................................................................................. xiii

List of Appendices ........................................................................................................................ xv

Chapter 1 ......................................................................................................................................... 1

1 Introduction ................................................................................................................................ 1

1.1 Introduction to Stem Cells .................................................................................................. 1

1.1.1 Adult Stem Cells ..................................................................................................... 1

1.1.2 Neural Stem Cells ................................................................................................... 2

1.1.3 Embryonic Stem Cells ............................................................................................ 2

1.2 Pluripotency ........................................................................................................................ 4

1.3 Nuclear Reprogramming ..................................................................................................... 7

1.3.1 Nuclear Transfer ..................................................................................................... 7

1.3.2 Cell Fusion .............................................................................................................. 9

1.3.3 Transcription Factor-Mediated Reprogramming .................................................. 10

1.3.4 Induced Pluripotent Stem Cell Technology .......................................................... 11

1.3.5 Improving iPS Cell Technology ........................................................................... 15

1.3.6 Induced Pluripotent Stem Cells Generated from Neural Stem Cells .................... 16

1.4 Reprogramming and Cancer ............................................................................................. 17

1.4.1 Introduction to Epigenetics ................................................................................... 17

1.4.2 Epigenetic Contribution to Cancer ........................................................................ 19

1.4.3 Chemotherapy Targeting Epigenetic Modifications in Cancer ............................ 21

Page 5: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

v

1.4.4 Pluripotency and Cancer ....................................................................................... 22

1.4.5 Nuclear Reprogramming of Cancer Cells ............................................................. 24

1.5 Cancer Stem Cells ............................................................................................................. 25

1.5.1 The Cancer Stem Cell Hypothesis ........................................................................ 26

1.6 Brain Tumours .................................................................................................................. 29

1.6.1 Human Glioma ...................................................................................................... 29

1.6.2 Mouse Glioma ....................................................................................................... 32

1.6.3 Brain Tumour Stem Cells ..................................................................................... 33

1.7 Thesis Rationale, Hypothesis, and Specific Aims ............................................................ 35

1.7.1 Rationale ............................................................................................................... 35

1.7.2 Hypothesis ............................................................................................................. 35

1.7.3 Specific Aims ........................................................................................................ 36

Chapter 2 ....................................................................................................................................... 37

2 Materials and Methods ............................................................................................................. 37

2.1 Characterization of Mouse Glioma Stem Cell Lines ........................................................ 37

2.1.1 Mouse Glioma Stem Cells .................................................................................... 37

2.1.2 Cell Culture and Differentiation ........................................................................... 37

2.1.3 RNA Isolation and Reverse-Transcriptase PCR ................................................... 37

2.1.4 Alkaline Phosphatase Staining .............................................................................. 38

2.1.5 MTT Assay ........................................................................................................... 38

2.2 Mouse iPS Cell Induction ................................................................................................. 39

2.2.1 EOS Lentiviral Infection ....................................................................................... 39

2.2.2 Retrovirus Production and Infection ..................................................................... 39

2.2.3 Isolation and Maintenance of iPS Cell Colonies .................................................. 40

2.3 Molecular Characterization of iPS Cell Lines .................................................................. 41

2.3.1 Immunocytochemistry .......................................................................................... 41

Page 6: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

vi

2.3.2 Quantitative RT-PCR ............................................................................................ 41

2.4 In Vitro Differentiation ..................................................................................................... 41

2.4.1 Embryoid Body Assays ......................................................................................... 41

2.4.2 Immunocytochemistry for Germ Layer Markers .................................................. 42

2.5 In Vivo Differentiation ...................................................................................................... 42

2.5.1 Teratoma Assays ................................................................................................... 42

2.5.2 Histology and Immunohistochemistry .................................................................. 43

2.6 Directed Differentiation to Neural Stem Cells .................................................................. 43

Chapter 3 ....................................................................................................................................... 45

3 Results ...................................................................................................................................... 45

3.1 Characterization of Mouse Glioma Stem Cell Lines ........................................................ 45

3.1.1 Mouse Glioma Stem Cells Express Neural Stem Cell Markers, but Not Core

Pluripotency Markers ............................................................................................ 45

3.1.2 Mouse Glioma Stem Cells Express Alkaline Phosphatase ................................... 47

3.1.3 Serum Alters the Growth Rate of Glioma and Normal Neural Stem Cells .......... 47

3.2 Defined Factors Can Reprogram Mouse Glioma Stem Cells ........................................... 49

3.2.1 Induction of Reprogramming with Four Transcription Factors ............................ 49

3.2.2 Induced Pluripotent Stem Cell Lines Express Pluripotency Factors, but Not

Neural Stem Cell or Astrocytic Cell Markers ....................................................... 51

3.2.3 Retroviral Silencing in Reprogrammed Cell Lines ............................................... 55

3.3 Altered Differentiation Potential of p53-Deficient iPS Cells ........................................... 57

3.3.1 Induced Pluripotent Stem Cell Lines Generate Embryoid Bodies with Reduced

Endodermal Differentiation in vitro ...................................................................... 57

3.4 p53-Deficient iPS Cell Lines Generate Highly Undifferentiated Tumours ..................... 61

3.5 Reprogrammed Glioma-Derived iPS Cell Lines Have Reduced Astrocytic

Differentiation in vivo ....................................................................................................... 65

3.6 Re-Differentiation of Glioma-Derived iPS Cell Lines to a Neural Stem Cell Fate .......... 67

Page 7: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

vii

Chapter 4 ....................................................................................................................................... 69

4 Discussion and Future Directions ............................................................................................ 69

4.1 Summary of Principle Findings ........................................................................................ 69

4.2 Why Reprogram Brain Tumour Stem Cells? .................................................................... 69

4.3 Mouse Glioma Stem Cells are Amenable to Reprogramming .......................................... 71

4.4 Reprogrammed Cell Lines Show Varying Degrees of Retroviral Silencing .................... 72

4.5 Analysis of the Differentiation Capacity of iPS Cell Lines .............................................. 73

4.6 Interplay between Retroviral Silencing, p53 Status, and In Vivo Differentiation

Potential ............................................................................................................................ 76

4.7 Reprogramming Mouse Versus Human Cells .................................................................. 77

4.8 Defining the Reprogrammed State .................................................................................... 78

4.9 Core Pluripotency Network Versus c-Myc Network in Cancer ....................................... 80

4.10 Expression of Pluripotency Factors in Human Glioma.................................................... 82

4.11 Future Directions .............................................................................................................. 84

Appendices .................................................................................................................................... 86

5 Appendix 1. Supplemental Methods ....................................................................................... 86

5.1 Primer Sequences .............................................................................................................. 86

6 Appendix 2. Supplemental Figures ......................................................................................... 88

6.1 Quantitative RT-PCR Results of Pluripotency Factor Expression ................................... 88

6.2 Alkaline Phosphatase Expression in Mouse Glioma Stem Cell Lines ............................. 89

6.3 Nanog Expression in MEF-Derived iPS Cell Lines ......................................................... 91

References ..................................................................................................................................... 92

Page 8: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

viii

List of Abbreviations

2i Dual inhibition

ABL v-abl Abelson murine leukemia viral oncogene homolog 1

Akt v-akt murine thymoma viral oncogene homolog/Protein kinase B

AP Alkaline phosphatase

APC Adenomatous polyposis coli

Ascl1 Achaete-scute homolog 1/Mash1

ATM Ataxia telangiectasia mutated

AZA 5-azacytidine

BCR Breakpoint cluster region

bFGF Basic fibroblast growth factor

Bmi1 Bmi1 polycomb ring finger oncogene

BMP Bone morphogenetic protein

Brn2 Pou3f2

BTSC Brain tumour stem cell

CD Cluster of differentiation

CD133 AC133/CD133/Prominin1

CD15 LewisX/CD15/Stage Specific Embryonic Antigen1

CDK Cyclin-dependent kinase

CDKN Cyclin-dependent kinase inhibitor

CML Chronic myeloid leukemia

CNPase 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase

CNS Central nervous system

CpG Poly cytosine-guanine

Cre Cre-recombinase

CSC Cancer stem cell

DMR Differentially methylated regions

DNMT DNA methyltransferase

e Embryonic day

EB Embryoid body

EC Embryonal carcinoma

Page 9: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

ix

EG Embryonic germ

EGF Epidermal growth factor

EGFR Epidermal growth factor receptor

ENU N-ethyl-N-nitrosourea

EOS Early transposon promoter and Oct4 and Sox2 enhancers

ER Estrogen receptor

ES Embryonic stem

Exo Exogenous

EZH2 Enhancer of zeste homologue 2

FBS Fetal bovine serum

Fbx15 F-box-containing protein/Fbxo15

FDA Food and Drug Administration

Flk1 Fetal liver kinase 1

FZD9 Frizzled-9

GBM Glioblastoma multiforme

GFAP Glial fibrillary acidic protein

GI Gastrointestinal

Glis1 Gli-similar protein 1

GSK3 Glycogen synthase kinase 3

GSTP3 Glutathione S-transferase pi 1

H&E Hematoxylin and eosin

HDAC Histone deacetylase

HOXA11 Homeobox A11

IDH1 Isocitrate dehydrogenase 1

Ink4/Arf Encodes p15INK4b

, ARF and p16INK4a

iPS Induced pluripotent stem

Jak Janus kinase

Klf4 Krüppel-like factor 4

LIF Leukemia inhibitory factor

LOH Loss of heterozygosity

Map2 Microtubule-associated protein 2

MAPK Mitogen-activated protein kinase

Page 10: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

x

MB Medulloblastoma

MDM2 Mdm2 p53 binding protein homolog

MeCP2 Methyl CpG binding protein 2

MEF Mouse embryonic fibroblast

miRNA Micro RNA

MEST Mesoderm-specific transcript homolog

MGMT O6-methylguanine DNA methyltransferase

MLH1 MutL homolog 1

MLL1 Mixed-lineage leukemia 1

MTT 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide

Myc Myelocytomatosis viral oncogene homolog

MyoD Myoblast determination protein

Mytl1 Myelin transcription factor 1-like

NF1 Neurofibromin 1

NOD Non-obese diabetic

NSC Neural stem cell

Oct4 Octamer-binding transcription factor 4/Pou5f1

p16INK4A

Cyclin-dependent kinase inhibitor 2A

p21 Cyclin-dependent kinase inhibitor 1/Waf1/Cip1

p53 Tumour protein 53

Pax6 Paired box gene 6

PcG Polycomb group

PDGFRA Alpha-type platelet-derived growth factor receptor

PGC Primordial germ cell

PI3K Phosphoinositide-3-kinase

PLO Poly-L-ornithine

PNET Primitive neuroectodermal tumour

PRC Polycomb repressive complex

Ptc1 Patched1

PTEN Phosphatase and tensin homolog

Ras Rat sarcoma

RASSF1A Ras association domain family 1 isoform A

Page 11: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

xi

RB1 Retinoblastoma 1

Rex1 Zinc finger protein 42/Zfp42

RT-PCR Reverse transcriptase polymerase chain reaction

S100β S100 calcium binding protein β

SCID Severe combined immune-deficient

SCNT Somatic cell nuclear transfer

SGZ Subgranular zone

SKP Skin-derived precursor

Sox2 SRY-box containing gene 2

SSEA-1 Stage-specific embryonic antigen 1

Stat Signal transducer and activator of transcription

Tbx3 T-box transcription factor 3

TES Testis derived transcript

TIMP3 Metalloproteinase inhibitor 3

TNFRSF10A Tumor necrosis factor receptor superfamily member 10A

SVZ Subventricular zone

Tgfβ Transforming growth factor β

TP53 Tumour protein 53 (human gene)

VPA Valproic acid

VHL Von Hippel-Lindau

WHO World Health Organization

Wnt Wingless

Page 12: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

xii

List of Tables

Table 1. Summary of the isolation of iPS cell colonies derived from four-factor inductions

and the number of clones derived ………………………………………………..... 50

Table 2. Summary of the latency of subcutaneous and intracranial tumour formation …... 62

Supplemental Table 1. Primer sequences used in this study …………………………….... 86

Page 13: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

xiii

List of Figures

Figure 1. The core signaling pathways and transcription factors regulating mouse ES cell

pluripotency. ............................................................................................................................... 6

Figure 2. Nuclear reprogramming to pluripotency by nuclear transfer, cell fusion, and

transcription factor transduction. ............................................................................................. 13

Figure 3. A comparison of the stochastic and cancer stem cell models of cancer. ...................... 28

Figure 4. Mouse glioma stem cell lines express neural stem cell-associated factors, but do

not express RNA encoding core pluripotency-associated factors. ........................................... 46

Figure 5. Serum alters the growth rate of glioma stem cell and neural stem cell lines. .............. 48

Figure 6. Molecular characterization of iPS cell lines reveals the expression of core

pluripotency-associated factors. ............................................................................................... 52

Figure 7. Reprogrammed neural stem cells and glioma stem cells fail to express neural stem

cell or astrocytic cell markers. ................................................................................................. 54

Figure 8. Analysis of retroviral vector expression in iPS cell lines. ............................................. 56

Figure 9. Glioma stem cell-derived iPS cell lines form EBs and generate cells of mesodermal

and ectodermal lineages in vitro, as detected by immunostaining. .......................................... 59

Figure 10. Differentiated cell markers are detectable at the transcript level in undifferentiated

iPS and ES cell populations, but are enhanced during EB differentiation. .............................. 60

Figure 11. Reprogrammed NSCs and glioma stem cells generate subcutaneous tumours with

similar macroscopic appearance. ............................................................................................. 63

Figure 12. Glioma stem cell-derived iPS cell lines generate highly undifferentiated tumours

in vivo. ...................................................................................................................................... 64

Figure 13. Glioma stem cell-derived iPS cell lines generate tumours expressing mesodermal

and ectodermal cell markers, but have significantly reduced astrocytic differentiation. ......... 66

Figure 14. Preliminary directed re-differentiation experiments generated Nestin+ cells with

neural stem cell-like morphology. ............................................................................................ 68

Figure S1. Glioma stem cell lines express L-Myc and neural stem cell-associated factors, but

do not express Nanog or Oct4. ................................................................................................. 88

Figure S2. Glioma stem cells, but not normal neural stem cells, express alkaline phosphatase

in stem cell conditions. ............................................................................................................. 89

Page 14: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

xiv

Figure S3. Glioma stem cells and normal neural stem cells fail to express alkaline

phosphatase in differentiation conditions................................................................................. 90

Figure S4. MEF-derived iPS cell lines express Nanog and 2i treatment enhances Nanog

expression. ................................................................................................................................ 91

Page 15: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

xv

List of Appendices

Appendix 1. Supplemental Methods …………………………………………………….... 86

Appendix 2. Supplemental Figures ……………………………………………………...... 88

Page 16: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

1

Chapter 1

1 Introduction

1.1 Introduction to Stem Cells

Stem cells are involved in embryonic and fetal development as well as tissue repair and

maintenance in adult organisms. These cells have the intrinsic ability to self-renew to regenerate

more stem cells and differentiate to produce mature, lineage-restricted cell types. In

development, pluripotent stem cells give rise to the cells of the three germ layers (endoderm,

mesoderm, and ectoderm) of a fetal organism. Multipotent stem cells are lineage-restricted and

generate specialized cell types residing within a tissue. The neural stem cell is an example of a

multipotent stem cell that differentiates to produce the cell types comprising the nervous system:

neurons, astrocytes, and oligodendrocytes. Multipotent stem cells persist throughout an

organism’s lifetime and are responsible for cellular regeneration and tissue replenishment.

1.1.1 Adult Stem Cells

The most restricted type of stem cell is the multipotent stem cell, which produces differentiated

cells of the tissue type in which it resides. Stem cell activity occurs not only during fetal

development, but also in adult tissues, when multipotent stem cells are required for the

maintenance of the tissues under normal conditions and repair after injury. Normal tissue

turnover occurs at significantly different rates, depending on the tissue type; brain and skeletal

muscle have relatively little turnover, whereas the highest turnover rates occurs in the intestines

and skin, which are replenished approximately every 5 days and 4 weeks, respectively [1]. In

tissues with little turnover, such as the brain, it was initially thought that stem cells were not

present and the generation of new neurons (neurogenesis) ceased just after birth, but the

identification of proliferating cells based on labeling experiments with tritiated thymidine in the

adult rat brain suggested otherwise [2]. Self-renewing, lineage-restricted neural stem cells

(NSCs) were later identified in the adult mammalian brain, and neurogenesis was shown to occur

not only during development, but also in the adult brain [3,4,5].

Page 17: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

2

1.1.2 Neural Stem Cells

The sites of adult neurogenesis, where adult NSCs are found, are the subventricular zone (SVZ)

lining the lateral ventricles and the subgranular zone (SGZ) of the dentate gyrus in the

hippocampus [6,7]. The regulation of proliferation and differentiation is mediated by the stem

cell niche – factors such as the microenvironment that provides extrinsic signals to the stem cell

and includes soluble growth factors, hormones, and contact with other cells and the extracellular

matrix [8]. Soluble factors such as epidermal growth factor (EGF) and basic fibroblast growth

factor (bFGF) were identified as potent mitogens regulating NSC proliferation and

differentiation in vivo; both growth factors enhanced neural stem cell self-renewal, but EGF

promoted the generation of glial cells and prevented neurogenesis, whereas bFGF promoted

neurogenesis [9,10]. These important mitogens are included in standard neural stem cell culture

conditions in vitro, allowing self-renewing, multipotent NSCs to be expanded in defined, serum-

free medium as spherical aggregates (neurospheres) or as an adherent monolayer on a substrate

such as laminin [3,11].

NSCs are among the best-characterized multipotent stem cells in terms of cell-surface

and intracellular marker expression; human NSCs can be identified by the cell-surface marker

CD133 (Prominin-1) and the intracellular expression of the neuroepithelial stem cell marker

Nestin and the stem cell-associated transcription factor Sox2 [3,12]. Mouse NSCs can also be

identified by these markers, but express the cell-surface carbohydrate moiety CD15 instead of

CD133 [13]. Multipotent NSCs are restricted in their differentiation capacity and generate

neurons, astrocytes, and oligodendrocytes, the differentiated cells of the mammalian CNS in

vitro and in vivo.

1.1.3 Embryonic Stem Cells

Embryonic stem (ES) cells are cultured cells that are derived from the inner cell mass of the

blastocyst after fertilization. These cells are defined as pluripotent stem cells and have the ability

to differentiate into cells of the three germ layers (endoderm, mesoderm, and ectoderm). When

aggregated with blastocysts, ES cells cannot generate extraembryonic tissue, such as trophoblast

cells that form the placenta, but give rise to cells and tissues of the endoderm (gastrointestinal

tract, lungs, stomach lining), mesoderm (muscle, bone, blood), and ectoderm (skin and the

nervous system).

Page 18: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

3

Human and mouse ES cells are isolated from the developing mammalian blastocyst at

embryonic day (e) 4-5 and can be grown in vitro in conditions promoting the maintenance of

pluripotency [14,15]. Standard mouse ES cell culture conditions include medium containing

serum (providing bone morphogenetic protein, or BMP) and the cytokine leukemia inhibitory

factor (LIF) [16,17]. Mouse ES cells are often grown on a mitotically-inactivated “feeder” layer

of cells, typically consisting of mouse embryonic fibroblasts (MEFs), which help to support the

growth of pluripotent ES cell colonies and secrete growth factors and cytokines [17,18].

Undifferentiated mouse ES cells can also be propagated in feeder-free conditions on a substrate

such as gelatin in the presence of LIF or MEF-conditioned culture medium [17,18]. Extrinsic

cytokine signaling, particularly mediated by LIF, counteracts the intrinsic tendency of ES cells to

differentiate [19]. LIF activates the Jak-Stat3 pathway in mouse ES cells, which directly

increases the expression of Klf4 and indirectly upregulates the expression of Sox2, as well as the

PI3K-Akt pathway, which indirectly enhances Nanog expression via the transcription factor

Tbx3 (Figure 1A) [19,20]. Kfl4 is a direct target of LIF signaling and binds to the promoter of

the Nanog gene, thereby regulating its expression [21]. The self-renewal of ES cells in an

undifferentiated state can be maintained in the absence of LIF by suppressing differentiation-

inducing signaling from mitogen-activated protein kinase (MAPK) and glycogen synthase kinase

3 (GSK3) using a cocktail of dual inhibition (2i) chemical inhibitors [22].

ES cells can self-renew indefinitely and exhibit tri-lineage differentiation in vitro, thus

recapitulating the characteristics of the cells of the inner cell mass in vivo. However, mouse ES

cells can be induced to differentiate in vitro into cells of extraembryonic origin, such as

extraembryonic endoderm (yolk sac) or trophectoderm (placenta) by knockdown of Nanog or

Oct4, respectively [23,24]. Mouse ES cells express cell-surface markers, such as stage-specific

embryonic antigen-1 (SSEA-1, or SSEA-4 in the case of human ES cells) and express the

transcription factors Nanog, Oct4, and Rex1 that cooperatively maintain pluripotency.

Functional pluripotency of mammalian ES cells is defined by in vitro embryoid body (EB)

formation and in vivo teratoma formation. EBs are spherical aggregates which form when

differentiated ES cells are grown in the absence of LIF. The EBs contain cells that express

markers of endodermal, mesodermal, and ectodermal lineages. Teratoma assays involve the

injection of ES cells into immune-compromised mice, typically in the kidney capsule, testes, or

Page 19: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

4

dorsal flank, and the subsequent identification of three germ layers by the detection of

characteristic histological structures that also express appropriate linage markers.

1.2 Pluripotency

A core transcription factor network involved in the maintenance of pluripotency has been

identified in human and mouse ES cells, which consists of the transcription factors Nanog, Oct4,

and Sox2 [25,26]. These factors form a cooperative regulatory loop that maintains the

pluripotent phenotype by binding to promoter regions of target genes, including each other

(Figure 1B). These transcription factors activate the expression of genes required for the

maintenance of pluripotency and self-renewal, including members of the Tgfβ and Wnt signaling

pathways [25]. Sox2 cooperates with Nanog, which binds to the promoter region and activates

the expression of Rex1, a transcription factor used as a marker of pluripotency in mouse ES cells

[27]. Nanog, Oct4, and Sox2 also inactivate the transcription of genes involved in differentiation

into extra-embryonic, endodermal, mesodermal, and ectodermal lineages. It has recently been

suggested that the regulation of germ layer differentiation is controlled in part by Oct4 and Sox2,

and that these factors not only repress differentiation-inducing transcription factors, but promote

different fate decisions [28]. Neural ectodermal differentiation is repressed by Oct4 and

enhanced by Sox2, whereas mesendodermal differentiation is promoted by Oct4 and inhibited by

Sox2 (Figure 1C) [28]. This finding was supported by the initial identification of Oct4 as a key

regulator of pluripotency in mouse ES cells [24]. This study showed that the levels of Oct4

expression in mouse ES cells are critical in maintaining pluripotent self-renewal and preventing

differentiation, since a two-fold increase in expression promotes lineage commitment to

primitive endoderm and mesoderm [24]. Therefore, a balance of the expression of these

dominant lineage specifiers must be maintained to sustain the undifferentiated self-renewal of ES

cells and to prevent differentiation and lineage commitment.

Nanog has been identified as the master regulator of the entry into the ground state of

pluripotency during embryonic development and late during the process of nuclear

reprogramming to the pluripotent state [29,30]. It is thought that Nanog controls the transition to

pluripotency by promoting the binding of Oct4 and Sox2 to target promoters of pluripotency-

associated genes [29]. Although Nanog deletion results in early embryonic lethality, Nanog-null

ES cells are viable and contribute to all three of the germ layers in chimeric mice, but cannot

Page 20: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

5

form mature germ cells [23,31]. This shows that Nanog is dispensable for established

pluripotent cells but plays a critical role in the formation of germ cells. Nanog-null blastocysts

fail to generate pluripotent ES cells and are unable to mature into epiblast [23]. Taken together,

these studies show that the function of Nanog is a key component that establishes a core

transcriptional regulatory network in mouse ES cells and is essential for the induction of full

pluripotency.

Page 21: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

6

A

B C

Figure 1. The core signaling pathways and transcription factors regulating mouse ES cell

pluripotency.

(A) LIF activates the Jak-Stat3 and PI3K-Akt pathways, which directly upregulate the

expression of Klf4 and Tbx3, respectively. Klf4 positively regulates the expression of Sox2

and Nanog, the latter of which is also regulated by Tbx3, to promote pluripotency.

Inhibition of MAPK and GSK3 signaling (MAPKi + GSK3i) maintains pluripotency in a

LIF-independent manner. (B) The core transcription factor network (yellow box) involved

in maintaining pluripotency in mouse ES cells, consisting of Nanog, Oct4, and Sox2. These

transcription factors form a cooperative regulatory loop and maintain pluripotency by

suppressing lineage commitment and positively regulating themselves. Nanog also binds to

the promoter region and activates the expression of Rex1. (C) Oct4 promotes mesoderm

and endoderm differentiation, but suppresses neural ectoderm differentiation, whereas

Sox2 has the opposite role in regulating lineage commitment.

Page 22: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

7

1.3 Nuclear Reprogramming

During development, cells that are committed to a specific lineage generally do not change fates

and produce cells of another germ layer; for example, neuroectodermal cells do not generate

endodermal cells or tissues. It was previously thought that cells constitutively inactivated genes

as they committed to a specific tissue lineage [32]. However, several classic experiments have

shown this to be untrue. Although it is now known that neural crest cells have differentiation

potential outside of the ectodermal lineage, the ability of these cells to generate mesodermal

tissue was explored in a series of experiments that raised questions about the degree of lineage

restriction in a given tissue. Transplantation experiments of quail neural crest cells into chick

embryos and subsequent tracking of the quail cells showed that ectodermal neural crest could

give rise to mesenchymal tissues [33]. It was concluded that the environment in which a cell

resides has the ability to dictate fate determination [33]. This was one of the first examples of

“transdifferentiation”, whereby a cell belonging to one tissue type could produce cells of another

type. Classical experiments have shown that differentiated cells exhibit some degree of cellular

plasticity. Through methods such as nuclear transfer, cell fusion, and transcription factor-

mediated induction of pluripotency (Figure 2), it has been shown that mature, differentiated

somatic cells can be reverted to a primitive, pluripotent state [32].

1.3.1 Nuclear Transfer

Somatic cell nuclear transfer (SCNT) is the process in which a nucleus from a differentiated

somatic cell is transplanted into an unfertilized enucleated oocyte, typically at the stage of

second meiotic metaphase [34]. Also known as cloning, SCNT generates an organism that is

genetically identical (i.e. a “clone”) of the original somatic cell. This process shows that somatic

cells retain all of the genetic information required to produce a fully developed organism, and

that pluripotent cells can be generated from differentiated cells. These studies also suggested

that the oocyte contains non-DNA molecules, RNA, or protein that are capable of directing

reprogramming of somatic cell DNA.

The first SCNT experiments were carried out by Briggs and King in the 1950s and

showed that tadpoles could be generated from frog blastula nuclei that were transplanted into

enucleated oocytes [35]. However, developmental arrest and death occurred using nuclei from a

Page 23: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

8

different donor species or from more specialized tissues [35]. Gurdon carried out similar SCNT

experiments involving nuclei from highly specialized intestinal epithelial cells and a different

species of frog, which generated viable tadpoles, albeit with relatively low efficiency [36,37].

The efficiency of generating viable adult frogs was roughly ten times greater if the donor nucleus

was derived from a more primitive cell (such as a blastula, which is formed after zygotic

cleavage during early embryonic development) than from highly specialized, differentiated cells

(such as intestinal epithelium) [36]. This provoked the debate that only a rare intestinal stem

cell, which may be more amenable to nuclear reprogramming, produced viable clones. The

argument that a rare, contaminating somatic stem cell being reprogrammed was put to rest

decades later when highly specialized mature lymphocytes were reprogrammed by nuclear

transfer [38]. Mice derived from tetraploid complementation with donor B-cell or T-cell nuclei

had rearranged immunoglobulin alleles or T-cell receptor genes in all tissues, thereby confirming

that terminally differentiated cells can be reprogrammed by nuclear transfer [38]. Therefore,

through the process of SCNT, endogenous reprogramming factors in oocytes reactivate a full

developmental program, overriding restricted lineage commitment to direct the development of a

viable organism from highly specialized nuclei.

Although the precise molecular mechanisms involved in nuclear reprogramming remain

unclear, it is generally thought that maternal factors present in unfertilized enucleated oocytes

initiate the derepression of previously silenced genes in somatic cells, primarily by chromatin

decompaction and demethylation of gene promoters [34]. In fact, one of the earliest events in

nuclear reprogramming is the demethylation of the promoter region and expression of Oct4 [34].

Since early mouse development relies on precise levels of Oct4 expression, incomplete

demethylation of the promoter of Oct4 and low expression levels may account for the relatively

low efficiency (approximately 2%) of generating an adult mouse from a differentiated cell by

SCNT [24,34]. Other developmental abnormalities commonly seen in cloned animals, such as

elongated telomeres, obesity, weakened immune systems, and cancer predisposition suggest that

nuclear reprogramming may not be a completely efficient process [32].

Page 24: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

9

1.3.2 Cell Fusion

The impact of one genome on another can be studied through cell fusion, a technique that may

aid in uncovering the precise molecular mechanisms required for nuclear reprogramming. The

process of cell fusion can result in proliferating hybrid cells containing a single fused nucleus, or

non-dividing heterokaryons with multiple nuclei [32]. Early cell fusion experiments revealed

that the neoplastic phenotype could be suppressed by fusing a malignant cell with a non-

malignant cell, suggesting the presence of trans-acting tumour suppressor proteins [39]. These

results suggested that the non-neoplastic state was dominant over the malignant state. In normal

tissues, the linage plasticity of differentiated cells was demonstrated when stable heterokaryons

generated by the fusion of human amniotic cells and mouse myotubes resulted in the activation

of expression of previously silent muscle-specific genes in the human non-muscle cells [40].

Subsequent experiments showed that the relative ratio of the nuclei present in the heterokaryon

determined the differentiated fate of the heterokaryon, and previously silent genes were activated

in cells from all three of the germ layers [41]. DNA synthesis was not required for

transdifferentiation in heterokaryons, but DNA demethylation was found to be essential, which

was one of the first instances that epigenetics were implicated in nuclear reprogramming [42].

These studies suggested that some degree of plasticity exists in the differentiated state of

mammalian cells, which is not a permanent state.

Pluripotency was first induced in hybrid mouse cells by fusing female embryonic germ

(EG) cells with thymocytes; the resulting hybrids showed demethylation of imprinted and non-

imprinted genes and contributed to all three of the germ layers in chimeric embryos [43]. In

contrast, pluripotent hybrids formed from the fusion of thymocytes with ES cells had remaining

allele-specific methylation, suggesting that ES cells were unable to fully erase parental imprints

[44]. Demethylation of promoters and Oct4 expression have been shown to be essential for

nuclear reprogramming to the pluripotent state; in fact, in the fusion of human B cells or

fibroblasts with mouse ES cells, it has been shown that the promoter regions of Nanog and Oct4

are demethylated and gene expression is activated in the mixed-species heterokaryons within 24

hours of fusion [45,46]. Taken together, these experiments have shown that factors present in

the pluripotent state can supersede the differentiated nuclear state during cell fusion, and have

Page 25: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

10

helped to suggest that transcription factors or other molecules can allow for the induction of

pluripotency in differentiated cells.

1.3.3 Transcription Factor-Mediated Reprogramming

Ectopic overexpression of transcription factors, or as few as a single master regulatory

transcription factor, can convert the fate of a cell to that of another cell type. This was first

shown in the late 1980s, when an extra set of legs was formed instead of antennae in Drosophila

melanogaster as a result of ectopic overexpression of Antennapedia, a homeotic gene [47].

Similarly, the ectopic expression of a single transcription factor, eyeless (known as Pax6 in the

mouse), generated functional eyes on the legs, wings, and antennae of D. melanogaster,

identifying this transcription factor as a master regulator of eye morphogenesis [48]. MyoD was

identified as a key regulator of tissue specification in mice, since overexpression of MyoD in a

fibroblast cell line induced myotube formation [49]. These studies suggest that the ectopic

overexpression of a single transcription factor can induce the activation of previously silenced

signaling pathways, resulting in transdetermination or transdifferentiation from one cell fate to

another closely related cell type derived from the same germ layer.

Recently, using strategies similar to induced pluripotent stem cells (see below), mouse

and human fibroblasts have been converted to functional neurons, bypassing the pluripotent

state, as a result of the introduction of the transcription factors Ascl1 (also known as Mash1),

Brn2, and Myt1l [50,51]. However, fibroblasts are derived from primitive mesenchyme, which

has primarily a mesodermal origin, but may also originate from neural crest cells, which may be

more amenable to reprogramming to a neural fate [33]. In addition, multipotent skin-derived

precursor (SKP) cells have been identified, which differentiate in culture to generate cells

expressing neuron-specific proteins, such as βIII tubulin [52]. This suggests that rare specialized

subtypes of cells capable of transdifferentiation may be responsible for the reprogramming seen

in these studies.

The most notable example of transcription factor-mediated reprogramming, however,

was the discovery of induced pluripotent (iPS) cells [53]. The finding that mature, differentiated

somatic cells can be converted to a pluripotent ES cell-like state using a defined set of

transcription factors has provided a novel method of studying the epigenetic state and plasticity

of differentiated cells and will be discussed in detail in the following sections.

Page 26: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

11

1.3.4 Induced Pluripotent Stem Cell Technology

The technology of generating induced pluripotent stem (iPS) cells has provided a technically

more feasible method of reprogramming somatic cells using a defined set of transcription factors

[32]. Pluripotency can be induced in fully differentiated somatic cells by retroviral transduction

of vectors encoding combinations of the transcription factors Oct4, Klf4, Sox2, and c-Myc.

Pluripotency was first achieved in mouse embryonic and tail-tip fibroblasts by these methods,

although selection of reprogrammed cell lines by Fbx15 expression, a transcription factor found

specifically in mouse ES cells and early embryos, generated iPS cell lines that were unable to

form chimeras [53]. Similar reprogramming experiments involving selection for Nanog or Oct4

expression yielded germline-competent iPS cell lines from mouse embryonic fibroblasts [54,55].

Reprogramming with selection for Nanog was achieved with a maximum efficiency of

approximately 0.05% and selection for Oct4 yielded iPS colonies with a maximum estimated

efficiency of 0.08%, roughly one tenth the efficiency of iPS cell generation through Fbx15

selection [53,55]. Nanog and Fbx15 are both binding targets of Oct4 and Sox2, but Fbx15 is not

required for the maintenance of ES cell pluripotency and is dispensable for mouse development

[56]. Pluripotency was induced in adult human fibroblasts and other cell types from humans and

mice using the same four transcription factors [57,58]. It has been shown that secondary iPS

cells (generated from mouse fibroblasts harbouring doxycycline-inducible vectors and obtained

from chimeric mice produced from primary iPS cells) can be generated with an efficiency

roughly 50 times greater than primary iPS cells [59]. Regardless of the method or type of cell

employed for reprogramming experiments, the efficiency at which a cell can be converted to a

pluripotent state remains relatively low, suggesting that transcription factor-mediated nuclear

reprogramming is context dependent and may involve a series of stochastic events [60,61].

Induced pluripotent stem cells are similar to ES cells in many ways, in that they share

similar gene expression profiles, including comparable gene expression levels of key

pluripotency factors such as Nanog, Oct4, and Rex1, and similar hypomethylation patterns at the

promoters of Nanog and Oct4 [54]. Both types of cells generate cells of the endodermal,

mesodermal, and ectodermal lineages in vitro and form teratomas containing cells derived from

all three of the germ layers in vivo. However, it has recently been shown that iPS and ES cells

have many differences in DNA methylation patterns, and that reprogramming is associated with

differentially methylated regions (DMRs), some of which reflect the epigenetic memory of the

Page 27: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

12

cell of origin [62]. Epigenetic memory refers to the similarities observed in the gene expression

patterns of reprogrammed cells and cells of the somatic tissue of origin, which does not result

from changes in DNA sequence, but from epigenetic modifications such as DNA methylation.

Epigenetic memory affects the differentiation potential of iPS cells, since iPS cells harbouring

residual epigenetic marks reflecting the somatic cell type of origin differentiate more readily

along lineages related to the tissue of origin, but have restricted differentiation potential to other

lineages [63,64]. Furthermore, some of the DMRs observed in iPS cells represent iPS cell-

specific methylation patters and are produced as a result of reprogramming, suggesting that iPS

cells harbor an epigenome that is not equivalent to that of ES cells.

Page 28: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

13

A Somatic Cell Nuclear Transfer

B Cell Fusion

C Transcription Factor-Mediated Reprogramming

Figure 2. Nuclear reprogramming to pluripotency by nuclear transfer, cell fusion, and

transcription factor transduction.

(A) In somatic cell nuclear transfer (SCNT), a nucleus from a somatic cell is transplanted

into an unfertilized enucleated oocyte. This results in the nuclear reprogramming of the

somatic cell nucleus to generate a blastocyst containing pluripotent cells. ES cells can be

Page 29: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

14

derived from the inner cell mass of the blastocyst or a cloned organism can be produced if

development proceeds to completion. (B) During cell fusion, two types of cells are fused to

form proliferative hybrids or non-proliferative multinucleated heterokaryons. The ratio of

the types of nuclei present prior to fusion determines the phenotype of the heterokaryon.

(C) In transcription factor-mediated reprogramming, pluripotency can be induced in

differentiated somatic cells by the ectopic expression of a combination of the transcription

factors Oct4, Klf4, Sox2, and c-Myc. The resulting iPS cells are phenotypically similar to

ES cells.

Page 30: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

15

1.3.5 Improving iPS Cell Technology

Several alterations to Yamanaka’s initial retroviral-mediated reprogramming procedure have

been reported with the overall goal of improving reprogramming efficiency, reducing the number

of exogenous transcription factors required for the induction of pluripotency, or reducing the

possibility of oncogenic transformation.

Altering the cellular environment or disrupting key signaling pathways has been shown to

increase reprogramming efficiency. Ascorbic acid or hypoxic exposure (5% O2) enhanced the

generation of mouse and human iPS cells by promoting the transition of the somatic gene

expression profile to that of ES cell [65,66]. Ascorbic acid, or vitamin C, prevented cell

senescence and promoted the transition of pre-iPS cells to the fully reprogrammed state, whereas

hypoxia enhanced proliferation and increased the levels of expression of Nanog and Oct4

[65,66]. The role of epigenetics in reprogramming to the pluripotent state was further

emphasized by the finding that DNA methyltransferase and histone deacetylase (HDAC)

inhibitors enhance the generation of mouse and human iPS cells [67,68]. The DNA

methyltransferase inhibitor, 5-azacytidine (AZA) enhanced mouse iPS cell generation by

promoting the transition to the fully reprogrammed state [67]. Similarly, the reprogramming

efficiency of generating mouse iPS cells was increased by approximately 100-fold using the

HDAC inhibitor valproic acid (VPA), and human fibroblasts were successfully reprogrammed

with only Oct4 and Sox2 in the presence of VPA [67,68]. Cellular senescence and the DNA

damage response mediated by the p53-p21 pathway and other cell cycle regulatory pathways

controlled by the Ink4/Arf locus have been shown to prevent reprogramming to the pluripotent

state [69,70,71,72,73]. The aforementioned studies have aided in improving the efficiency of

generating iPS cells and have helped elucidate the molecular events and signaling pathways

implicated in the reprogramming of somatic cells to a pluripotent state.

The retroviruses commonly used in reprogramming experiments can cause insertional

mutagenesis, which may complicate the study of cancer with iPS cells and prevent their use in

clinical applications. Methods of generating iPS cells without viruses were therefore developed,

such as piggyBac transposition [74], recombinant protein delivery [75], transfection of plasmid

vectors or non-integrating episomal vectors [76,77], synthetic mRNA delivery [78], and ectopic

miRNA expression [79]. However, it has been shown that human iPS cells generated from five

Page 31: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

16

different methods acquired not only epigenetic modifications, but also genetic alterations during

the reprogramming process; these reprogramming-associated mutations included genes

implicated in cancer [80]. Furthermore, the reprogramming process results in de novo copy

number variations and deletions of genomic regions containing tumour suppressor genes in

human iPS cells [81,82].

The exclusion of the potent oncogene c-Myc from the reprogramming process was

investigated due to the fact that this potent oncogene was reactivated in iPS cells injected in vivo

to produce tumours in chimeric mice and their offspring [54]. This transcription factor was

found to be dispensable for the induction of pluripotency in mouse and human cells, although

reprogramming efficiency was substantially reduced if c-Myc was excluded during the

reprogramming procedure [83]. An alternative to the omission of c-Myc is its replacement with

L-Myc, which has been shown to promote iPS cell generation and germline transmission with

less potent oncogenic effects [84]. Recently, it has been shown that the efficiency of

reprogramming is further enhanced by the addition of Glis1, a transcription factor found in

oocytes but not ES cells [85]. This finding represents the first transcription factor involved in

nuclear reprogramming of somatic cells to a pluripotent state, but not expressed in pluripotent ES

cells.

1.3.6 Induced Pluripotent Stem Cells Generated from Neural Stem Cells

Adult mouse and human NSCs have been successfully reprogrammed with two factors (Oct4 and

Klf4) [86,87] or one factor alone (Oct4) [88,89] to generate iPS cells, albeit with remarkably

lower efficiencies than with all four Yamanaka factors (Oct4, Klf4, c-Myc, and Sox2) [53].

Although Klf4 is an oncogene and could potentially be reactivated in vivo to produce tumours, its

inclusion as a reprogramming factor enhances the efficiency of generating iPS cells from mouse

NSCs by approximately 10-fold compared to one-factor reprogramming with Oct4 alone [89].

The generation of iPS cells from mouse NSCs by the ectopic expression of Oct4 and Klf4 is

generally associated with a reprogramming efficiency of approximately 0.11% [86], which is

similar to the efficiency of producing four-factor iPS cells from MEFs [54]. A small-molecule

histone methyltransferase inhibitor, BIX-01294, was found to increase the efficiency of

reprogramming mouse NSCs with ectopic Oct4 and Klf4 to the efficiency achieved for four-

Page 32: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

17

factor reprogramming, again implicating epigenetic modifications in the process of nuclear

reprogramming [90].

The expression of the stem cell maintenance gene Sox2 by NSCs is partly responsible for

the enhanced efficiency of generating iPS cells from NSCs. In fact, human and mouse NSCs

endogenously express Sox2 and c-Myc at greater levels than ES cells, and also express markers

associated with the iPS cell phenotype, such as alkaline phosphatase (AP) and

CD15/LewisX/SSEA-1 [89]. It is also interesting that a pluripotency marker used to identify iPS

cells, such as SSEA-1 is also a marker of adult neural progenitor cells [13]. It has been shown

that AP and SSEA-1 are activated sequentially and relatively early during the reprogramming of

mouse somatic cells, and are followed by the expression of Nanog and Oct4 [30]. The

expression of stem cell maintenance genes, particularly Sox2, makes NSCs good candidates for

reprogramming to a pluripotent state using few exogenous factors.

1.4 Reprogramming and Cancer

Cancer could be considered to be a disease of pathological “reprogramming”, since normal cells

can be transformed into cancer cells as a result of combinations of genetic and epigenetic

aberrations. Initially proposed in a 1971 study on retinoblastoma, Knudson’s “Two Hit”

Hypothesis of cancer suggested that at least two mutational events are required for the initiation

of cancer [91]. In other words, for a genetically unstable cell to be transformed into a cancer

cell, the process requires a second perturbing event, or “hit” [92]. Genetic predisposition and

environmental factors increase an individual’s risk of developing cancer [92]. Therefore, cancer

results from the accumulation of genetic and epigenetic (non-genetic) changes over time. We

now understand that both inherited and sporadic cancers can be caused by the inactivation of

tumour suppressor genes and/or the activation of oncogenes as a result of two independent hits,

which involves a combination of genetic and epigenetic aberrations [93]. Studies assessing the

role of epigenetics in cancer, and the plasticity and potential pluripotency of cancer cells, have

provided valuable insights into the non-genetic factors influencing cancer.

1.4.1 Introduction to Epigenetics

Epigenetics is the term describing the inheritable changes in gene expression levels by

mechanisms other than changes in DNA sequence, which are mediated by processes such as

Page 33: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

18

DNA methylation, histone modification, and chromatin remodeling [94]. Stable gene expression

alterations are caused by epigenetic processes, which are retained through mitosis; for instance,

DNA methylation patterns are transmitted during cell division [95]. Interplay exists between

epigenetic processes, since DNA methylation recruits histone-modifying enzymes and vice-

versa.

The most common targets for methylation are cytosine-guanine (CpG) sites, which

cluster in 0.5-2 kb regions of the genome termed “CpG islands” [96]. Predominantly situated in

promoter and coding regions of genes, CpG islands remain, for the most part, unmethylated in

autosomal genes. Methylation of CpG islands in promoter regions alters protein-DNA

interaction, since methylcytosine protrudes into the major groove of DNA, and reduces the

binding affinity of some transcription factors, thereby inhibiting transcription initiation [95].

Furthermore, methyl CpG binding protein 2 (MeCP2) and similar proteins bind to methylated

regions of DNA and recruit HDAC enzymes [97,98]. Nucleosomes consist of DNA wrapped

around an octamer of histone proteins, which are subject to reversible post-translational

modifications, including acetylation and methylation, among others. Generally, genes silenced

by DNA methylation are associated with histones that are hypermethylated and hypoacetylated at

specific lysine residues [99]. Nucleosome remodeling complexes bind to modified histone

proteins and reposition nucleosomes, thus exposing or concealing genes to be transcribed or

repressed.

The importance of DNA methylation during mammalian development was initially

exemplified by gene targeting in mouse ES cells to mutate a putative DNA methyltransferase

(DNMT) gene, resulting in a recessive lethal phenotype in embryos [100]. Dnmt1 homozygous

mutant ES cells and embryos showed approximately 3-fold reduction in genomic methylcytosine

levels compared to controls; the ES cells were viable in culture, whereas the embryos died mid-

gestation [100]. The subsequent identification and inactivation of Dnmt3a and Dnmt3b genes,

which are highly expressed in undifferentiated ES cells but downregulated in differentiated cells

of adult mammals, revealed that these DNMTs are essential for de novo methylation and

mammalian development [101,102]. Inactivation of both Dnmt3a and Dnmt3b in mice resulted

in embryonic lethality before e11.5, due to growth arrest and impaired development shortly after

gastrulation [102]. Dnmt1 is involved in the maintenance of DNA methylation marks during cell

division, whereas Dnmt3a and Dnmt3b are responsible for de novo methylation during

Page 34: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

19

development [102]. Somatic cells harbour predominantly methylated CpG sites, with the

promoter or coding regions of genes containing regions with a high proportion of unmethylated

CpG islands [96]. Inactivation of the X chromosome, genomic imprinting, and silencing of

autosomal Alu insertional elements (a type of short interspersed element) and L1

retrotransposons are processes that rely on methylation of CpG islands in the promoter regions of

genes [96]. Gene expression is silenced by methylation of CpG islands and methylation patterns

are heritable, which provides a method of altering levels of gene expression without affecting the

underlying DNA sequence.

1.4.2 Epigenetic Contribution to Cancer

Cancer is a disease of aberrant genetics and epigenetics, but the relative contribution of each to

tumour progression remains largely unclear. DNA methylation is the most thoroughly

characterized epigenetic process involved in cancer [96]. In broad terms, cancer is associated

with genomic hypomethylation and CpG island hypermethylation. The first epigenetic

aberration to be associated with cancer was, in fact, global hypomethylation, which may promote

malignant transformation by causing chromosomal instability, promoting the loss of imprinting,

or reactivating transposable elements [93,103]. The aberrant methylation in cancer may involve

decreases in the overall amount of methylcytosine, demethylation of specific loci, de novo

methylation of CpG islands, or overexpression of DNMT enzymes [96]. It was first shown that

sites of cytosine methylation often correlated with mutated regions of DNA; the spontaneous

deamination of 5-methylcytosine resulted in transition mutations of cytosine to thymine and

subsequent failure to correctly repair the mismatch [104]. This study established a link between

methylated DNA regions and genomic mutations.

Similar to genes in normal cells, tumour suppressor genes in transformed human cells are

susceptible to methylation, including TP53, RB1, VHL, CDKN2A, CDKN2B, MLH1, and APC

[95]. The promoter regions of these genes harbour CpG islands, which are targeted for

methylation and subsequent gene silencing. For instance, approximately one quarter of the point

mutations in the tumour suppressor p53 gene are associated with CpG islands; in colon cancer,

this fraction is increased to nearly one half [96]. In addition to promoter hypermethylation, point

mutation and loss of chromosomal material through homozygous deletion or loss of

heterozygosity (LOH) may contribute to gene inactivation in cancer [95]. Aberrant methylation

Page 35: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

20

in cancer may be mediated by increased expression or deregulated activity of de novo

methyltransferases, although the precise molecular mechanisms by which global methylation

levels are altered in cancer remain unclear [94].

It was long disputed whether de novo methylation in cancer was causative and directly

involved in initiating oncogenic transformation, or secondary and merely accompanying tumour

incidence [95]. The causal role of DNA methylation in cancer was investigated by gene

targeting of Dnmt1, which is a classical DNMT involved in the maintenance of methylation; not

only was genomic imprinting impaired, but the incidence of intestinal cancer was significantly

reduced [105,106]. Interestingly, cancer cells show a loss of genomic imprinting that arises as a

result of aberrant DNA methylation [107]. Furthermore, Dnmt3a and Dnmt3b, which are

involved in de novo methylation, are frequently overexpressed in cancer; depletion of Dnmt3b

reactivated genes that were silenced by hypermethylation and induced apoptosis in human cancer

cells in vitro [102,108]. Finally, promoter hypermethylation of important oncogenes in their

wildtype form, such as VHL and RB1, directly suppresses gene expression and has been

confirmed in familial cases of VHL syndrome and retinoblastoma, respectively [95]. Taken

together, these studies suggest that DNMTs involved in the maintenance and initiation of new

methylation patterns play an important causative role in cancer.

In addition, methylation of CpG islands within the promoter region of genes encoding

DNA repair enzymes silences the expression of these genes and promotes mutagenesis [96]. A

notable example is O6-methylguanine DNA methyltransferase (MGMT), which removes

mutagenic alkyl groups from O6-methylguanine [109]. This alkylated nucleotide binds

preferentially to thymine, causing transition mutations, or cross-links with cytosine residues,

impeding DNA replication [109]. Aberrant methylation of the MGMT promoter occurs in 40%

of glioma brain tumours and colorectal carcinomas, and in 25% of non-small cell lung cancers,

lymphomas, and head and neck carcinomas, but rarely in other types of tumours [109]. Silenced

MGMT expression as a result of promoter hypermethylation is an important factor in the

pathogenesis of these types of cancer by promoting DNA mutagenesis.

Aberrant methylation patterns also play a role in deregulating the activity of histone-

modifying enzymes. Histone methyltransferases, such as mixed-lineage leukemia 1 (MLL1)

create transforming fusion proteins involved in leukemogenesis, and the polycomb group (PcG)

Page 36: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

21

complex protein Bmi1 silences the tumour suppressor genes p16INK4a

and p14ARF

in lymphoma,

brain, and other types of cancer [110,111,112]. Bmi1 is also essential for the regulation of

proliferation of normal and leukemic stem cells, resulting in cell cycle arrest, differentiation, and

apoptosis in Bmi1-deficient stem cells [113]. PcG proteins have a role in maintaining the long-

term silencing of differentiation-inducing genes in stem cells through the alteration of histone

modifications, particularly through trimethylation of lysine 27 of histone 3 [114]. PcG proteins

also have target genes that are often hypermethylated in their promoter regions and silenced in

cancer [115]. This was thought to create an “epigenetic stem cell signature” in cancer, which

could promote the abnormal expansion of cells in the early stages of malignant transformation

[115]. Evidently, a great deal of interplay exists between aberrant methylation patterns and

altered activity of histone-modifying enzymes in cancer.

1.4.3 Chemotherapy Targeting Epigenetic Modifications in Cancer

The epigenetic modifications involved in cancer pathogenesis and progression are theoretically

fully reversible; therefore, processes such as the aberrant methylation in cancer are ideal targets

for chemotherapeutic agents. A caveat could be if a genetic mutation exists in an epigenetic

regulator, such as a DNMT or histone methyltransferase. By restoring normal methylation levels

to CpG islands within the promoter regions of tumour suppressor genes, expression of these

genes could be reactivated to reestablish normal growth control [96]. In fact, AZA was the first

drug approved by the FDA as a methyltransferase inhibitor and is the common treatment for

myelodysplastic syndromes, which are hematopoietic stem cell disorders associated with

hypermethylation of the cell cycle regulator, p15INK4B

[116]. Methylated DNA recruits HDACs

through the assistance of MeCP2 and other methyl CpG binding proteins, which further

emphasizes the interplay between aberrant methylation and histone-modifying enzymes in cancer

[97,98]. AZA and HDAC inhibitors are currently being evaluated in clinical trials for other

types of tumours [117,118].

Page 37: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

22

1.4.4 Pluripotency and Cancer

Cancer cells are characterized by many features, including uncontrolled self-renewal and failure

to senesce, which are similar characteristics of pluripotent ES cells, since these cells self-renew

indefinitely in vitro. The aforementioned epigenetic stem cell signature in cancer involves the

preferential silencing of differentiation-inducing PcG target genes by promoter

hypermethylation, thereby promoting the self-renewal of neoplastic cells [115]. In fact, cancer-

specific DNA hypermethylation was 12-fold more likely to be associated with PcG target genes

than with non-PcG targets [115]. These findings are corroborated by a study that implicated a

pluripotent ES cell-like identity, including silencing of PcG-regulated genes, in poorly

differentiated human tumours [119]. Aggressive tumours often consist of poorly differentiated

cells, and cancers such as high-grade estrogen receptor (ER)-negative breast cancer, high-grade

glioma, and bladder carcinomas showed preferential overexpression of activation targets of

Nanog, Oct4, Sox2, and c-Myc [119]. The ES cell-like gene expression profile in ER-negative

breast cancer was a negative prognostic indicator, predicting significantly higher mortality rates

[119]. The ES cell-like expression signature in aggressive tumours did not involve the

expression of Nanog, Oct4, or Sox2, but simply the overexpression of gene sets representing the

core expression signature of ES cells.

However, it is likely that a gene expression network governed by c-Myc accounts for a

significant portion of the similarities between the transcriptional profiles of ES and cancer cells

[120]. The interpretation that cancer represents a regression to an ES cell-like state due to

dedifferentiation of cells is suspect, since the pluripotency signature overlaps significantly with

the c-Myc regulatory network. This network is involved in regulating cellular metabolism, cell

cycle, and protein synthesis pathways during oncogenic transformation [120]. Myc is also

implicated in somatic cell reprogramming to the pluripotent state. A similarity between

pluripotent and cancer cells can be found in the p53-p21 tumour suppressor pathway, which

prevents the transition to the pluripotent state during transcription factor-mediated nuclear

reprogramming in a manner that parallels its role in suppressing oncogenic transformation

[69,70,71,72]. Overlap between the c-Myc network and the core pluripotency network regulated

by Nanog, Oct4, and Sox2 makes it difficult to conclude whether a true ES cell-like signature

exists in cancer cells.

Page 38: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

23

Nevertheless, similar to normal cells, precancerous and cancerous cells display some

degree of cellular plasticity. It was recently shown that phenotypic plasticity occurs in healthy

human epithelial breast tissue, with a rare subpopulation of epithelial cells expressing Nanog and

Oct4 and harbouring the capacity to generate teratomas containing derivatives of the three germ

layers in vivo [121]. These cells, termed “endogenous provisionally pluripotent stem cells”, are

distinct from human ES cells and mesenchymal stem cells, expressing markers of endoderm,

mesoderm, and ectoderm upon differentiation, and may act as breast cancer precursors [121].

Furthermore, phenotypic plasticity may occur in cancer, as some neoplastic cells have an

intrinsic capacity for endothelial differentiation, which may play a role in the adaptation to a

hypoxic tumour microenvironment through the process of vasculogenic mimicry [122]. This

process has been associated with aggressive forms of skin, brain, breast, prostate, ovarian, lung,

and rhabdomyosarcoma cancers, among others [122,123,124]. Vasculogenic mimicry is similar

to embryonic vasculogenesis, with cancer cells activating the expression of previously silenced

signaling molecules and endothelium-associated genes to form extracellular matrix-rich fluid-

conducting vessels [122]. This example of phenotypic plasticity may reflect the unstable

epigenetics of cancer cells and should not be interpreted as true pluripotency.

Historically, a link was initially made between pluripotency and cancer by seminal

studies in the mid-1960s showing that single embryonal carcinoma (EC) cells are pluripotent

stem cells of a teratocarcinoma and can produce tissues derived from all three of the germ layers

when injected in vivo [125]. This study also demonstrated the heterogeneity of EC cells derived

from a teratocarcinoma. Upon blastocyst injection, mouse teratocarcinoma cells gave rise to the

generation of viable chimeric mice devoid of tumours and contributed to germline transmission

[126,127]. By directing postimplantation development, the teratocarcinoma cells activated the

expression of many somatic and germline genes that had been silenced in the original tumour

[126]. These studies suggested that embryo-derived teratocarcinomas, which are tumours of

embryonic or germ cell origin, have not only pluripotent potential, but can direct the

development of a viable organism. It was later shown that EC cells express Oct4, which may

confer a pluripotent phenotype in these cells [128]. Oct4 is also expressed in embryonic germ

(EG) cells, primordial germ cells (PGCs), and identifies pluripotent cells in germ cell tumours,

which is suggestive of a PGC cell-of-origin theory for germ cell tumours [129,130]. In germ cell

tumours, Oct4 may be a dose-dependent oncogenic determinant; overexpression of Oct4

Page 39: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

24

increases tumour malignancy, whereas suppression of Oct4 prevents tumour growth [129].

Although Oct4 and Nanog expression have been detected in some somatic tumours, including

aggressive breast and brain cancers, the functional role of these transcription factors in the

context of cancer remains unclear [131,132,133,134]. It is highly unlikely that Oct4 and Nanog

have the same pluripotency-associated function in cancer cells as they do in normal cells [134].

1.4.5 Nuclear Reprogramming of Cancer Cells

Taking into account the extensive involvement of epigenetic processes in cancer, a question that

arises is whether resetting the epigenetic program of cancer cells through the process of nuclear

reprogramming could attenuate the neoplastic phenotype. A classical experiment in the late

1960s showed that renal carcinoma nuclei from frogs could be transplanted into enucleated

oocytes to give rise to viable tadpoles [135]. Initially attributed to the pluripotent nature of the

renal carcinoma cells, the wide variety of cell types employed in the classical nuclear

reprogramming studies previously discussed suggests that pluripotency is induced during

reprogramming and is not necessarily a characteristic of the donor cell.

Nuclear transfer has been employed to reprogram several mouse models of cancer. Cells

from the classical Patched1 heterozygous (Ptc1+/-

) mouse model of medulloblastoma were

reprogrammed by nuclear transfer, a process that attenuated tumourigenicity and suppressed

proliferation [136]. The epigenetically reprogrammed Ptc1+/-

medulloblastoma nuclei were able

to direct preimplantation development and formed postimplantation embryos devoid of

malignancies, suggesting that tumourigenicity was suppressed, at least at an early developmental

stage. Similarly, nuclei from experimentally induced leukemia, lymphoma, and breast cancer

cells were reprogrammed by nuclear transplantation and formed blastocysts, but failed to

generate viable ES cells [137]. By contrast, Ras-inducible melanoma nuclei were reprogrammed

by nuclear transplantation to generate viable chimeras, although a variety of tumours arose

rapidly in the chimeric mice, due to the reactivation of Ras in vivo [137]. Furthermore, in

tetraploid complementation experiments (when implanted ES cells produce the embryo and

tetraploid host cells generate extra-embryonic tissues), the ES cells derived from Ras-inducible

melanoma nuclei could only support development until e9.5 [137]. Similarly, mouse EC cell

lines were reprogrammed by nuclear transfer, and the resulting ES cells had the same

tumourigenic and developmental potential as the donor EC cells [138]. The failure to alter the

Page 40: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

25

tumour-initiating ability of EC cells by SCNT was attributed to permanent genetic lesions, rather

than epigenetic aberrations [138]. Taken together, these finding suggests that the autonomous

developmental potential of cancer cells may be limited by permanent genetic lesions and that

reprogramming by SCNT is insufficient to block tumour-initiating ability.

Recently, gastrointestinal (GI) cancer cells and chronic myeloid leukemia (CML) cells

have been reprogrammed using Yamanaka factors to generate iPS cells [139,140]. It is

interesting to note that the GI cancer-derived iPS cells were associated with an increased

sensitivity to differentiation-inducing treatment and to the chemotherapeutic agent 5-

fluorodeoxyuridine, whereas the CML-derived iPS cells lost the dependency on continuous

signaling of the BCR-ABL oncogene fusion protein and became resistant to the BCR-ABL

inhibitor imatinib [139,140]. Furthermore, re-differentiation of CML-derived iPS cells to a

hematopoietic cell fate restored BCR-ABL dependency; cells expressing hematopoietic markers

(CD34, CD43, and CD45) were killed as a result of imatinib treatment, whereas the viability of

non-hematopoietic cells was not affected [140]. These studies implicated the epigenetic state of

cancer cells in their sensitivity to differentiation-inducing chemicals and chemotherapeutic

agents.

1.5 Cancer Stem Cells

Cancer stem cells (CSCs) comprise the subset of cells in a cancer with the ability to initiate

tumour growth. CSCs share several properties with normal stem cells, such as self-renewal and

differentiation, but are the cells responsible for cancer progression and recurrence. CSCs were

initially identified in human acute myeloid leukemia based on the isolation of cells with the

hematopoietic stem cell surface phenotype (CD34+CD38

-) that could engraft and serially

transplant the disease in sub-lethally irradiated non-obese diabetic/severe combined immune-

deficient (NOD/SCID) mice [141]. This study established a standard in vivo model of serially

transplanting cells into NOD/SCID recipients to verify the cancer-initiating capacity of putative

CSCs, which must also produce a xenograft that recapitulates or phenocopies the original cancer.

Through similar methods, CSCs were first identified in solid tumours by isolating cells

from primary human breast cancers based on the cell surface marker expression profile

CD44+CD24

-/Lineage

-; these CSCs engrafted and initiated tumour growth that phenotypically

mimicked the primary human tumour when as few as 1,000 cells were injected into the

Page 41: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

26

mammary fat pad of NOD/SCID mice [142]. CSCs were later identified in solid primary

tumours including cancer of the brain [143], bone [144], prostate [145], skin [146], colon [147],

lung [148], head and neck [149], pancreas [150], and mesenchymal tissue [151]. Early

controversial studies involving the transplantation of cancer cells subcutaneously in human

subjects were conducted in the mid-20th

century were the first to demonstrate the functional

heterogeneity of human tumours [152,153,154]. One study showed that the incidence of tumour

formation was only 50% when one million cancer cells were implanted in human subjects [154].

Initially attributed to the “host resistance to cancer”, these results can be described by the Cancer

Stem Cell Hypothesis, which relates to the functional heterogeneity occurring within a tumour.

1.5.1 The Cancer Stem Cell Hypothesis

The hierarchical model of tumour progression involving cancer stem cells suggests that not every

cell within a tumour has an equivalent potential for self-renewal, differentiation, or tumour

initiation. The Cancer Stem Cell Hypothesis proposes that only CSCs are able to regenerate

tumours when cancer cells are injected into immune-compromised animals. The CSC therefore

lies at the apex of the hierarchy of cells within a tumour, initiates tumour growth, and can self-

renew to generate CSCs or differentiate to generate non-cancer stem cells of the tumour bulk

(Figure 3). This model is in contrast to the stochastic model of cancer, which suggests that every

cell within a tumour has the potential to proliferate and initiate tumour growth, although the

probability of tumour initiation remains low for any given cell [155]. The stochastic model of

cancer implies functional homogeneity within a tumour, whereas the Cancer Stem Cell

Hypothesis suggests functional heterogeneity of cells in a tumour. The isolation of CSCs from a

primary tumour based on markers or phenotype, therefore, enriches for clonogenic cells in vitro

and tumour-initiating cells when injected into immune-deficient recipients in vivo. Importantly,

the Cancer Stem Cell model does not predict the cell of origin of a cancer. Although CSCs share

several characteristics with normal stem cells, such as self-renewal and differentiation, this does

not imply that CSCs occur as a result of oncogenic transformation of normal stem cells.

The Cancer Stem Cell Hypothesis has important clinical implications in cancer,

particularly in treatment and tumour recurrence, since treatment regimens targeting non-CSCs,

which constitute the majority of cells within the tumour (i.e. tumour bulk), fail to cure the

disease and lead to recurrence if CSCs remain after treatment. The Cancer Stem Cell Hypothesis

Page 42: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

27

is supported by experimental results suggesting that CSCs are more resistant to chemotherapy

and radiation treatment compared to non-CSCs, due to expression of drug transporters and

increased DNA repair capacity [156,157,158]. Furthermore, recent evidence from acute

lymphoblastic leukemia suggests that genetic diversity exists in leukemia-initiating cells and that

multi-clonal evolution occurs during leukemic progression [159]. Therefore, due to the

functional heterogeneity of cells within a cancer of the blood or solid tissues, treatments

specifically targeting CSCs are essential in curing cancer and preventing recurrence.

Page 43: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

28

A

B

Figure 3. A comparison of the stochastic and cancer stem cell models of cancer.

(A) The stochastic model of cancer (left) posits that every cell within a tumour is

functionally equivalent and has similar, albeit low, tumour-initiating potential. The cancer

stem cell model of cancer (right) implies functional heterogeneity within a tumour, with the

tumour-initiating CSC located at the apex of the hierarchy. This model suggests that only

rare CSCs have the ability to initiate and maintain tumour growth, through self-renewal

and differentiation to produce non-CSCs of the tumour bulk. (B) Treatments failing to

target CSCs lead to tumour recurrence by sparing CSCs, whereas targeted treatments

eliminate the CSCs and cause tumour regression.

Page 44: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

29

1.6 Brain Tumours

Brain tumours comprise a diverse group of cancers occurring in the central nervous system

(CNS). In the developing brain of children (aged 0-14) and young adults (aged 15-29), tumours

are thought to arise as a result of spontaneous oncogenic transformation of proliferating cells,

whereas the gradual accumulation of genetic lesions has been attributed to the cause of brain

tumours in adults (>30 years) [160]. In fact, the most common types of brain tumours occurring

in younger individuals vary significantly from those occurring most commonly in the aged

population. The most common types of brain tumours occurring in children and young adults are

embryonal tumours such as primitive neuroectodermal tumours (PNETs) and medulloblastoma

(MB), low-grade astrocytomas, and ependymomas [160]. These tumours are considerably rarer

in adults, who are most frequently affected by astrocytic glioma brain tumours. The World

Health Organization (WHO) has applied a histological grading scale of I-IV to CNS tumours,

with grade I tumours consisting of benign growths with low proliferative potential and grade IV

glioblastoma multiforme (GBM) consisting of anaplastic growths displaying high proliferative

indices, angiogenesis, cellular invasion, and necrosis [161]. High-grade GBM is the most

common type of brain tumour occurring in adults and is associated with a median survival of

only 12.1 months with radiotherapy alone or 14.6 months with the addition of temozolomide

chemotherapy [162]. Therefore, while non-metastatic embryonal tumours arising in children,

such as MB, are associated with relatively high survival rates with aggressive therapy, high-

grade gliomas affecting adults are one of the most devastating types of cancer.

1.6.1 Human Glioma

Gliomas are characterized pathologically as primary parenchymal tumours that exhibit glial

differentiation and are classified as astrocytomas, oligodendrogliomas, ependymomas, or mixed

gliomas, based on the predominant cell type present [161]. Grade IV malignant gliomas can be

identified as a primary GBM upon initial diagnosis, or can develop from a low-grade

astrocytoma over the course of approximately 5 years to produce a secondary GBM. The

majority of GBMs (>90%) arise de novo in patients aged 62 years (average) and progress rapidly

over the course of only a few months, whereas secondary GBMs occur in a younger population

(aged 45 years, average) and are associated with longer survival [161]. It has been proposed that

although primary and secondary GBM share several genetic, pathological, and biological

Page 45: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

30

features, they consist of two distinct clinical entities, due to striking differences in the genetic

pathways affected. For instance, both types are characterized by LOH 10q (approximately 70%

of cases), but primary GBMs are most often associated with EGFR amplification (36%), p16INK4a

deletion (31%), and TP53 mutation (28%), whereas TP53 mutation is the most frequent genetic

mutation (65%) implicated in secondary GBMs, followed by p16INK4a

deletion (19%) and EGFR

amplification (8%) [163]. Therefore, several genes have been implicated in the pathogenesis of

high-grade glioma, but the frequency of specific genetic aberrations differs significantly between

primary and secondary GBM.

More recently, genome-wide sequencing studies have identified other important genetic

aberrations commonly occurring in stage IV glioma, including homozygous deletion of PTEN,

NF1, RB1, and IDH1, as well as mutation or amplification of CDK4, PDGFRA, and MDM2

[164,165]. Furthermore, the signaling pathways more commonly altered in GBM were identified

as PI3K (88% of cases), p53 (87%), and Rb (78%) [164]. All three of the signaling pathways

were altered in 74% of cases [164]. These genomic analyses have helped to identify common

genetic aberrations occurring in the most aggressive form of glioma, thereby presenting genes

and signaling pathways that could be targeted by novel therapeutics and in animal models of the

disease.

Although the genetic changes associated with high-grade glioma have been well

characterized [164,165], it has also been recognized that extensive epigenetic changes are

involved. Epigenetic processes commonly implicated in cancer also apply to brain tumour

pathogenesis, such as aberrant hypermethylation of CpG islands in promoter regions of tumour

suppressor genes and altered histone modification. In a broad study investigating the gene

hypermethylation profiles of human cancers, the following genes were associated with

hypermethylated promoters in brain tumours (although the types of brain tumours were not

specified): MGMT (34% of samples), p16INK4A

(30%), TIMP3 (26%), p14ARF

(9%), and GSTP1

(5%) [166]. In glioma brain tumours, promoter hypermethylation silences the expression of

genes involved in cell cycle control (p16INK4A

and p15INK4b

) tumour suppression (RB1, VHL, and

RASSF1A), DNA repair and genome integrity (MGMT and MLH1), among other functions [167].

The heterogeneous methylation profile of high-grade gliomas is exemplified by another

study that identified several other genes as frequently hypermethylated in GBM, including

Page 46: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

31

FZD9, TNFRSF10A, MEST, TES, and HOXA11 [168]. Interestingly, this study found that genes

with hypermethylated promoters in GBM are significantly enriched (41%, p < 0.001) for targets

of the Polycomb repressive complex (PRC)2 in ES cells [168]. This study, therefore, established

a link between promoter hypermethylation and PRC2 target genes in GBM. Furthermore, the

core component of PRC2, EZH2, and the PcG protein and oncogene, BMI1 (a member of PRC1),

were found to be overexpressed in GBM tumours, particularly in the CD133+ cancer stem cell

fraction, and were required for self-renewal [169]. Stable knockdown of BMI1 or EZH2 reduced

the clonogenic potential of GBM cells in vitro, while BMI1 knockdown reduced brain tumour

formation in vivo [169]. BMI1 typically targets the INK4A/ARF locus to suppress expression of

p16INK4A

and p14ARF

, but there is evidence that BMI1 may target other tumour suppressor

pathways in human and mouse glioma [112,169]. Furthermore, BMI1 may promote radiation

resistance in GBM by recruiting DNA damage response machinery, including ATM kinase and

γH2AX [170]. EZH2, the core component of PRC2, directly targets c-Myc in GBM and is

considered to be essential for BTSC self-renewal in vitro and tumour-initiating ability in vivo

[171]. Taken together, these studies show that PRC1 and PRC2, which regulate PcG target gene

expression through chromatin modification, may play a role in the maintenance of self-renewal

of cancer cells in high-grade glioma.

Aberrant promoter hypermethylation of genes encoding DNA repair enzymes has also

been associated with glioma pathogenesis. Silencing of the MGMT gene by promoter

hypermethylation occurs in approximately 40% of gliomas, but less frequently in non-glial

tumours, thereby reducing the expression levels of a key DNA repair enzyme and promoting

mutagenesis [109]. Loss of expression of MGMT results in a failure to remove alkyl groups

from guanine residues, which binds preferentially to thymine, resulting predominantly in G:C to

A:T transition mutations [109]. Aberrant promoter methylation and silencing of MGMT has

been implicated in the early stages of the transformation from low-grade gliomas to high-grade

secondary GBM, since hypermethylation occurs in 48% of low-grade diffuse astrocytomas and

in 75% of secondary GBM (compared to only 36% of primary GBM) [172]. Furthermore,

hypermethylation and silencing of MGMT is associated with increased G:C to A:T transition

mutations in the key tumour suppressor gene, TP53 [172]. For instance, 92% of secondary

GBMs with MGMT hypermethylation showed mutations in TP53, whereas only 25% of those

without MGMT hypermethylation harboured TP53 mutations [172]. Therefore,

Page 47: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

32

hypermethylation of the promoter of MGMT, which encodes a key DNA repair enzyme,

promotes the accumulation of mutations in TP53 and perhaps other tumour suppressor genes.

Nevertheless, promoter hypermethylation of MGMT predicts a more favourable outcome

for patients treated with the common chemotherapeutic alkylating agent, temozolomide, since

alkyl adducts are not removed from guanine residues, thus triggering chemotherapy-induced

apoptosis of tumour cells [173,174]. Therefore, promoter hypermethylation and silencing of the

MGMT gene promotes the preservation of mutagenic alkylated nucleotides, but may also act as a

favourable prognostic marker when combined with alkylating drugs. In fact, GBM patients with

long-term survival (>2 years) more frequently harbour MGMT promoter hypermethylation than

classic GBM survivors (78% versus 39%) [167]. In addition, epigenetics may play a role in

recurrence of high-grade gliomas; approximately two thirds of relapsed GBMs show

significantly different DNA methylation patterns compared to the primary GBMs from which

they arose, primarily due to hypermethylation of promoters of pro-apoptotic genes [175]. The

identification of distinct epigenetic profiles associated with individual subtypes of glioma and

different tumour stages, or as predictive biomarkers, may encourage the development of novel

chemotherapeutic agents targeting DNA methylation or chromatin modification.

1.6.2 Mouse Glioma

Mouse models of cancer can be produced by exposure to carcinogens, targeted deletion or

mutation of tumour suppressor genes, forced expression of oncogenes, or a combination thereof.

Mouse models of glioma targeting the tumour suppressor p53 are clinically representative of the

human disease, since p53 signaling is inactivated in 87% of high-grade gliomas, with mutations

or homozygous deletions of TP53 occurring in 35% of GBM cases [164]. It has been shown that

homozygous or heterozygous deletion of the mouse p53 gene results in predisposition to various

types of cancer and 50% survival rates of approximately 20 weeks and >70 weeks, respectively

[176]. In p53-/-

mice, thymic lymphoma is the most common type of cancer, whereas sarcomas

most often occur in p53+/-

mice [176]. On the other hand, Cre-mediated deletion of p53 in cells

expressing the astrocytic cell marker, glial fibrillary acidic protein (GFAP), has been shown to

generate high-grade brain tumours with 90% penetrance and high specificity for malignant

gliomas (>85%, based on histopathology) [177]. Therefore, targeted deletion of p53 in the CNS

of mice is one molecular mechanism for the generation of high-grade malignant brain tumours.

Page 48: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

33

In addition to targeted deletion of tumour suppressors, mouse models of glioma have

been reported that incorporate carcinogen treatment in utero. A common carcinogen for such

applications is the alkylating agent, N-ethyl-N-nitrosourea (ENU), which causes nucleotide

transitions, resulting in random point mutations [178]. Malignant gliomas are generally a rare

occurrence in p53-/-

mice, but ENU treatment of p53-/-

mice in utero generates gliomas in 50-

70% of p53-/-

mice with a median survival of approximately 10-12 weeks [179,180]. However,

this mouse model does not generate glioma exclusively, and the mice also succumb to

lymphoma, leukemia, sarcoma, and lung cancer [179,180]. The incidence of glioma formation is

increased if p53 knockout is targeted exclusively to the CNS; our lab has previously shown that

the Nestin-Cre mediated deletion of p53 in the CNS of mice combined with ENU treatment at

e13.5 causes 100% brain tumour incidence, 91% of which were identified histologically as high-

grade malignant gliomas, with a median survival of 19.3 weeks and the absence of other tumour

types [180]. Similar to human gliomas, the primary mouse gliomas were phenotypically

heterogeneous, with a high percentage (>70%) of these cells express markers of NSCs (Nestin+

and Sox2+), while a minority of cells express the mature cell markers of astrocytes (S100β

+),

neurons (Map2+), and oligodendrocytes (CNPase

+) [180]. Sorting for the cell surface marker

CD15 enriched for the clonogenic population, with tumours occurring in vivo from the orthotopic

injection of as few as 1,000 cells [180]. Stem cell lines derived from the mouse model of glioma

are associated with gene expression patterns that show similarities to both the typical cancer

genotype as well as the normal NSC program [180,181]. Most strikingly, the gene expression

profile of brain tumour stem cells correlates closely to the expression pattern of normal NSCs,

suggesting that there are relatively few changes in gene expression between normal and tumour

stem cells and that epigenetics play a significant role in the tumourigenic phenotype of these

cells. The CNS-specific deletion of p53, combined with ENU treatment in utero constitutes,

therefore, a clinically-representative and effective chemical-genetic mouse model of high-grade

glioma.

1.6.3 Brain Tumour Stem Cells

Cancer stem cells have been identified in human brain tumours and are likely responsible for

tumour progression and recurrence [143,182]. Dissociating and culturing primary adult or

pediatric brain tumours in standard neural stem cell conditions (serum-free medium with EGF

and bFGF) produced neurospheres that were phenotypically similar to normal neural stem cells

Page 49: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

34

in terms of self-renewal and differentiation into neurons, astrocytes, and oligodendrocytes under

appropriate conditions (growth factor withdrawal or addition of serum) [3,143,183,184,185].

The neurospheres grown from primary brain tumours engrafted in NOD/SCID mice

subcutaneously or intracranially and regenerated tumours that were phenotypically similar to the

primary human tumour [182].

Brain tumour stem cells (BTSCs) can be prospectively isolated from primary human

tumours by sorting for the neural stem cell surface marker, CD133 (Prominin-1) [12,143].

BTSCs comprise a subpopulation of cells within a primary tumour; CD133+ cells constitute a

range of 3.5-46% of cells in a human tumour, with higher tumour grades generally containing a

higher CD133+ fraction [143,182]. In fact, CD133

+ CSCs in GBM are associated with increased

radiation resistance compared to the non-CSCs comprising the tumour bulk, with CSCs

activating Chk1/2 DNA damage checkpoint kinases and the downstream damage response

machinery [158]. BTSCs have potent tumour initiating-ability in xenograft models; the injection

of as few as one hundred CD133+ BTSCs resulted in the formation of phenotypically

heterogeneous and serially transplantable brain tumour in NOD/SCID recipients, whereas 105

CD133- cells engrafted in the brain, but did not form tumours [182]. CD133 therefore enriches

for tumour-initiating cells.

Although it remains possible that other cell surface or intracellular markers identify CSCs

within brain tumours, CD133 is considered the most reliable marker of BTSCs. In mouse

models of glioma and medulloblastoma, sorting for CD15/SSEA-1 enriches for clonogenic cells

in vitro and tumour-initiating cells in vivo [180,186,187]. Markers such as CD15/SSEA-1 or

autofluorescence have been used to enrich for human glioma tumour-initiating cells in vitro and

in vivo [188,189]. Interestingly, autofluorescent glioma-initiating cells were found not only to

express Sox2, but also Nanog and Oct4, although these findings remain controversial in the field

[189].

BTSCs have many phenotypic similarities to normal NSCs, including the expression of

NSC markers (Nestin, Sox2, CD133) and the ability to self-renew or differentiate to produce

neurons, astrocytes, and oligodendrocytes. The regulation of self-renewal and differentiation of

BTSCs and normal NSCs is mediated by many of the same signaling pathways, including BMP,

Hedgehog and Notch. Increased BMP signaling has been shown to reduce BTSC proliferation

Page 50: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

35

and tumour-initiating ability, and promote neuronal differentiation in vitro and in vivo [190].

Hedgehog and Notch signaling also regulate normal NSC self-renewal and differentiation;

blockade of these pathways in BTSCs reduces self-renewal in vitro and tumour-initiating ability

in vivo [132,191]. BTSCs and normal NSCs can also be cultured as cell lines in vitro as

neurospheres or in adherent conditions on a matrix of poly-L-ornithine and laminin; adherent

culture conditions result in a more homogeneous cell population and offer some practical

advantages regarding infection, transfection, and clonal isolation [181]. Glioma BTSC lines can

also initiate aggressive malignant brain tumours in NOD/SCID mice upon injection of as few as

100 cells [181]. Therefore, the discovery of a tumour-initiating cell in human brain tumours and

the ability to isolate BTSCs based on the expression of CD133 allows for their expansion and

manipulation in vitro so that their genetics and epigenetics can be studied in depth.

1.7 Thesis Rationale, Hypothesis, and Specific Aims

1.7.1 Rationale

Cancer arises from a combination of genetic and non-genetic changes, including epigenetic

aberrations, all of which contribute to the neoplastic phenotype. Reprogramming somatic cell

nuclei by SCNT, cell fusion, or transcription factor-mediated methods dramatically alters the

epigenetic state of cells, resulting in a change in developmental potential [32]. The ability to

reprogram mouse models of medulloblastoma and melanoma by SCNT suggests that

reprogrammed cancer cell nuclei can direct normal development and that the neoplastic

phenotype can be at least partially attenuated [136,137]. In addition, normal mouse NSCs can be

reprogrammed by one or more transcription factors to generate iPS cells [86,89]. The gene

expression profile of mouse BTSCs correlates closely to the expression pattern of normal NSCs,

which may make mouse BTSCs amenable to reprogramming to the pluripotent state with few

exogenous factors. This study will provide an opportunity to assess the influence of induced

pluripotency on the self-renewal, differentiation, and tumour-initiating ability of BTSCs.

1.7.2 Hypothesis

We propose that CSCs, which are the driving force of brain tumour growth, can be

reprogrammed to pluripotency by the ectopic expression of a defined set of transcription factors

(Oct4, Klf4, Sox2, and c-Myc). Reprogramming BTSCs may alter the epigenetic contribution to

Page 51: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

36

the neoplastic phenotype and suppress tumourigenicity. If successful, this study will allow a

better understanding of the relative contributions of DNA genetic changes and non-genetic

cancer-associated changes to neoplastic behaviour.

1.7.3 Specific Aims

The overall goal of this project was to characterize BTSCs derived from a mouse model of

glioma in terms of pluripotency marker expression, and to generate iPS cells from BTSCs and

appropriate control NSCs by ectopic expression of transcription factors. A thorough molecular,

phenotypic, and functional characterization of the resulting iPS cell lines was performed,

including the assessment of in vitro and in vivo differentiation potential. Future experiments

involve the complete phenotypic characterization of NSC generated from the re-differentiation of

BTSC-derived iPS cell lines, which will also be assessed for tumourigenicity by in vivo

injections. The tumour-forming capacity of iPS cell-derived NSCs will be compared to that of

the BTSCs from which they were derived, with the goal of determining whether resetting the

epigenetic program of glioma BTSCs attenuates tumourigenicity. Comparing the gene

expression profiles of these cells may also aid in identifying novel genes or pathways that are

altered or differentially regulated in glioma tumours.

Page 52: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

37

Chapter 2

2 Materials and Methods

2.1 Characterization of Mouse Glioma Stem Cell Lines

2.1.1 Mouse Glioma Stem Cells

Our lab previously developed a clinically representative chemical-genetic mouse model of

glioma involving the Cre-mediated deletion of p53 in the central nervous system [180]. Nestin-

Cre;p53+/-

male mice were time-mated with p53flox/flox

females, which were then administered the

chemical carcinogen N-ethyl-N-nitrosourea at e13.5. This resulted in 100% spontaneous brain

tumour incidence in the offspring by 6 months of age [180]. Glioma stem cell lines were

established from dissection and dissociation of unsorted tumours and were grown in adherent

serum-free conditions, as described below.

2.1.2 Cell Culture and Differentiation

Mouse GNS and normal NS cell lines were grown as adherent monolayers in vitro in mouse

NSC medium (serum-free DMEM/F12 (1:1) with 2 µg/mL heparin, 20ng/ml EGF and 20ng/ml

bFGF with 2 µL laminin (Sigma, L2020) per 1 mL of culture medium on Primaria tissue culture

dishes (BD Biosciences) as previously described [3,181]. Cells were grown in a 37°C incubator

with 5% CO2 and cultures were replated every three days, with a split ratio of approximately 1 in

4. Cells were replated by mild dissociation treatment with 0.5-1 mL of Accutase (Sigma,

A6964) for approximately 5 minutes at 37°C, resuspended in DMEM, centrifuged, and

resuspended in complete NSC medium, from which approximately one quarter of the volume

was placed in a new dish and supplemented with 2 µL laminin per 1 mL of culture medium.

Serum-based differentiation was carried out in mouse NSC medium without growth factors,

supplemented with 10% fetal bovine serum (FBS; Gibco, 16000-044).

2.1.3 RNA Isolation and Reverse-Transcriptase PCR

Cells were lysed directly on the tissue culture dish with RLT buffer (Qiagen, 79216) containing

10 µL/mL β-mercaptoethanol, collected with a cell scraper, and frozen at -80°C for RNA

extraction. RNA was extracted with the QIAshredder (Qiagen, 79656) and RNeasy Mini Kit

Page 53: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

38

(Qiagen, 74106) according to the manufacturer’s instructions. An on-column DNase digestion

step was included (Qiagen, 79254). Synthesis of cDNA was carried out with Transcriptor

Reverse Transcriptase (Roche, 03 531 295 001) according to the manufacturer’s protocol, using

1 µg of RNA and oligo dT primers. PCRs were carried out for 35 cycles with primers to detect

Nanog, Oct4, Rex1, Klf4, Sox2, c-Myc, and GAPDH. Quantitative RT-PCR was carried out by

Stephanie Dobson (a rotation student in the lab) to assess the levels of expression of L-Myc, Klf4,

Klf2, Nanog, Sox2, Oct4, Klf5, and c-Myc in the glioma stem cell lines, p53-/-

NSCs, p53+/+

NSCs, and J1 mouse ES cells. See Appendix 1 for all primer sequences.

2.1.4 Alkaline Phosphatase Staining

Alkaline phosphatase staining was carried out for the three glioma NS cell lines, p53+/+

NS, and

p53-/-

NS cell lines, with J1 mouse ES cells and MEFs as positive and negative controls,

respectively, as described elsewhere [192]. Briefly, cells were fixed with 4% formaldehyde at

room temperature for 15 minutes, washed with PBS, and stained with a solution of 1 mg/mL Fast

Red TR hemi(zinc chloride) salt (Sigma, F8764) and 0.4 mg/mL naphthol phosphate disodium

salt (Sigma, N7255) in 0.1 M Tris-HCl, pH 8.6 for approximately 5 minutes. The enzymatic

product couples with the Fast Red TR salt to generate a red fluorescent azo dye adduct that can

be visualized by light or fluorescence microscopy [193]. Cells were washed several times with

PBS and stored in 4% formaldehyde for visualization with a Leica DMIL microscope fitted with

a Leica DC500 camera for imaging.

2.1.5 MTT Assay

Cells were seeded at a density of 2,000 cells per well in stem cell conditions (serum-free NSC

medium with EGF and bFGF) or growth factor-free medium with 10% (v/v) heat-inactivated

serum in 96-well tissue culture plates with 2 µL laminin per 1 mL of medium. Proliferation was

measured on days 5, 10, and 14 by adding 10 µL of 5 mg/mL MTT reagent (Roche, 11 465 007

001) to each well and incubating at 37°C for 4 hours. MTT solubilization buffer (10% SDS in

0.01 M HCl) was added at a volume of 100 µL per well and incubated overnight at 37°C.

Spectrophotometry was carried out at 575 nm using a Versamax microplate reader (Molecular

Devices, Sunnyvale, CA) and analyzed using SoftMax Pro software (Molecular Devices).

Page 54: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

39

2.2 Mouse iPS Cell Induction

2.2.1 EOS Lentiviral Infection

The early transposon promoter and Oct4 and Sox2 enhancer (EOS) lentivirus was produced as

described elsewhere and was provided by Dr. Akitsu Hotta from the Ellis Lab [192,194].

Twenty-four well plates were coated with 0.01% poly-L-ornithine (PLO; Sigma, P4957) for

approximately 20 minutes, washed with PBS, and coated with 5 µg/mL laminin in PBS

overnight. Target cells (GNS3, p53+/+

NS, and p53-/-

cell lines) were seeded at a density of

20,000 cells per well of a 24-well plate and infected the following day with 1 µl of EOS

lentivirus or PGK-GFP lentivirus in 500 µl of complete mouse NS cell medium containing 4

µg/mL polybrene (Sigma, 107689). The virus-containing medium was removed and replaced

with fresh mouse NS cell medium approximately 24 hours post-infection. GFP expression was

visualized using a Zeiss Axiovert 200M fluorescence microscope to assess infection efficiency

and cells were expanded into 6-well plates 5 days after infection. EOS-infected cell stocks were

frozen in complete mouse NS cell culture medium containing 10% (v/v) DMSO.

2.2.2 Retrovirus Production and Infection

The Platinum-E (Plat-E) packaging cell line was used to produce retroviruses and was cultured in

DMEM containing 10% (v/v) heat-inactivated FBS, 0.1 mM nonessential amino acids (Gibco,

11140-050), 1 µg/mL puromycin, and 10 µg/mL blasticidin. Plat-E cells were seeded at a

density of 2x106 cells in T-25 flasks and transfected with 10 µg of one of the following plasmids

using Opti-MEM I media (Gibco, 31985-070) and Lipofectamine 2000 (Invitrogen, 52887)

according to the manufacturer’s instructions: pMX-mOct4, pMX-mKlf4, pMX-mSox2, pMX-

mcMyc, or pMX-mRFP. Plat-E medium was replaced with complete mouse neural stem cell

medium supplemented with 0.1 mM nonessential amino acids approximately 16 hours after

transfection. Target cells were seeded at a density of 1x105

cells in 6-well plates coated with

0.01% PLO and 5 µg/mL laminin in PBS as previously described and infected with retroviruses

approximately 24 hours later. Briefly, viral supernatants were filtered through 0.45 µm low

protein binding filters (Pall Life Sciences, 4184) and viral cocktails were produced containing

two factors (Oct4 + Klf4), three factors (Oct4 + Klf4 + Sox2), or four factors (Oct4 + Klf4 +

Sox2 + c-Myc) with equal volumes of each factor and 4 µg/mL polybrene. Low-passage CD1

MEFs were infected with 3- and 4-factor combinations of retroviruses as a control.

Page 55: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

40

Approximately 24 hours post-infection, the virus-containing medium was replaced with fresh

mouse neural stem cell medium and RFP expression was visualized using a Zeiss Axiovert 200M

fluorescence microscope.

2.2.3 Isolation and Maintenance of iPS Cell Colonies

Two days after retrovirus infection, 1x105 cells infected with two, three, or four factors were

seeded on Mitomycin C-treated e15.5 MEF feeder layers in 100 mm tissue culture dishes

containing mouse ES cell medium consisting of Glasgow Minimum Essential Medium (GMEM;

Gibco, 11710-035) supplemented with 10% (v/v) heat-inactivated ES certified FBS (Gibco,

16141-079), 0.1 mM MEM nonessential amino acids (Gibco, 11140-050), 1 mM sodium

pyruvate (Gibco, 11360-070), 0.1 mM β-mercaptoethanol, and 100 units/mL recombinant human

LIF (Millipore, LIF1005). Approximately 4-6 weeks after infection, iPS colonies were isolated

from induction plates by manually picking colonies with a 27G needle, aspirating the colony

with a pipette, dissociating colonies in 0.025% trypsin-EDTA for approximately 10 minutes, and

replating dissociated colonies on fresh e15.5 MEF feeder layers in 24-well plates.

Approximately 3-5 days later, colonies were trypsinized and expanded to 12-well plates and

subsequently to 6-well plates, at which point iPS cell lines were frozen in complete ES cell

medium containing 10% (v/v) DMSO.

Mouse ES and iPS cell lines were routinely grown on Mitomycin C-treated e15.5 MEF

feeder layers in tissue culture dishes in mouse ES cell medium. Cells were propagated by

treatment with 0.025% trypsin-EDTA for approximately 5 minutes at 37°C, inactivating the

trypsin with an equal volume of complete medium, and replating cells on fresh feeder layers at a

split ratio of 1:10 to 1:15. The culture medium was replaced daily. For RNA extraction, feeder

depletion was performed by trypsinizing cells, inactivating trypsin by adding a 10-fold greater

volume of complete ES cell medium, and plating the cell suspension in 100 mm Nunclon Δ

tissue culture dishes (Thermo Scientific, 150350) for two sequential incubations of 10 minutes at

37°C to allow the MEFs to adhere to the plastic. Mouse ES and iPS cell lines were grown in

feeder-free conditions in 6-well plates (Corning, 3516) coated with 0.1% gelatin.

Dual inhibitor (2i) treatment was carried out in complete ES culture medium with 1 µM

PD0325901 (Santa Cruz Biotechnology, sc-205427) and 3 µM CHIR99021 (Stemgent, 04-0004)

or twice as concentrated (2i[2X]) to inhibit MAPK signaling and GSK3β, respectively [22,195].

Page 56: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

41

2.3 Molecular Characterization of iPS Cell Lines

2.3.1 Immunocytochemistry

Cells were seeded in 24-well plates (Falcon, 353047) coated with 0.1% gelatin and fixed in 4%

PFA for 10 minutes at room temperature two days later. Fixed cells were permeabilized with

0.2% NP-40 for 15 minutes at room temperature, washed in PBS, and blocked in 10% NGS for 1

hour at room temperature. Primary antibodies were diluted in 5% NGS and incubated overnight

at 4°C, as follows: rabbit anti-Nanog (1:500) (Abcam, ab80892); rabbit anti-Oct4 (1:500)

(Abcam, ab19857); rabbit anti-GFAP (1:1,000) (Dako, Z0334); mouse anti-Nestin (1:1,000) (BD

Pharmingen, 556309). Following three washes with PBS, secondary antibodies were diluted in

5% NGS and incubated at room temperature for 1 hour: anti-rabbit::A488 (1:500)

(Invitrogen/Molecular Probes, A11008) or anti-mouse::A488 (1:500) (Invitrogen/Molecular

Probes, A11029). After three washes with PBS, 1X DAPI (0.5 µg/mL) in fluorescent mounting

medium (Dako, S3023) was placed on glass coverslips and inverted onto the cells in each well.

All fluorescence imaging was performed on a Zeiss Axiovert 200M fluorescence microscope

with a Zeiss AxioCam HRm camera and analyzed using AxioVision software.

2.3.2 Quantitative RT-PCR

RNA extraction (following feeder depletion) and cDNA synthesis were performed as previously

described. Quantitative RT-PCR was carried out for 45 cycles using a Chromo4 DNA Engine

gradient cycler (PTC-200, MJ Research) and the data was analyzed with Opticon Monitor 3

software. See Appendix 1 for the list of primers used to detect exogenous retroviral vector

expression in iPS cell lines.

2.4 In Vitro Differentiation

2.4.1 Embryoid Body Assays

Mouse ES and iPS cell lines were grown in feeder-free conditions in 6-well plates (Corning,

3516) coated with 0.1% gelatin for three passages to deplete cultures of MEF feeder cells.

Embryoid body assays were initiated by seeding cells at a density of 2x106 cells in 10 mL of

mouse ES medium without LIF in 100 mm polystyrene Petri dishes (Fisher Scientific, 0875712).

The culture medium was changed every 2 days by transferring the cell suspension to a 15 mL

conical tube with a 10 mL pipette, allowing the EBs to settle to the bottom of the tube for

Page 57: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

42

approximately 5 minutes, and carefully aspirating the old medium. The EBs were gently

resuspended in 10 mL of fresh ES medium (without LIF) and transferred to a new 100 mm Petri

dish. EBs were visualized and imaged using a Leica DMIL light microscope with a Leica

DC500 camera. After 8 days in suspension culture, approximately 5-10 EBs were seeded in ES

medium (without LIF) in each well of a 24-well plate coated with 0.1% gelatin. The culture

medium was changed every 2 days and the adherent EBs were imaged using a Zeiss Axiovert

200M fluorescence microscope with a phase contrast filter and a Zeiss AxioCam HRm camera.

2.4.2 Immunocytochemistry for Germ Layer Markers

Embryoid bodies seeded in gelatin-coated 24-well plates were fixed after 6 days in adherent

conditions and permeabilized as previously described. Cells were blocked in 5% BSA for 1 hour

at room temperature and primary antibodies were incubated in 0.5% BSA overnight at 4°C as

follows: mouse anti-GATA4 (1:100) (Santa Cruz Biotechnology, sc-25310); rabbit anti-Alpha

Smooth Muscle Actin (1:100) (Abcam, ab5694); goat anti-Brachyury (1:20) (R&D Systems,

AF2085); mouse anti-Nestin (1:500) (BD Pharmingen, 556309); rabbit anti-βIII Tubulin (1:500)

(Abcam, ab52901). After three PBS washes, the appropriate secondary antibodies were

incubated in 0.5% BSA for 1 hour at room temperature, each at a dilution of 1:500: anti-

mouse::A568 (Invitrogen/Molecular Probes, A11004), anti-mouse::A488 (Invitrogen/Molecular

Probes, A11029), anti-rabbit::A568 (Invitrogen/Molecular Probes, A11036), anti-rabbit::A488

(Invitrogen/Molecular Probes, A11008), or anti-goat::A488 (Invitrogen/Molecular Probes,

A11055). After three washes with PBS, coverslips were mounted and the cells were visualized

as previously described.

2.5 In Vivo Differentiation

2.5.1 Teratoma Assays

All mouse procedures were carried out by Lilian Lee in accordance with the Hospital for Sick

Children’s Animal Care Committee guidelines. For intracranial injections, 2.5x105 cells in a

volume of 2.5 µl of PBS were injected into the frontal cortex of 5.5 week old female NOD/SCID

mice. The injection coordinates were 4 mm anterior of lambda, 2 mm to the right of the midline,

and 4.5-5.0 mm in depth. Subcutaneous injections were carried out three days later with cells of

one greater passage number. For subcutaneous injections, 1.5x106 cells were prepared in 50 µl

Page 58: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

43

of PBS, to which 50 µL of Matrigel were added (growth factor reduced; BD Biosciences,

354230). Each cell line was injected into the dorsal flank of 6 week old female NOD/SCID

mouse (one injection per side). Tumour formation was monitored over the course of 12 weeks,

and animals demonstrating symptoms were sacrificed by CO2 asphyxiation. Tumours were fixed

in 4% PFA overnight at 4°C and stored in 70% ethanol at 4°C until processed.

2.5.2 Histology and Immunohistochemistry

Tissue sections were generated using standard protocols for paraffin-embedded tissues, and

hematoxylin and eosin (H&E) staining was performed by standard methods.

Immunohistochemistry was carried out on paraffin-embedded sections of tumours or brains

following heat-mediated antigen retrieval. Briefly, deparaffinized sections were heated in citric

acid buffer (pH 6.0) in a pressure cooker, cooled and blocked in 5% BSA for one hour at room

temperature. Immunohistochemistry was performed using the following antibodies: mouse anti-

GATA4 (1:500) (Santa Cruz Biotechnology, sc-25310); rabbit anti-Alpha Smooth Muscle Actin

(1:500) (Abcam, ab5694); goat anti-Brachyury (1:20) (R&D Systems, AF2085); mouse anti-

Nestin (1:500) (BD Pharmingen, 556309); rabbit anti-GFAP (1:1,000) (Dako, Z0334).

Following three PBS washes, the appropriate secondary antibodies listed above were incubated

in 0.5% BSA for 1 hour at room temperature, each at a dilution of 1:500. Sections were washed

twice with PBS, stained with 1X DAPI in PBS for 10 minutes, washed briefly with PBS,

mounted in fluorescent mounting medium, and visualized as previously described.

2.6 Directed Differentiation to Neural Stem Cells

Directed differentiation of iPS and ES cells to generate NSCs was induced in serum-free

adherent conditions, as described elsewhere [11,196,197]. In brief, iPS and J1 mouse ES cell

lines were grown in feeder-free conditions in 6-well plates (Corning, 3515) coated with 0.1%

gelatin for three passages to remove MEF feeder cells from the cultures. ES and iPS cells were

seeded at a density of 1x104 cells/cm

2 in T25 flasks (Falcon, 353109) coated with 0.1% gelatin in

complete mouse ES cell medium. Approximately 24 hours later, the culture medium was

removed and replaced with N2B27 medium, consisting of a 1:1 mixture of DMEM/F12

supplemented with modified N2 (25 µg/mL insulin, 100 µg/mL apotransferrin, 6 ng/mL

progesterone, 16 µg/mL putrescine, 30 nM sodium selenite, and 50 µg/mL bovine serum

albumin fraction V) and Neurobasal medium (Gibco, 21103) supplemented with B27 (Gibco,

Page 59: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

44

17504). Fresh N2B27 medium was replenished every 2 days. After 7 days, 3x106 cells were

replated in uncoated 100 mm Nunclon Δ tissue culture dishes (Thermo Scientific, 150350) in NS

expansion medium consisting of mouse NeuroCult basal medium (Stem Cell Technologies,

05700) supplemented with modified N2 and 10 ng/mL of both EGF and bFGF. The culture

medium was replenished 3 days later. Five days after the transfer to NSC expansion medium

containing EGF and FGF, cellular aggregates growing in suspension were collected, pelletted by

gravity, and dissociated by accutase treatment for approximately 10 minutes. Cells were seeded

in 100 mm Primaria tissue culture dishes (Falcon, 353803) in NSC expansion medium

supplemented with 2 µL laminin per 1 mL of medium. Two days later, cells were replated in

Primaria tissue culture dishes coated with PLO/laminin as previously described to establish NSC

lines. Early passage iPS and ES cell-derived NS-like cell lines were frozen in NSC expansion

medium with 10% (v/v) DMSO for future experiments.

Page 60: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

45

Chapter 3

3 Results

I generated all of the data and figures in the Results section; Figure S1 in Appendix 2 was

produced by Stephanie Dobson, a rotation student that I supervised in the laboratory.

3.1 Characterization of Mouse Glioma Stem Cell Lines

3.1.1 Mouse Glioma Stem Cells Express Neural Stem Cell Markers, but

Not Core Pluripotency Markers

The expression of pluripotency factors was assessed in the three glioma stem cell lines derived

from independent tumours arising in the mouse model of glioma (GNS1, GNS2, and GNS3), and

in non-neoplastic p53-/-

NS, and p53+/+

NS cell lines. RT-PCR analysis showed that all five of

the cell lines grown in stem cell conditions (serum-free medium containing EGF and bFGF)

failed to express the core pluripotency factors Nanog, Rex1, and Oct4, but expressed the neural

stem cell-associated factors Sox2, Klf4, and c-Myc at levels comparable to J1 mouse ES cells

(Figure 4). The expression of pluripotency factors was also assessed in medium containing 10%

serum to determine whether the expression levels of Sox2, Klf4, and c-Myc were maintained in

differentiation conditions. It was found that the cell lines continued to express Sox2, Klf4, and c-

Myc transcripts when grown in serum-containing medium for one week, despite strong induction

of expression of differentiated markers [180].

The results were validated by quantitative RT-PCR analysis to detect the expression of L-

Myc, Klf4, Klf2, Nanog, Sox2, Oct4, Klf5, and c-Myc (Appendix 2, Figure S1). Quantitative RT-

PCR confirmed that Nanog and Oct4 expression levels are essentially undetectable in glioma

stem cells and normal NSCs (with the exception of low detection levels in GNS2 in

differentiation conditions). Expression levels of L-myc, Sox2, and particularly c-Myc were

increased in glioma stem cells and normal NSCs compared to ES cells. Klf4 expression

increased in differentiation conditions in two of the three glioma stem cell lines relative to ES

cells. Therefore, mouse glioma stem cells express the same reprogramming-associated factors

(Sox2, Klf4, and c-Myc) as normal neural stem cells, but fail to express genes associated with the

core pluripotency program (Nanog, Rex1, and Oct4). The expression of endogenous Nanog,

Page 61: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

46

Rex1, and Oct4 would therefore be an ideal readout to assess the pluripotency of these cells after

reprogramming.

Figure 4. Mouse glioma stem cell lines express neural stem cell-associated factors, but do

not express RNA encoding core pluripotency-associated factors.

The three mouse glioma stem cell lines (GNS1, GNS2, and GNS3), p53-/-

NS, and p53+/+

NS

cell lines failed to express the core pluripotency-associated factors Nanog, Rex1, and Oct4

in stem cell conditions (serum-free medium with EGF and bFGF) and in differentiation

conditions (medium containing 10% serum). The five cell lines expressed the neural stem

cell-associated factors Sox2, Klf4, and c-Myc in stem cell conditions or differentiation

conditions at levels comparable to J1 mouse ES cells (used as a positive control), as detected

by RT-PCR. GAPDH was a housekeeping gene used as a technical control.

Page 62: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

47

3.1.2 Mouse Glioma Stem Cells Express Alkaline Phosphatase

Alkaline phosphatase (AP) staining is used as a rapid and nonspecific assay to differentiate

between pluripotent ES cells and differentiated cells such as fibroblasts, since ES cells express

AP, whereas differentiated cells fail to express this enzyme. The product of the hydrolysis

reaction catalyzed by AP couples with the Fast Red TR salt to generate a red fluorescent

compound that can be visualized by light or fluorescence microscopy.

The three mouse glioma stem cell lines grown in stem cell conditions expressed relatively

high levels of AP compared to J1 mouse ES cells, whereas the p53+/+

and p53-/-

NS cell lines did

not express detectable levels of AP (Appendix 2, Figure S2). Growing the glioma NS, p53+/+

NS, and p53-/-

NS cell lines in medium containing 10% serum significantly reduced AP

expression (Appendix 2, Figure S3). The detection of AP expression in the GNS cell lines in

stem cell conditions, therefore, made this an unreliable assay to use in subsequent experiments

involving reprogrammed cells.

3.1.3 Serum Alters the Growth Rate of Glioma and Normal Neural Stem Cells

Preliminary reprogramming experiments resulted in overgrowth of non-reprogrammed glioma

stem cells in the induction plates, so the growth rates of the three glioma stem cell lines, p53-/-

NS, and p53+/+

NS cell lines were examined in stem cell conditions (serum-free medium

supplemented with EGF and bFGF) and differentiation conditions (growth factor-free medium

supplemented with 10% serum). The cell lines GNS1 and GNS2 had similar growth rates in

stem cell and differentiation conditions, whereas the GNS3 cell line grew significantly slower in

medium containing serum than in serum-free medium (Figure 5). The GNS3 cell line was

therefore selected for subsequent reprogramming experiments, in order to optimize differential

growth of non-reprogrammed and reprogrammed cells in serum-containing ES cell medium

during the induction process.

Page 63: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

48

Figure 5. Serum alters the growth rate of glioma stem cell and neural stem cell lines.

The proliferation of one of the three glioma stem cell lines (GNS3) is reduced in serum-

containing medium compared to mouse NSC (mNSC) culture conditions, similar to p53-/-

and p53+/+

NSC lines. The GNS3 cell line was therefore chosen for reprogramming

experiments. Values are means ± SEM; * denotes p<0.05 based on Student’s t-test.

Page 64: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

49

3.2 Defined Factors Can Reprogram Mouse Glioma Stem Cells

3.2.1 Induction of Reprogramming with Four Transcription Factors

Preliminary experiments involving the piggyBac-mediated reprogramming of GNS1, GNS2, and

GNS3 with Oct4 and Klf4 did not yield any iPS cell lines, since the glioma stem cell lines grew

too rapidly in serum-containing media; therefore, non-reprogrammed cells took over the culture.

Also, selection for the expression of the Oct4 plasmid or the EOS vector with hygromycin or

puromycin, respectively, proved to be challenging, since the glioma stem cell lines are

intrinsically highly resistant to both hygromycin and puromycin, and the MEF feeder layers are

not resistant at such high concentrations. Colonies with characteristic iPS cell morphology were

not generated from the glioma NS, p53-/-

NS, or p53+/+

NS cell lines infected with EOS, after the

piggyBac-mediated transposition of Oct4 and Klf4.

Subsequent reprogramming attempts involved the retroviral-mediated transduction of

GNS3 with combinations of two, three, and four of the reprogramming factors. Two-factor and

three-factor inductions of GNS3 without c-Myc did not generate iPS cell colonies that could be

manually isolated from the induction plate, since the glioma stem cells continued to proliferate

during the 4-6 week induction period and overtook any colonies that might have formed. The

addition of c-Myc as a reprogramming factor increased the efficiency at which colonies were

generated and reduced the amount of time required for the emergence of iPS cell colonies that

were large enough to be picked. CD1 MEFs were reprogrammed in parallel and generated iPS

cell-like colonies from three- and four-factor inductions (without and with c-Myc). Even though

it was not possible to isolate iPS cell colonies from two- or three-factor infections of GNS3,

colonies resembling iPS cells were isolated from four-factor infections of GNS3, p53+/+

NS, and

p53-/-

cell lines (Table 1). Some of the iPS cell lines were derived from EOS-infected cells,

while others were not.

Page 65: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

50

Cell Line Number of

Colonies Picked

Number of Clones

(12-well plate stage)

Number of Clones

(final 6-well plate stage)

GNS3

GNS3-EOS

30

36

12

7

6

4

p53-/- NS

p53-/- NS-EOS

48

18

12

4

7

3*

p53+/+ NS

p53+/+-NS-EOS

CD1 (MEFs)

24

42

24

13

16

15

2

7

12

Table 1. Summary of the isolation of iPS cell colonies derived from four-factor inductions

and the number of clones derived.

(* Indicates clones that had lost typical iPS cell morphology.)

Page 66: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

51

3.2.2 Induced Pluripotent Stem Cell Lines Express Pluripotency Factors, but Not Neural Stem Cell or Astrocytic Cell Markers

Approximately 4-6 weeks after infecting GNS3, p53+/+

NS, and p53-/-

NS cell lines with pMX-

based retroviruses encoding Oct4, Klf4, Sox2, and c-Myc, and culture on feeders in medium

containing serum and LIF, iPS colonies were isolated and expanded in culture. The majority of

the iPS cell lines had silenced retroviral RFP expression, whereas others failed to fully silence

RFP, indicating that different degrees of reprogramming had been achieved. Nanog and Oct4

protein expression was detected by immunocytochemistry, but at lower levels compared to J1

mouse ES cells (Figure 6A). Eight p53-/-

NS-iPS, four p53+/+

NS-iPS, and six GNS-iPS cell lines

were selected for RT-PCR analysis, which revealed the activation of expression of endogenous

pluripotency factors, Nanog, Oct4, and Rex1 in some of the clones (Figure 6B). CD1 MEFs

were reprogrammed in parallel and generated iPS-cell like colonies that were isolated, the

majority of which expressed Nanog (Appendix 2, Figure S4).

The iPS cell lines that were chosen for subsequent experiments and further

characterization (p53-/-

NS-iPS1, p53-/-

NS-iPS2, p53+/+

NS-iPS1, p53+/+

NS-iPS2, GNS-iPS1,

and GNS-iPS2) were selected based on expression of endogenous pluripotency factors and

maintenance of iPS cell-like morphology. Candidate iPS cell lines that failed to generate iPS cell

colonies after several passages in culture were discarded. Two of the iPS cell lines (GNS-iPS2

and p53+/+

NS-iPS2) were derived from EOS-infected cells, although no GFP+ colonies were

observed. Failure to activate EOS expression was attributed to low infection efficiency of the

EOS lentivirus.

Dual inhibition of MAPK and GSK3 signaling by treatment with the 2i cocktail enhanced

Nanog expression in the GNS-iPS2 cell line, as detected by quantitative RT-PCR analysis [195]

(Figure 6C). The p53-/-

NS-iPS1 cell line had been treated with 2i prior to RT-PCR analysis.

Nanog expression was also enhanced in CD1 MEF-derived iPS cell lines as a result of 2i

treatment, as detected by end-point RT-PCR analysis (Appendix 2, Figure S4).

Page 67: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

52

A

B C

Figure 6. Molecular characterization of iPS cell lines reveals the expression of core

pluripotency-associated factors.

(A) Representative figures of the morphology of p53+/+

NS-iPS, p53-/-

NS-iPS, and GNS-iPS

cell lines that have silenced retroviral RFP expression. Low levels of Nanog and Oct4

protein expression were detected by immunocytochemistry (with DAPI nuclear staining

inset). J1 mouse ES cells are shown as a positive control. (B) The expression of

endogenous pluripotency factors (Nanog, Rex1, and Oct4), as detected by RT-PCR. MEFs

and J1 mouse ES cells were used as negative and positive controls, respectively. The iPS

cell lines chosen for subsequent experiments (labeled clones 1 and 2) are highlighted by

dashed boxes. The p53-/-

NS-iPS1 and GNS-iPS2 cell lines had been treated with 2i and

2i[2X], respectively. (C) Nanog expression was enhanced in a glioma-derived iPS cell line

by dual inhibition (2i) of MAPK signaling and GSK3β by 1 µM PD0325901 and 3µM

CHIR99021, respectively, as determined by quantitative RT-PCR. Values are means ±

SEM for three biological replicates; * denotes p<0.05 (Student’s t-test).

Page 68: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

53

The expression levels of the neural stem cell marker Nestin and the astrocytic cell marker

GFAP were assessed by immunocytochemistry in mouse ES cell culture conditions. The

reprogrammed cells and J1 mouse ES cells failed to express Nestin, and only rare cells stained

positive for GFAP (Figure 7A). Control experiments for p53+/+

NS and GNS3 cell lines were

carried out in stem cell conditions (serum-free medium with EGF and bFGF) in parallel with

differentiation conditions (growth factor-free medium with 10% serum). Both cell lines

expressed Nestin in stem cell conditions, but the expression of this neural stem cell marker was

reduced upon growth factor withdrawal and serum addition (Figure 7B). This was especially

apparent for the glioma stem cell line, which did not express detectable levels of Nestin in

differentiation conditions. By contrast, the wild type NSCs and GNS3 cell line expressed low

levels of GFAP in stem cell conditions and significantly higher levels of the astrocytic cell

marker in differentiation conditions. Since mouse ES cell medium contains serum, it can be

concluded that the reprogrammed cells did not retain phenotypic marker memory of the cell of

origin, since the iPS cell colonies did not express the astrocytic cell marker GFAP.

Page 69: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

54

A

B

Figure 7. Reprogrammed neural stem cells and glioma stem cells fail to express neural

stem cell or astrocytic cell markers.

(A) Immunocytochemistry revealed negligible expression of the neural stem cell marker

Nestin and rare cells expressing the astrocytic cell marker GFAP in reprogrammed cell

lines, similar to J1 mouse ES cells. (B) Control experiments showing the reduced

expression of Nestin and increased expression of GFAP in p53+/+

NS and GNS3 cell lines in

differentiation conditions (10% serum) compared to stem cell conditions.

Page 70: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

55

3.2.3 Retroviral Silencing in Reprogrammed Cell Lines

Pluripotent stem cells and fully-reprogrammed iPS cells silence retroviral vector expression

[198]. The expression levels of retroviral transcripts encoding exogenous Oct4, Klf4, Sox2, and

c-Myc were assessed by quantitative RT-PCR for the six iPS cells lines selected, control four

factor-infected cell lines, and J1 mouse ES cells (Figure 8). The data for each iPS cell line was

compared to the expression levels of the respective control cell line using RNA harvested within

one week of infection with the four factor (4F) retroviral cocktail and normalized to GAPDH.

The results from three biological replicates suggest that the expression of the retroviral

vector encoding Oct4 was silenced in both of the glioma-derived and p53-/-

NSC-derived iPS cell

lines. Both of the GNS-iPS cell lines silenced exogenous Sox2 and c-Myc expression, although

only GNS-iPS1 silenced exogenous Klf4 expression. The two p53-/-

NSC-derived iPS cell lines

failed to silence the expression of retroviruses encoding Klf4, Sox2, and c-Myc. Both of the

p53+/+

NSC-derived iPS cell lines failed to silence any retroviral vector expression, with p53+/+

NS-iPS2 showing increased retroviral vector expression of all four of the exogenous factors

compared to the freshly infected 4F control. It should be taken into account that the 4F controls

were not sorted to isolate the population of cells with successful viral integration; therefore, the

bulk culture may reflect lower retroviral expression, since only approximately 80% infection

efficiency was obtained. The iPS cell colonies from which the cell lines were derived, on the

other hand, presumably arose as a result of successful viral integrations, so the majority of cells

in the bulk iPS cell lines contained the retroviral vectors.

Page 71: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

56

Figure 8. Analysis of retroviral vector expression in iPS cell lines.

The expression of exogenous (Exo) retroviral vectors encoding Oct4, Klf4, Sox2, and c-Myc

was analyzed by quantitative RT-PCR for glioma NS (GNS)-derived iPS p53-/-

NS-derived,

and p53+/+

NS-iPS cell lines. The expression levels are shown relative to the appropriate

freshly infected four factor (4F) control cell line. J1 mouse ES cells are shown as a negative

control and have <0.01 fold expression relative to the 4F control. Varying degrees of

retroviral vector silencing were observed, with both of the GNS-iPS cell lines silencing the

majority of the retroviruses (with the exception of exo-Klf4 in GNS-iPS2). Exo-Oct4 was

silenced in both of the p53-/-

NS-iPS cell lines, but they failed to silence the expression of the

other retroviral vectors. Both of the p53+/+

NS-iPS cell lines failed to silence retroviral

vector expression. The results were obtained from three biological replicates normalized to

GAPDH; values are means ± SEM.

J1

Page 72: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

57

3.3 Altered Differentiation Potential of p53-Deficient iPS Cells

3.3.1 Induced Pluripotent Stem Cell Lines Generate Embryoid Bodies with Reduced Endodermal Differentiation in vitro

Embryoid body (EB) assays were used to test the in vitro differentiation capacity of the iPS cell

lines derived from GNS3, p53+/+

NS, and p53-/-

NS cell lines. All of the iPS cell lines that were

tested formed EBs in suspension culture in serum-containing medium without LIF and produced

differentiated outgrowths of cells when grown in adherent conditions on gelatin (Figure 9A).

The EBs formed from glioma-derived iPS cell lines were more irregularly-shaped than those

formed from p53+/+

or p53-/-

NS cells or from ES cells. The non-reprogrammed GNS3, p53+/+

NS, and p53-/-

NS cell lines from which the iPS cells were derived failed to form embryoid

bodies under the same culture conditions after being transferred from stem cell culture conditions

(serum-free media with EGF and FGF) or from ES culture conditions (serum-containing media

with LIF). The GNS3 cells adhered to the dishes and did not form aggregates in suspension,

whereas the majority of the p53+/+

NS and p53-/-

NS cells died, especially when cells experienced

LIF withdrawal after being transferred from serum-containing medium with LIF to EB medium

without LIF. Therefore, embryoid body formation was a property associated with only the

reprogrammed cell lines and not the parental cell lines.

Immunocytochemistry was performed for the embryoid body differentiation assays.

After 8 days of suspension culture and 6 days of adherent culture conditions, embryoid bodies

were stained for markers of endoderm (GATA4), mesoderm (α-Smooth Muscle Actin), and

ectoderm (Nestin and βIII Tubulin) (Figure 9B). Cells staining for ectoderm and mesoderm

lineage markers were detected for all of the iPS cell lines, but endodermal lineage markers were

less readily detected. The in vitro differentiation of J1 mouse ES cells was carried out in parallel

and revealed the definitive presence of GATA4+ cells, which suggests that cells of the

endodermal lineage were generated by differentiated J1 cells, but that the iPS cell lines may have

a preference for the mesodermal and ectodermal lineages during in vitro differentiation.

The expression of markers of mesoderm (Flk1 and Brachyury), endoderm (Sox17 and

GATA4), and ectoderm (Nestin and Sox1) was also assessed by end-point RT-PCR for feeder-

depleted ES and iPS cell lines in ES cell conditions (serum-containing medium with LIF; Figure

Page 73: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

58

10A) and EBs after 4 and 8 days in suspension culture in differentiation conditions (serum-

containing medium with LIF; Figure 10B). The amplification of Flk1 and Nestin transcripts in

ES cell conditions occurred for the majority of the iPS cell lines tested, in addition to J1 mouse

ES cells, which also expressed detectable levels of all of the other germ layer markers. This

suggests that some spontaneous differentiation may have occurred in culture and that low levels

of transcripts encoding differentiated cell markers may have been present. Upon removal of LIF

in differentiation conditions, additional germ layer markers were detected in the iPS cell-derived

EBs, including Sox17 and Sox1, in addition to Flk1 and Nestin. Although Brachyury was only

detected in ES cell-derived EBs at day 4, Flk1 was detected in both of the glioma stem cell-

derived iPS and one p53+/+

NSC-derived iPS cell lines. Similarly, GATA4 was only detected in

ES cell-derived EBs, but another endodermal cell marker, Sox17, was detected at relatively high

levels in GNS-iPS2 and both of the p53+/+

NS-iPS cell lines. Nestin and Sox1 were detected in

all of the EB populations after 8 days in differentiation conditions. Interestingly, EBs formed

from one of the glioma stem cell-derived iPS cell lines (GNS-iPS2) expressed markers of

mesoderm (Flk1), endoderm (Sox17) and ectoderm (Nestin and Sox1) after 8 days in

differentiation conditions. Similarly, EBs formed from p53+/+

NS-iPS2 or J1 ES cells expressed

these four germ layer markers, with J1-derived EBs also expressing Brachyury and GATA4. The

minimal expression of germ layer markers by p53-/-

NSC-derived iPS cell lines suggests that a

potential blockage in differentiation may have occurred.

Page 74: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

59

A

B

Figure 9. Glioma stem cell-derived iPS cell lines form EBs and generate cells of

mesodermal and ectodermal lineages in vitro, as detected by immunostaining.

(A) EBs were formed in suspension culture in serum without LIF for each of the iPS cell

lines tested (top panels) and produced differentiated outgrowth of cells when grown in

adherent conditions on gelatin-coated dishes (bottom panels). Representative figures are

shown. (B) Representative figures of immunocytochemistry performed for adherent EBs.

Differentiated cells stained positive for the mesodermal marker α-Smooth Muscle Actin

(αSMA), whereas the endodermal marker GATA4 was only detected in the outgrowth from

EBs formed from J1 mouse ES cells. Ectodermal cells were detected in all of the EB assays,

based on the expression of the neural lineage markers Nestin and βIII Tubulin (βIII Tub).

Page 75: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

60

A

B

Figure 10. Differentiated cell markers are detectable at the transcript level in

undifferentiated iPS and ES cell populations, but are enhanced during EB differentiation.

(A) Undifferentiated iPS cell lines expressed detectable Flk1 and Nestin at the transcript

level, whereas ES cells (J1) expressed low levels of additional differentiation markers, even

in the presence of LIF. (B) Markers of mesodermal, endodermal, and ectodermal cells

were expressed by iPS cell lines and J1 cells after 4 or 8 days in differentiation conditions

(serum-containing medium without LIF). Of note, EBs formed from GNS-iPS2 expressed

Flk1, Sox17, Nestin, and Sox1 after 8 days in differentiation conditions.

Page 76: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

61

3.4 p53-Deficient iPS Cell Lines Generate Highly Undifferentiated Tumours

Teratoma assays were carried out to assess in vivo differentiation of the iPS cell lines, by the

subcutaneous injection of 1.5x106 cells into the dorsal flank of NOD/SCID mice, with J1 mouse

ES cells injected as a positive control. Subcutaneous tumours were obtained for each iPS cell

line injected and for J1 mouse ES cells with a latency of 2-6 weeks (Table 2). The glioma stem

cell-derived and p53-/-

NSC-derived iPS cell lines grew at similar rates as J1 cells, whereas the

p53+/+

NSC-derived iPS cell lines grew slowest in vivo. Visible growths were produced in the

dorsal flanks of NOD/SCID mice and the tumours had similar gross pathology (Figure 11). Non-

reprogrammed GNS3 cells generated a tumour at one of the two subcutaneous injection sites.

No tumours were obtained from the subcutaneous injection of p53-/-

or p53+/+

NSCs, or for

mouse embryonic fibroblasts, which were injected as a negative control. The tumour-initiating

capacity of the iPS cell lines in the forebrain was assessed by injecting 2.5x105 cells

intracranially into NOD/SCID mice, which resulted in tumour formation for 4 of the 6 iPS cell

lines (representing each genotype) in 2.5-12 weeks (Table 2). The intracranial injection of 1x105

of the non-reprogrammed glioma stem cells had previously been carried out and tumours were

generated in 3.5-8 weeks in NOD/SCID mice [180]. Intracranial tumours were not generated

from the injection of non-reprogrammed p53-/-

or p53+/+

NSCs in NOD/SCID recipients.

Histological analysis of the tumours was carried out following H&E staining (Figure 12).

The primary tumour in the mouse model was identified as a high-grade malignant glioma. The

non-reprogrammed GNS3 cells produced an anaplastic brain phenotype tumour following

injection into the brain or subcutaneous space of immune-deficient mice. The subcutaneous

tumours produced from the injection of p53-/-

NSC-derived iPS and glioma stem cell-derived iPS

cell lines were highly undifferentiated, resembling primitive malignant tumours. These tumours

were anaplastic, with a high nuclear to cytoplasmic ratio. A p53+/+

NSC-derived iPS cell line

also generated a highly undifferentiated tumour, whereas another p53+/+

NSC-derived iPS cell

line produced a differentiated subcutaneous tumour, similar to the teratoma produced from the

injection of J1 mouse ES cells. These teratomas contained structures representative of the three

germ layers, such as neural rosettes or keratinizing epithelium (ectoderm), gut-like epithelium

(endoderm), and cartilage or muscle (mesoderm). Phenotypically similar forebrain tumours were

Page 77: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

62

generated for each sample injected intracranially. Taken together, these results suggest that p53

status may play a role in the differentiation of iPS cell lines in vivo.

Cell Line Subcutaneous Tumours Intracranial Tumours

GNS-iPS 2 weeks (2/2) 2.5 weeks (2/2)

p53-/-

NS-iPS 2-2.5 weeks (2/2) 5 weeks (1/2)

J1 Mouse ES 2.5 weeks (1/1) 3 weeks (1/1)

Parental GNS 3 weeks (1/1) 3.5-8 weeks* (1/1)

p53+/+

NS-iPS 4.5-6 weeks (2/2) 12 weeks (1/2)

p53-/-

NS (0/1) (0/1)

p53+/+

NS (0/1) (0/1)

MEFs (0/1) ND

Table 2. Summary of the latency of subcutaneous and intracranial tumour formation.

NOD/SCID mice were sacrificed 2-6 weeks after the subcutaneous injection of 1.5x106 cells

in the dorsal flank or 2.5-12 weeks after the intracranial injection of 2.5x105 cells in the

forebrain, due to tumour formation. Non-reprogrammed p53-/-

and p53+/+

NSCs failed to

form subcutaneous or intracranial tumours. MEFs were injected subcutaneously as a

negative control and failed to generate tumours. Data indicates the number of cell lines

generating tumours/number of cell lines injected. *Obtained from previous experiments

[180]; ND, not determined.

Page 78: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

63

A

B

Figure 11. Reprogrammed NSCs and glioma stem cells generate subcutaneous tumours

with similar macroscopic appearance.

(A) Representative images of the growths generated 2 weeks after the subcutaneous

injection of 1.5x106 reprogrammed glioma stem cells (GNS-iPS1 and GNS-iPS2) in the

dorsal flanks of NOD/SCID recipients (arrows point to tumours). (B) The reprogrammed

cell lines generated 2-6 weeks after the subcutaneous injection of p53-/-

NS-iPS cells (#1 and

2), GNS-iPS cells (#3 and 4), p53+/+

NS-iPS cells (#5 and 6), J1 mouse ES cells (#7), and the

parental non-reprogrammed glioma stem cell line (#10).

Page 79: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

64

Figure 12. Glioma stem cell-derived iPS cell lines generate highly undifferentiated tumours

in vivo.

The primary high-grade malignant mouse glioma generated glioma stem cell lines that

produced anaplastic tumours when injected subcutaneously or orthotopically into

NOD/SCID mice. Tumours generated from p53-/-

NSC-derived iPS cell lines (p53-/-

NS-

iPS2 shown) and glioma stem cell-derived iPS cell lines were atypical, undifferentiated, and

exhibited large regions of necrosis. One p53+/+

NSC-derived iPS cell line generated highly

undifferentiated tumours, whereas the other p53+/+

NSC-derived iPS cell line produced

differentiated tumours similar to those produced from the injection of J1 mouse ES cells.

These tumours contained structures representative of multiple germ layers.

Page 80: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

65

3.5 Reprogrammed Glioma-Derived iPS Cell Lines Have Reduced Astrocytic Differentiation in vivo

In order to confirm the presence or absence of differentiated cells in the teratomas,

immunohistochemical analysis was carried out using markers of the three germ layers: Nestin

and GFAP (ectoderm), GATA4 (endoderm), and α-Smooth Muscle Actin (mesoderm).

Immunohistochemical analysis of the subcutaneous and intracranial tumours revealed similar

results, regardless of the injection site, and showed significantly different differentiation

capacities between the reprogrammed cell lines (Figure 13). One of each of the p53+/+

and p53-/-

NS cell-derived iPS lines generated tumours with extensive differentiation to generate GATA4+

endoderm, α-Smooth Muscle Actin+ (αSMA

+) mesoderm, and Nestin

+ ectoderm, whereas the

markers were less frequently detected in tumours generated from the other p53+/+

NS cell-

derived iPS cell line, albeit still detected. The other p53-/-

NS cell-derived iPS cell line and both

of the glioma stem cell-derived iPS cell lines generated tumours that did not contain any

GATA4+ cells (Figure 13A), suggesting impaired endodermal differentiation in vivo. Tumours

formed from the injection of glioma stem cell-derived iPS cell lines showed reduced expression

of the astrocytic cell marker, GFAP, in comparison to tumours generated from the non-

reprogrammed GNS cell line or from p53-/-

NSC-derived iPS cells (Figure 13B). GFAP

expression was also absent from forebrain tumours generated from the intracranial injection of

glioma stem cell-derived iPS cells. These results suggest that reprogrammed glioma stem cells

have reduced differentiation potential in vitro and in vivo, although the analysis of additional

glioma stem cell-derived iPS cell lines may be required to strengthen this finding.

Page 81: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

66

A

B

Figure 13. Glioma stem cell-derived iPS cell lines generate tumours expressing

mesodermal and ectodermal cell markers, but have significantly reduced astrocytic

differentiation.

(A) Immunohistochemistry shows the expression of markers of endoderm (GATA4) and

mesoderm (αSMA). Glioma stem cell-derived iPS cell lines form tumours that fail to

differentiate into endodermal cells. (B) Immunohistochemistry reveals that glioma stem

cell-derived iPS cell lines have reduced expression of the astrocytic cell marker GFAP in

vivo compared to the non-reprogrammed glioma stem (GNS) cell line or J1 mouse ES cells.

Page 82: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

67

3.6 Re-Differentiation of Glioma-Derived iPS Cell Lines to a Neural Stem Cell Fate

The directed neural re-differentiation of the iPS cell lines was carried out in parallel with J1

mouse ES cells in serum-free adherent conditions to establish iPS cell-derived NSC lines

[11,196,197]. Significant cell death was observed when the cells were transferred from ES cell

conditions to N2B27, which normally occurs when cells are transferred from serum-containing

medium to serum-free conditions [197]. Immunostaining for Nestin and Oct4 on days 3 and 5 of

culture in N2B27 medium showed that Nestin expression increased and Oct4 expression

decreased slightly by the fifth day in N2B27 conditions compared to ES cell conditions (Figure

14A). On the seventh day of culture in N2B27 medium, the cells were transferred to NSC

expansion medium containing EGF and bFGF, and spherical aggregates of cells formed within

three days. The spheres were dissociated and cells adhered to PLO/laminin-coated tissue culture

dishes. The early-passage NS-like cells showed typical NSC bipolar morphology (Figure 14B)

and proliferated in serum-free NSC conditions in the presence of EGF and bFGF.

Taken together, the results suggest that BTSCs derived from a clinically-representative

mouse model of glioma are amenable to reprogramming with a defined set of transcription

factors. The resulting glioma stem cell-derived iPS cell lines activated the expression of

endogenous pluripotency factors and silenced the expression of exogenous reprogramming

factors. Reprogrammed glioma stem cells formed EBs that expressed endodermal, mesodermal,

and ectodermal cell markers at the transcript level and generated undifferentiated tumours in

vivo. Preliminary directed re-differentiation experiments of the reprogrammed cell lines suggest

that NS-like cells can be generated from CSC-derived iPS cell lines.

Page 83: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

68

A

B

Figure 14. Preliminary directed re-differentiation experiments generated Nestin+ cells with

neural stem cell-like morphology.

(A) Preliminary immunocytochemistry results of directed re-differentiation experiments to

generate neural stem cells from GNS-iPS cells suggest that Nestin expression increased,

while Oct4 expression decreased slightly during the first 5 days in N2B27 medium. Day

zero represents the starting population in ES cell medium. (B) Phase contrast images of

iPS cell-derived NS-like cells expanded in NSC conditions (serum-free medium with EGF

and bFGF). The appearance of bipolar NSC morphology suggests the directed

differentiation to a NSC fate.

Page 84: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

69

Chapter 4

4 Discussion and Future Directions

4.1 Summary of Principle Findings

This project has shown a proof-of-principle that mouse glioma stem cells, which fail to express

defining pluripotency factors Nanog, Oct4, and Rex1, can be reprogrammed by retroviral-

mediated transduction of Oct4, Klf4, Sox2, and c-Myc, to generate iPS cell lines that express

these endogenous factors. The reprogrammed cells formed EBs, expressed ectodermal and

mesodermal cell markers at the protein level and formed undifferentiated teratomas in vivo.

Although incomplete differentiation of the BTSC-derived iPS cell lines was observed in vitro

and in vivo, this could be attributed to several factors, such as a partially reprogrammed state or

the preexisting aberrant epigenetic or genetic program of the parental glioma stem cells.

Although the acquisition of endogenous Nanog and Oct4 expression may not fully define a true

pluripotent state, the data in this thesis suggest that at least one type of CSC can be remarkably

changed in its phenotypic behaviour and also in its lineage potential by transduction with a

defined set of transcription factors. Future studies will be required to understand the factors that

may contribute to a functional pluripotent state in reprogrammed CSCs and to assess the relative

genetic and non-genetic contributions to the neoplastic phenotype of glioma.

4.2 Why Reprogram Brain Tumour Stem Cells?

The rationale for reprogramming BTSCs is that it enables the study of the relative genetic and

non-genetic contributions to the neoplastic phenotype of brain tumours. Any genetic or non-

genetic change favourably regulating the survival of cells by conferring the ability to replicate

indefinitely, evade apoptosis, or prevent differentiation and senescence would promote the

abnormal clonal expansion of CSCs. If the genetic program of CSCs remains intact as these

cells differentiate to generate non-CSCs comprising the tumour bulk, epigenetics are evidently

involved in the differentiation of CSCs.

Differentiation therapy, therefore, is an attractive concept for cancer treatment if cancer is

considered as a stem cell hierarchy. It has been shown, for instance, that the treatment of human

GBM cells in vitro with BMP4 reduces proliferation, induces neural differentiation, and depletes

Page 85: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

70

the CD133+ CSC fraction, without affecting overall cell viability [190]. BMP4 treatment in vitro

or in vivo reduced the tumour-initiating capacity of GBM cells by inducing their differentiation

[190]. Similarly, the transcription factor-mediated reprogramming of GI cancer cells to the

pluripotent state caused the resulting iPS cells to be more highly sensitive to chemotherapy and

differentiation-inducing treatment [139]. These studies suggest that the epigenetic program of

cancer cells may influence the susceptibility of these cells to chemotherapeutic agents or

differentiation-inducing chemicals, which could be applied in the clinic as cancer treatments.

Reprogramming cancer cells may enable the reactivation of the expression of aberrantly

hypermethylated and silenced genes, including tumour suppressor genes, which would restore

normal growth control. It has been shown that the transcription factor-mediated reprogramming

of transformed human cells reactivates stable expression of the tumour suppressor gene p16INK4A

(CDKN2A) in the resulting iPS cells by erasing aberrant epigenetic marks, thereby restoring

normal cell cycle control [199]. Regions of DNA with de novo methylation were enriched for

PcG targets and were generally demethylated as a result of reprogramming [199]. In addition to

the p16INK4A

gene, 24 other gene promoters that are often hypermethylated in cancer were

demethylated during the reprogramming process, including the promoters of RASSF1, VHL,

FHIT, and GATA5 [199]. These findings suggest that aberrant CpG island hypermethylation of

tumour suppressor gene promoters can be erased during the reprogramming process in a manner

that parallels the removal of developmentally-related epigenetic marks. This study, however,

relied on the use of telomerase-immortalized human embryonic lung fibroblasts, which silence

p16INK4A

(CDKN2A) after extensive culturing [199]. These transformed cells fail to grow in soft

agar and are non-tumourigenic in immune-compromised mice, which is only suggestive of a

premalignant phenotype, and not a malignant phenotype [199]. Therefore, conducting a similar

study with primary human or mouse CSCs was of primary importance.

This thesis involved the reprogramming of BTSCs that, according to the Cancer Stem

Cell Hypothesis, are the cells responsible for the initiation and maintenance of tumour growth.

We developed a chemical-genetic mouse model of glioma that we believe is clinically relevant;

the CNS-specific deletion of p53 in the mouse model is representative of the human disease,

since p53 signaling is altered in 87% of high-grade glioma [164]. The chemical-genetic

approach to modeling the human disease in the mouse is ideal, since tumours arise spontaneously

as a result of mutagenesis, and not as a result of targeting additional oncogenes or tumour

Page 86: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

71

suppressors. By reprogramming mouse glioma BTSCs, the cells directly involved in the

initiation and maintenance of tumour growth can be studied.

4.3 Mouse Glioma Stem Cells are Amenable to Reprogramming

Mouse BTSCs derived from the chemical-genetic mouse model of glioma have a NSC-related

gene expression program, expressing Sox2, c-Myc, and Klf4 at similar levels as NSCs. Mouse

glioma stem cells, however, fail to express a pluripotency-associated program involving Nanog,

Oct4, and Rex1. This was initially detected by end-point RT-PCR and confirmed with

quantitative RT-PCR analysis. These results suggest that the self-renewal of mouse glioma stem

cells is regulated by a NSC-related program, likely driven by c-Myc, and not a pluripotent ES

cell-related signature. The similarities between glioma stem cells and normal NSCs, such as the

expression of the reprogramming factors Sox2, c-Myc, and Klf4 was initially hypothesized to

facilitate the generation of iPS cells from BTSCs with few exogenous factors, since adult mouse

NSCs had been reprogrammed with Oct4 and Klf4 or Oct4 alone [86,89]. However, technical

difficulties arose, such as the ability of the mouse glioma stem cells to proliferate extensively

during prolonged culture in serum-containing medium and their high intrinsic resistance to

selection agents such as puromycin and hygromycin. The preliminary piggyBac-mediated

reprogramming attempts with one or two factors, therefore, were unsuccessful, primarily due to

overgrowth of non-reprogrammed glioma stem cells. The GNS3 cell line, which grows more

slowly than GNS1 or GNS2 in culture conditions containing serum, was selected for subsequent

reprogramming experiments. Retroviral-mediated reprogramming of GNS3 with four exogenous

factors yielded more favourable results, since the inclusion of exogenous c-Myc generated iPS

cell-like colonies that could be isolated more readily from the induction plates. The appearance

of colonies with morphology characteristic of iPS cells suggested that the retroviral-mediated

reprogramming of glioma stem cells, p53+/+

and p53-/-

NSCs, and CD1 MEFs with four factors

was successful.

The molecular characterization of the resulting iPS cell lines revealed the expression of

core pluripotency factors, including endogenous expression of Nanog, Oct4, and Rex1. Fewer

iPS cell lines derived from BTSCs activated the expression of all three of the pluripotency

factors; in fact, endogenous Oct4 expression was only detected for one of the six GNS-iPS cell

lines. Nanog expression was considered the most important marker of pluripotency, since it is

Page 87: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

72

essential for the transition to the pluripotent state and is activated late in the process of

reprogramming mouse somatic cells [29,30]. The results were consistent with the finding that

p53 deficiency promotes reprogramming, since a higher proportion of p53-deficient NSC-

derived iPS cell lines expressed Nanog and generally at higher levels than the other iPS cell

lines. Although the glioma stem cells were also p53-deficient, Nanog expression was not

enhanced in BTSC-derived iPS cell lines. Treatment with the 2i cocktail of chemicals was

attempted, which consists of 1 µM PD0325901 and 3 µM CHIR99021 to inhibit MAPK

signaling and GSK3β, respectively, to promote complete reprogramming of mouse somatic cells

[195]. Treatment with 2i promoted Nanog expression in CD1 MEF-derived iPS cell lines, as

detected by end-point RT-PCR, and in a glioma stem cell-derived iPS cell line, as detected by

quantitative RT-PCR. The fold change in Nanog expression was only statistically significant if

2i treatment was carried out with twice the usual concentration of these inhibitors, which may

reflect the intrinsic drug resistance of CSCs. Relatively low levels of Nanog and Oct4 were

detected in the reprogrammed cell lines at the protein level by immunocytochemistry, whereas

Nestin (a marker of NSCs) and GFAP (a marker of astrocytic cells) were not detected. The non-

reprogrammed NSCs and glioma stem cell lines express high levels of GFAP in serum-

containing medium, whereas the reprogrammed cell lines significantly downregulated GFAP

expression, even in ES cell medium containing serum. Taken together, these results show that

the expression of endogenous core pluripotency genes (Nanog, Oct4, and Rex1) was activated in

the reprogrammed cell lines, and the expression of NSC and differentiated astrocytic cell

markers (Nestin and GFAP) was reduced. This may reflect the removal of developmentally-

regulated epigenetic marks and erasure of the epigenetic memory of the cell of origin.

4.4 Reprogrammed Cell Lines Show Varying Degrees of Retroviral Silencing

Different levels of transgene expression of exogenous Oct4, Klf4, Sox2, and c-Myc were detected

by quantitative RT-PCR in the iPS cell lines derived from BTSCs, p53+/+

NSCs, and p53-/-

NSCs.

Retroviral silencing is a property of pluripotent stem cells, including iPS cells, and has been

proposed to indicate complete reprogramming of somatic cells [198]. Exogenous Oct4

expression was silenced in each of the iPS cell lines derived from p53-deficient cells (BTSCs

and NSCs), which may reflect the enhanced ability to generate iPS cells from p53-deficient cells.

The glioma stem cell-derived iPS cell lines showed some retroviral silencing, whereas the p53-/-

Page 88: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

73

NSC-derived iPS cell lines failed to silence the expression of retroviruses encoding Klf4, Sox2,

and c-Myc. Both of the p53+/+

NSC-derived iPS cell lines failed to silence the expression of all

of the exogenous reprogramming factors, although four-factor iPS cell lines derived from wild

type NSCs have been shown to silence Oct4, Klf4, Sox2 and c-Myc transgenes [86]. The results

suggest that it was possible to derive iPS cell lines from mouse glioma stem cells, p53+/+

NSCs,

and p53-/-

NSCs, although the varying extent of retroviral silencing in each cell line suggests that

different degrees of reprogramming may have been achieved.

4.5 Analysis of the Differentiation Capacity of iPS Cell Lines

The initial indication that the reprogrammed cells had acquired new differentiation capabilities in

vitro was the results of the EB assays. Only the reprogrammed cell lines formed spherical EB-

like structures similar to those formed from J1 mouse ES cells in suspension culture in serum-

containing medium without LIF. The non-reprogrammed GNS3, p53+/+

NS, and p53-/-

NS cell

lines failed to form EBs and either adhered to the uncoated plastic dishes or died when

transferred from standard NSC medium or serum-containing medium to EB culture conditions.

The expression of various germ layer markers was assessed at the transcript level by RT-

PCR and at the protein level by immunostaining. At the transcript level, Sox17 (a marker of

endodermal cells) was detected at relatively high levels in EBs formed from one of the two

BTSC-derived iPS cell lines, as well as both of the p53+/+

NSC-derived iPS cell lines, and at low

levels in the p53-/-

NSC-derived iPS cell lines. The expression of this marker was detected at

much lower levels in the undifferentiated controls. The expression of ectodermal cell markers

(Nestin and Sox1) was detected in all of the cell lines at various time points in EB differentiation.

Brachyury (a marker of mesodermal cells) and GATA4 (a marker of endodermal cells) were only

detected in EBs formed from J1 mouse ES cells, whereas expression of the mesodermal cell

marker Flk1 was detected in EBs formed from glioma stem cells, p53-/-

NSCs, and p53+/+

NSCs.

Brachyury expression was only detected in EBs at day 4 of differentiation, but not at day 8,

which is consistent with the established kinetics of expression of this marker, since Brachyury

expression peaks at day 3 of differentiation of mouse ES cells but is undetectable by RT-PCR by

day 5 [200]. One of the glioma stem cell-derived iPS cell lines expressed markers of mesoderm

(Flk1), endoderm (Sox17) and ectoderm (Nestin and Sox1) at the transcript level after 8 days in

differentiation conditions, suggesting that a pluripotent phenotype was achieved. Also, EBs

Page 89: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

74

formed from one of the p53+/+

NSC-derived iPS cell lines expressed these four germ layer

markers, similar to EBs formed from ES cells (which also expressed Brachyury and GATA4).

Immunostaining revealed that the iPS cell-derived EBs expressed α-Smooth Muscle Actin (a

marker of mesodermal cells) and Nestin and βIII tubulin (markers of ectodermal cells) in

adherent conditions. The expression of GATA4 (a pan-endodermal cell marker) was only

detected in EBs formed from differentiated mouse ES cells, although the assessment of the

expression of additional endodermal cell markers at the protein level may be required. The

failure of the EBs to express GATA4 transcript or protein may be indicative of incomplete

endodermal differentiation. Nanog regulates endodermal differentiation by preventing

extraembryonic endodermal differentiation; Nanog-deficient mouse ES cells upregulate GATA4

and GATA6 expression and differentiate into extraembryonic endodermal cells [23]. Therefore,

achieving low endogenous levels of Nanog expression in the reprogrammed cell lines would

predict increased differentiation into GATA4+ cells, which was not observed. The minimal

expression of germ layer markers by p53-/-

NSC-derived iPS cell lines at the transcript level

suggests that a blockage in differentiation occurred.

Nevertheless, these results call into question the fidelity of germ layer markers in

assessing pluripotency, since the expression of both mesoderm markers (Brachyury and Flk1) or

both endoderm markers (Sox17 and GATA4) did not always occur in the same samples.

Furthermore, the detection of differentiated cell markers occurred at the transcript level in ES

cell conditions in the presence of LIF, which promotes the undifferentiated self-renewal of

mouse ES cells [19,20]. The detection of transcripts encoding differentiated cell markers in

presumably non-differentiating conditions can be attributed to the high sensitivity of end-point

RT-PCR amplifying basal levels of expression in undifferentiated cells or rare transcripts present

due to the spontaneous differentiation of a minority of cells. Even though Nestin expression was

detected at the transcript level in the majority of the iPS cell lines and J1 cells in ES cell

conditions, this NSC marker was not detected at the protein level by immunocytochemistry. It

would be beneficial to carry out epigenetic analyses, such as bisulphite sequencing at the

promoters of Nestin and GFAP to determine whether epigenetic memory is present and whether

the reprogrammed cells have retained memory of the cell type of origin.

The differentiation capacity of the reprogrammed cell lines was also assessed in vivo by

subcutaneous and intracranial injections with subsequent histological and immunohistochemical

Page 90: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

75

analysis. Highly undifferentiated, atypical tumours were generated from the injection of each of

the p53-/-

NSC-derived and glioma stem cell-derived iPS cell lines, as determined by histological

analysis. However, the p53-/-

NS-iPS2 cell line formed tumours that stained positive for markers

of mesoderm (α-Smooth Muscle Actin), endoderm (GATA4), and ectoderm (Nestin), whereas

p53-/-

NS-iPS1 and both of the GNS-iPS cell lines failed to generate tumours with GATA4+ cells.

One of the p53+/+

NSC-derived iPS cell lines also generated undifferentiated tumours, whereas

the other produced highly differentiated tumours with structures representative of the three germ

layers, similar to teratomas formed from J1 mouse ES cells. It should be noted that blood vessels

express αSMA, although vascular αSMA+ cells were readily distinguished from αSMA

+ tumour

cells based on morphology and the presence of autofluorescent erythrocytes. Furthermore, this

mesodermal cell marker is expressed in mesenchymal cells such as smooth muscle, which may

originate from neural crest cells [33]. The expression of additional mesodermal markers of non-

mesenchymal cells and non-neural ectodermal markers should be assessed to establish a

definitive differentiation profile of the reprogrammed cells.

Furthermore, the glioma stem cell-derived iPS cell lines formed subcutaneous and

intracranial tumours that failed to show GFAP+ cells, which was in stark contrast to the tumours

formed from the injection of the non-reprogrammed glioma stem cell line or J1 mouse ES cells.

The non-reprogrammed GNS3 cell line produced tumours with high GFAP expression, which is

consistent with previous results from our laboratory suggesting that this cell line is biased toward

a glial phenotype in vivo [180]. The reprogrammed glioma stem cell lines even failed to

generate GFAP+ astrocytic cells in the forebrain niche. These results suggest that the

reprogrammed glioma stem cell lines may have a blockage in astrocytic differentiation in vivo,

which may represent a suppression of the endogenous lineage commitment of the parental

glioma stem cells, although additional in vivo experiments are required to strengthen this finding.

The preliminary directed re-differentiation of the iPS cell lines generated cells with

typical NSC morphology that proliferated in adherent conditions in serum-free medium with

EGF and bFGF. An increase in Nestin and slight decrease in Oct4 staining was observed during

the directed re-differentiation procedure, suggesting that lineage commitment was in progress.

Future experiments will assess the expression of neural stem cell markers and pluripotency

factors, and will evaluate the multipotent differentiation capacity of the putative NSC lines.

Page 91: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

76

4.6 Interplay between Retroviral Silencing, p53 Status, and In Vivo Differentiation Potential

Retroviral silencing of exogenous expression of Oct4, Klf4, Sox2, and c-Myc is generally thought

to be required for the differentiation of iPS cells in vivo [30,198]. The constitutive ectopic

expression of the four reprogramming factors may prevent the differentiation of iPS cells in

teratomas by disrupting the endogenous signaling pathways involved in regulating differentiation

[30]. For instance, Sox2 suppresses endodermal and mesodermal differentiation, but promotes

neural ectodermal differentiation [28]. Oct4 has the converse role in promoting endodermal and

mesodermal differentiation while suppressing neural ectodermal differentiation [28]. The role of

c-Myc in the maintenance of pluripotency is through the repression of primitive endoderm

differentiation [201]. Therefore, it is likely that the sustained overexpression of retroviral

vectors encoding the reprogramming factors may influence the differentiation capacity of the iPS

cell lines.

A surprising result was the extensive differentiation of the p53+/+

NS-iPS1 cell line,

which expressed exogenous reprogramming factors, following subcutaneous or intracranial

injection in vivo. Large regions of cells stained positive for each of the germ layer markers

assessed. This is inconsistent with the literature, which suggests that the downregulation of

transgene expression of Oct4, Klf4, Sox2, and c-Myc is required for the differentiation of iPS

cells in vivo [30]. However, the overexpression of Oct4 has been shown to promote primitive

endodermal and mesodermal differentiation [24]. This may account for the increased

endodermal and mesodermal differentiation observed when the p53+/+

NS-iPS1 cell line was

injected in vivo, since this cell line failed to silence exogenous Oct4 expression. However, the

role of Sox2 in suppressing endodermal and mesodermal differentiation was not corroborated by

the results of this study, since high exogenous Sox2 expression in p53+/+

NS-iPS1 did not prevent

the differentiation of endodermal and mesodermal cells in vivo [28].

The tumours generated from the subcutaneous or intracranial injection of the

reprogrammed cell lines showed varying degrees of differentiation, which seemed to be

regulated largely by p53 status. In contrast to the differentiated tumours generated by the p53+/+

NS-iPS1 cell line, the p53-deficient GNS-iPS cell lines, which had silenced exogenous

expression of the four reprogramming factors, generated highly undifferentiated tumours. The

Page 92: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

77

growths resembled primitive malignant tumours, showing high nuclear to cytoplasmic ratio and

failing to show any structures representative of the three germ layers, based on histological

analysis. This blockage in differentiation was independent of retroviral silencing and was

therefore attributed to p53 deficiency, genetic lesions or epigenetic aberrations suppressing

differentiation. Similar undifferentiated tumours were obtained from the injection of p53-

deficient NSC-derived iPS cell lines, which had silenced exogenous Oct4 expression, but

continued to express Klf4, Sox2, and c-Myc transgenes. Although some endodermal

differentiation was detected in vivo for the p53-deficient NSC-derived iPS cell lines based on the

expression of the endodermal cell marker GATA4, these tumours did not show any structures

representative of the three germ layers upon histological analysis. This may be indicative of

impaired differentiation capacity, likely mediated by p53 status or ectopic expression of other

reprogramming factors.

It has been shown that p53 deficiency, combined with transduction of c-Myc, generates

growths consisting primarily of undifferentiated tissues, with only small areas of differentiated

cells, thus resembling tumours [72]. Nevertheless, p53-deficient iPS cells generated from four

factor inductions are able to form chimeric mice, although incomplete silencing of exogenous

expression of retroviral c-Myc may account for the observation that chimeras were not viable

past seven weeks [72]. The overall expression levels of Klf4, Sox2, and c-Myc are higher in p53-

deficient iPS cells derived from four-factor inductions than in p53-deficient iPS cells derived

from three-factor inductions (omitting c-Myc) or in p53-heterozygous iPS cells [72]. They

suggest that exogenous c-Myc expression may prevent retroviral silencing in iPS cells with a

p53-deficient background, although this hypothesis and the mechanism by which c-Myc acts to

suppress retroviral silencing remain to be confirmed conclusively.

4.7 Reprogramming Mouse Versus Human Cells

The mouse system offers the advantage in reprogramming experiments, since the attainment of

complete reprogramming can be verified by creating chimeric mice and assessing germline

transmission. Human GBM stem cell lines grown in similar adherent culture conditions could

also be reprogrammed in parallel with patient-matched fibroblasts to compare the gene

expression profiles of the resulting iPS cell lines. This would help to identify any cancer-specific

genetic or epigenetic changes that may be involved in the neoplastic phenotype or may prevent

Page 93: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

78

reprogramming to pluripotency. Furthermore, although primary tumour samples can be obtained

from human brain cancer, the normal NSCs that may give rise to these tumours are inaccessible.

Being able to model genetic changes in NSCs differentiated from BTSC-derived iPS cells using

this reprogramming and re-differentiation strategy may be a means to understand the influence of

cancer-associated genetic changes on NSC behaviour. As well, reprogramming normal patient

fibroblasts into iPS cells and re-differentiating to produce NSCs will allow us to interrogate

patient specific cancer genetic changes on the behaviour of NSCs, which are a potential cell of

origin for human brain tumours. For example, the patient’s own oncogenic changes could be

introduced into their reprogrammed NSCs to see which lesion or combination of mutations

contributes to human NSC transformation.

In human iPS experiments, however, teratoma assays are the best indication of

pluripotency, but these experiments are not standardized and fail to indicate the degree of

reprogramming that was achieved, since even partially reprogrammed iPS cells generate

teratomas [192,202]. As a result, an open-access bioinformatic assay was developed to assess

the pluripotency of human iPS cell lines based on the comparison of their genome-wide

expression profile to that of approximately 450 human ES cells, iPS cell lines, stem cell lines,

differentiated cells, and developing and adult tissues [203]. This resource will provide an

extremely valuable tool to assess the quality and pluripotency of human iPS cell lines in the

future. As the only in vivo measure of pluripotency in human iPS cells is the teratoma assay,

there remains controversy over the definition of pluripotency in reprogrammed cells.

4.8 Defining the Reprogrammed State

The reprogrammed state of iPS cells may be especially difficult to define in the context of cancer

cells as the reprogramming target, since cancer cells are associated with both genetic and

epigenetic aberrations. The induction of Nanog expression may not confer functional

pluripotency in cancer cell-derived iPS cells, due to the potential for genetic lesions or abnormal

epigenetic marks in key differentiation-regulating genes in cancer cells. Furthermore, copy

number variations, mutations, or amplifications of lineage determining genes in cancer cells

could result in cell death of a particular lineage during in vitro differentiation of CSC-derived

iPS cell lines. In addition, in the chemical-genetic mouse model of glioma, in utero treatment

with the carcinogen ENU resulted in genetic mutations in the BTSCs, some of which may affect

Page 94: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

79

the ability of these cells to be reprogrammed to pluripotency, particularly if epigenetic regulators

are aberrantly expressed. If genes involved in chromatin remodeling and gene expression

regulation, such as those encoding PcG proteins, are mutated or have altered expression levels in

cancer cells, it would be reasonable to hypothesize that the ability of these cells to be

reprogrammed to the pluripotent state may be altered or that the differentiation profile of cancer

cell-derived iPS cells may be affected. For instance, the expression of the PcG protein and

oncogene, Bmi1, is essential for the development of mouse glioma and is highly enriched in the

CD133+ BTSC population in human glioma, which makes it a potential candidate to regulate

differentiation in glioma stem cell-derived iPS cells [112,169]. Furthermore, the INK4A/ARF

locus is silenced in iPS and ES cells, but Bmi1 has an established role in regulating cellular

differentiation in an INK4A/ARF-independent manner in both mouse and human glioma, which

further supports its potential role in regulating differentiation in iPS cells. A role for Bmi1 in the

induction of pluripotency has also been established; it was recently shown that MEFs could be

reprogrammed by retroviral transduction of Bmi1 to generate NS-like cells, which were then

reprogrammed with Oct4 alone to generate germline-competent iPS cells [204]. Chemical

activation of sonic hedgehog signaling in MEFs enhanced the expression of Bmi1, Sox2, N-Myc,

and Klf4, and exogenous Oct4 was sufficient to reprogram these cells to the iPS cell state [204].

It is believed that Bmi1 has two main roles in promoting the generation of iPS cells, through the

suppression of the INK4A/ARF locus (which encodes p16INK4a

and p19ARF

) and by enhancing the

expression of Sox2 and N-Myc [204]. The role of Bmi1 in regulating the differentiation of iPS

cells remains to be confirmed.

In addition, epigenetic memory of the cell type of origin and permanent genetic mutations

may complicate reprogramming studies involving cancer cells [63]. Epigenetic memory occurs

when epigenetic marks, such as DNA methylation, are retained in reprogrammed cells and

reflect the somatic cell type from which they were derived [63]. These residual epigenetic marks

confer different gene expression profiles and promote the differentiation of reprogrammed cells

along lineages associated with the somatic tissue of origin, while suppressing commitment to

other lineages of differentiation [63,64]. Epigenetic memory is generally associated with early-

passage human and mouse iPS cells and can be erased by prolonged culture, differentiation and

serial reprogramming, or as a result of treatment with HDAC inhibitors or DNMT inhibitors

[63,64]. Early- and late-passage (p4 versus p>10) mouse iPS cells have similar properties,

Page 95: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

80

including the promoter demethylation and expression of endogenous pluripotency genes, and

formation of teratomas and chimeras [64]. Although both classes of iPS cells are transgene-

independent, early-passage iPS cells harbour epigenetic memory and are therefore

transcriptionally distinguishable from late-passage iPS cells and have biased differentiation

toward the tissue type of origin [64]. By contrast, partially reprogrammed cells are dependent

upon the expression of exogenous pluripotency factors, fail to express endogenous pluripotency

factors, and form teratomas but not chimeras [64]. The differentiation experiments in this thesis

were carried out with late-passage iPS cell lines (p>10), but it may be beneficial to repeat

differentiation experiments after extended time in culture to ensure the complete erasure of

epigenetic memory. Furthermore, reprogramming-associated mutations involving genes

implicated in cancer have been identified in human iPS cells [80]. Similarly, human iPS cells

harbour de novo copy number variations and genomic deletions of tumour suppressor genes

[81,82]. These findings show that not only epigenetic modifications, but also permanent genetic

alterations are involved in reprogramming somatic cells to the pluripotent state, which may

complicate the study of genetics and epigenetics in CSC-derived iPS cells.

4.9 Core Pluripotency Network Versus c-Myc Network in Cancer

The putative ES cell signature attributed to the gene expression profiles of aggressive human

cancers remains controversial in the cancer field. Some groups have recognized an ES cell-like

gene expression program in aggressive human tumours, based on the overexpression of genes

targeted by Nanog, Oct4, and Sox2, with c-Myc identified as the activator of the ES cell program

[119,205]. However, it was not determined whether the putative ES cell signature exists in rare

CSCs or in bulk tumour cells, which makes it difficult to draw any conclusions relating to the

functional implications of this signature [205]. The proposed ES cell signature in epithelial

cancers was interpreted to represent the dedifferentiation of cancer cells to an ES cell-like state

[205]. However, a significant proportion of genes targeted by Nanog, Oct4, and Sox2 are also

regulated by c-Myc, which suggests that a pluripotency signature is in fact a c-Myc signature

[120]. A recent study attributed the similarities between the gene expression profiles of human

cancer and ES cells to a common transcriptional network regulated by c-Myc [120]. The

conflicting studies concurred, however, that PcG target genes were repressed in human cancers,

similar to ES cells, thereby confirming a role for PRC proteins in cancer initiation or progression

[119,120]. Nevertheless, the extensive overlap between the network regulated by c-Myc and the

Page 96: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

81

core pluripotency network governed by Nanog, Oct4, and Sox2 makes it extremely difficult to

identify a true ES cell signature in cancer.

The findings of this study support the concept that c-Myc, and not the core pluripotency

network of Nanog, Oct4, and Sox2, regulates gene expression in mouse glioma stem cells. The

gene expression profiles of human and mouse glioma stem cells were found to align closely with

those of human and mouse normal NSCs [180]. Two distinct gene expression patterns were

identified when mouse glioma stem cells and p53-/-

NSCs were compared to p53+/+

NSCs: a p53-

deficient signature and a glioma signature [180]. Interestingly, the p53-deficient signature

shared by glioma stem cells and p53-/-

NSCs was enriched for genes implicated in p53, apoptosis,

and MAPK signaling pathways, whereas the glioma-associated signature was represented by the

enrichment for transcripts involved in various human cancers, metabolic pathways, insulin and

mTOR signaling, and ubiquitin-mediated proteolysis, among others [180]. No enrichment for

pluripotency-related gene expression was detected. It is likely that enriching for CSCs would

effectively diminish the putative ES cell signature in cancer cells and further enhance the

prevalence of the c-Myc regulatory network.

Additional evidence supporting the importance of the c-Myc network in mouse glioma

stem cells is the finding that these cells express neural stem cell-related transcripts, such as Sox2,

c-Myc, and Klf4. Although these transcription factors have a role in reprogramming somatic

cells to the pluripotent state, they are also expressed in adult mouse neural stem cells and are,

therefore, not ES cell-specific transcription factors [86]. Sox2, which is considered to be part of

the core transcriptional network in ES cells (in addition to Nanog and Oct4), regulates the

maintenance of self-renewal and multipotent differentiation in NSCs in a manner that parallels

its role in ES cells, but without activating the core pluripotency network. Indeed, mouse glioma

stem cells fail to express the pluripotency factors, Nanog, Oct4, and Rex1, even though they

express relatively high levels of Sox2. This suggests that Sox2 expression does not activate the

core pluripotency network in mouse glioma stem cells. Furthermore, the cell surface marker

SSEA-1/CD15 and the enzyme alkaline phosphatase are expressed by mouse glioma stem cells

and ES cells, but these markers are by no means specific to pluripotent cells and do not reflect

any functional similarity between glioma stem cells and ES cells. In fact, although SSEA-1

enriches for clonogenic cells in vitro and tumour-initiating cells in vivo in mouse models of

glioma and medulloblastoma, the functional role of this marker in the context of cancer remains

Page 97: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

82

unclear [180,186,187]. Therefore, the results support the importance of a NSC-related c-Myc

network, but not a core pluripotency network involving Nanog and Oct4, in mouse glioma stem

cells.

4.10 Expression of Pluripotency Factors in Human Glioma

Due to the relatively low efficiency of generating iPS cells through transcription factor-mediated

reprogramming, it could be argued that a rare population of cells may be more amenable to

reprogramming, potentially due to the expression of pluripotency-associated factors. The

expression of core pluripotency factors, Nanog and Oct4, has been detected in primary human

GBM tumours in a small cohort of studies, although the function of these factors in GBM

pathogenesis remains elusive [132,133,189,206,207,208]. Levels of Oct4 transcript and protein

expression were found to correlate with glioma grade, with high-grade GBM expressing greater

amounts of Oct4 than low-grade astrocytomas [207]. It has been hypothesized, therefore, that

the augmented malignancy of GBM is a result of reactivated expression of stem cell genes, such

as Oct4, although this remains to be shown conclusively [133]. It has been shown that the

expression of neural stem cell markers (Nestin and Sox2) remains unaltered in human glioma

cells in vivo and in vitro, whereas pluripotency factor expression (Nanog and Oct4) occurs in

vivo, but is lost after approximately 20 passages in vitro [133]. The cause for the downregulated

expression of Nanog and Oct4 in human GBM cells in vitro is not clear. Our lab has detected

Oct4 protein in primary human GBM tumours [209], but not at the transcript level in mouse

glioma stem cells in vitro, as shown in this study by end-point RT-PCR and quantitative RT-

PCR. Therefore, it is unlikely that a subset of mouse glioma stem cells exists that is more

amenable to reprogramming due to the expression of core pluripotency factors.

The functional role of Nanog and Oct4 expression in human glioma has not been

established. Nanog was found to regulate proliferation of CD133+ cells sorted from primary

human GBM tumours and was essential for tumour initiation in vivo [208]. In an elegant

competition experiment involving lentiviral-mediated fluorescent labeling of primary GBM cells

combined with Nanog knockdown using short hairpin RNA, GFP-control or GFP-shNanog were

injected intracranially with RFP-control cells [208]. Without Nanog knockdown, the resulting

tumours were a combination of GFP+ and RFP

+ cells, whereas the injection of RFP-control and

GFP-shNanog cells resulted in tumours consisting entirely of RFP+ cells, with GFP-shNanog

Page 98: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

83

cells unable to engraft and form tumours [208]. These results suggest that Nanog expression is

required for the engraftment and tumour-initiating capability of human GBM cells in orthotopic

xenograft experiments.

The self-renewal of CD133+ cells sorted from primary human GBM tumours and

increased expression of human ES cell markers, including Nanog, Oct4, and c-Myc, were

promoted in hypoxic (2% oxygen) conditions [206,210]. The functional relevance of the

enhanced expression of Nanog, Oct4 and c-Myc in hypoxic conditions is not clear, since

upregulation of these ES cell markers were not only seen in CD133+ CSCs sorted from primary

human GBM tumours, but also in CD133- cells [210]. Hypoxia stabilizes hypoxia inducible

factor alpha (HIFα) subunits and allows the HIF complex to bind to hypoxia response elements

and activate the expression of target genes, including c-Myc and Oct4 [210]. It is likely that the

misexpression of ES cell-related transcription factors in glioma is a result of the hypoxic

activation of aberrant gene expression programs and has very little functional implication in

pluripotency; the CSCs responsible for tumour growth and maintenance do not form teratomas in

vivo containing tissues derived from the three germ layers. Although cells from primary human

GBM tumours may express detectable levels of Nanog and Oct4 transcript or protein, it is

unlikely that the levels of expression are similar to those occurring in pluripotent human ES

cells. It is probable that a balance of the transcription factors belonging to the core pluripotency

network (Nanog, Oct4, and Sox2) is required for functional pluripotency.

The expression of pluripotency factors, particularly Oct4, in human gliomas and other

types of cancer is complicated by the fact that several pseudogenes and splice variants exist.

This has been confirmed for the human Oct4 gene, but not the mouse Oct4 gene. The expression

of three distinct Oct4 pseudogenes has been detected at the transcript and protein levels in

primary samples and cell lines derived from human glioma and breast cancer [211,212].

Conversely, Oct4 was not detected in these tumour types, among others, by

immunohistochemistry of tissue arrays [129,130]. Although the Oct4 pseudogenes have high

sequence homology to the human Oct4 gene, they encode protein products that do not have the

same function in promoting pluripotency and self-renewal of cells [211]. Furthermore, through

alternative splicing, three isoforms of the Oct4 protein can be generated: Oct4A, Oct4B, and

Oct4B1 [213,214]. The Oct4A isoform is the classic Oct4 protein involved in the maintenance

of pluripotent self-renewal in ES and EC cells, whereas the Oct4B isoform is expressed in non-

Page 99: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

84

pluripotent somatic cells and has no established function in sustaining the self-renewal of ES

cells [213,214]. Oct4A and Oct4B possess a DNA binding domain, whereas Oct4B1 lacks a

portion of this domain and has no established function, although it is expressed in ES and EC

cells and is downregulated during differentiation [214]. The presence of multiple pseudogenes

and splice variants of Oct4 raise the possibility of false-positive results at the transcript and

protein levels if primers or antibodies are not selected carefully to identify the classic Oct4A

variant [215]. In fact, studies in which Oct4 expression was detected at the transcript level in

human cancer cells using primers to amplify sequences common to both Oct4A and Oct4B, such

as one study claiming that human embryonic genes are re-expressed in cancer, may simply be

detecting Oct4B or a pseudogene of Oct4 [215,216]. Similarly, Nanog encodes ten processed

pseudogenes in the human, three of which can be expressed, and only two pseudogenes in the

mouse in addition to the bona fide Nanog protein product [217]. Therefore, the expression of

pseudogenes and splice variants, particularly for the human Oct4 gene, complicates the study of

this pluripotency factor in human somatic tumours.

4.11 Future Directions

The glioma stem cell-derived iPS and appropriate control iPS cell lines generated from this

project will be a valuable resource in carrying out important experiments in the future. A

potential caveat is that additional iPS cell lines derived from other glioma stem cell lines and

different tumour types may be required to strengthen the findings of this study. Ideally, the

glioma stem cell-derived and control iPS cell lines should show equivalent degrees of

reprogramming. Furthermore, non-integrating methods of generating iPS cells should be

employed to minimize the genetic effects of reprogramming to pluripotency. It may be

beneficial to repeat the reprogramming experiments and replace exogenous c-Myc with L-Myc,

which has been shown to promote more efficient reprogramming with less potent neoplastic

transformation than c-Myc [84]. It remains to be shown whether different primary brain tumour

samples can be reprogrammed more fully or whether genetic lesions block complete

reprogramming.

The molecular and functional characterization of the iPS cell-derived NSC lines obtained

in this study will be carried out to assess the expression of neural stem cell markers and to

determine whether the cells are multipotent and can generate neurons, astrocytes, and

Page 100: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

85

oligodendrocytes upon growth factor withdrawal in vitro. Global DNA methylation analysis,

bisulphite sequencing of candidate promoter regions of pluripotency-associated genes (Nanog

and Oct4) and NSC-associated genes (Nestin and GFAP), and chromatin immunoprecipitation

coupled with high-throughput sequencing to detect histone modifications can be carried out to

compare the epigenetic patterns of the non-reprogrammed glioma stem cells, iPS cell lines, and

iPS cell-derived NSC lines. An important experiment to be performed involves the genomic

DNA sequencing of the iPS cell lines and the original cell lines from which they were derived, in

order to identify potential reprogramming-associated mutations [80]. This will not only provide

an assessment of the genetic mutations that may have occurred as a result of retroviral

integration, but will also identify mutations in the non-reprogrammed glioma stem cells, which

were induced by ENU treatment in the chemical-genetic mouse model of glioma. In addition,

microarray analysis of the non-reprogrammed glioma stem cells, iPS cells, and iPS-derived

NSCs must be carried out to compare the gene expression profiles of the various cell types. In

combination with the sequencing data, the comparison of the gene expression profiles of the non-

reprogrammed glioma stem cells and iPS-derived NSCs, in particular, would help to elucidate

the epigenetic changes occurring during reprogramming and subsequent re-differentiation of

cancer cells. Intracranial injections will be carried out for the iPS cell-derived NSCs and

parental glioma stem cell lines to study tumourigenicity. These experiments will allow us to

study the relative contributions of genetic and non-genetic factors to the cancer phenotype and

will aid in determining whether reprogramming mouse glioma stem cells alters their tumour-

initiating ability.

Page 101: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

86

Appendices

5 Appendix 1. Supplemental Methods

5.1 Primer Sequences

The following primer sequences were used to amplify transcripts for RT-PCR and quantitative

RT-PCR (qRT-PCR) applications.

Supplemental Table 1. Primer sequences used in this study.

Target Primer Sequence Annealing

Temp (°C)

Application

Nanog

for: ACACTGACATGAGTGTGGGTCTTCC

63

RT-PCR

rev: GCAGGTCTTCAGAGGAAGGGCGAG

Oct4 for: CACGAGTGGAAAGCAACTCA

rev: TGGGAAAGGTGTCCCTGTAG

60 RT-PCR

Endogenous

Oct4

for: TCTTTCCACCAGGCCCCCGGCTC

rev: TGCGGGCGGACATGGGGAGATCC

60 RT-PCR

Klf4 for: TACCCCTACACTGAGTCCCG

rev: GTGTGGGTGGCTGTTCTTTT

53 RT-PCR

Sox2 for: ATACAAGGGAATTGGGAGGG

rev: AAACAAGACCACGAAAACGG

58 RT-PCR

c-Myc for: GGACTGTATGTGGAGCGGTT

rev: TCGTCTGCTTGAATGGACAG

58 RT-PCR

Rex1 for: GGAAGAAATGCTGAAGGTGGAGAC 55 RT-PCR

rev: AGTCCCCATCCCCTTCAATAGC

Flk1 for: TAGGTGCCTCCCCATACCCTGG

rev: TGGCCGGCTCTTTCGCTTACTG

60 RT-PCR

Brachyury for: TAATGGAGGAACCGGGGGACTGC

rev: TGTCCGCATAGGTTGGAGAGCTGT

60 RT-PCR

Sox17 for: TATGGTGTGGGCCAAAGACGA

rev: AACGCCTTCCAAGACTTGCCT

60 RT-PCR

GATA4 for: CTGGAAGACACCCCAATCTC

rev: CACAGGCATTGCACAGGTAG

60 RT-PCR

Nestin for: AACTCTCGCTTGCAGACACCTG

rev: AGGTGCTGGTCCTCTGGTATCC

60 RT-PCR

Sox1 for: TTCCCCAGGACTCCGAGGCG

rev: GCTGTGTGCCTCCTCTGCGG

60 RT-PCR

Page 102: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

87

GAPDH for: AACTTTGGCATTGTGGAAGG

rev: ACACATTGGGGGTAGGAACA

60

58-60

RT-PCR

qRT-PCR

Nanog

for: TTCTTGCTTACAAGGGTCTGC

rev: AGAGGAAGGGCGAGGAGA

60 qRT-PCR

Oct4 for: ACATCGCCAATCAGCTTGG

rev: AGAACCATACTCGAACCACATCC

60 qRT-PCR

Rex1 for: TCTTCTCTCAATAGAGTGAGTGTGC

rev: GCTTTCTTCTGTGTGCAGGA

60 qRT-PCR

Klf4 for: GCACACCTGCGAACTCACAC 60 qRT-PCR

rev: CCGTCCCAGTCACAGTGGTAA

Klf2 for: CTCAGCGAGCCTATCTTGCC

rev: CAGACCGTCCAATCCCATGG

60 qRT-PCR

Klf5 for: ATTCGCCAACTCTCCCACCT

rev: TCGCCCGTATGAGTCCTCAG

60 qRT-PCR

Sox2 for: ACAGATGCAACCGATGCACC 60 qRT-PCR

rev: TGGAGTTGTACTGCAGGGCG

c-Myc for: CCACCAGCAGCGACTCTGA 60 qRT-PCR

rev: TGCCTCTTCTCCACAGACACC

L-Myc for: GGACGGCACTCCTAGTCTGG

rev: GGTGACTGGCTTTCGGATGT

60 qRT-PCR

Exogenous

Oct4

for: TCTCCCATGCATTCAAACTG

rev: CTTTTATTTTATCGTCGACC

58 q-RT-PCR

Exogenous

Klf4

for: CCACCTTGCCTTACACATGA

rev: CTTTTATTTTATCGTCGACC

58 qRT-PCR

Exogenous

Sox2

for: CTCCGGGACATGATCAGC

rev: CTTTTATTTTATCGTCGACC

58 qRT-PCR

Exogenous

cMyc

for: TGAGGAAACGACGAGAACA

rev: CCCTTTTTCTGGAGACTAAATAAA

58 qRT-PCR

Page 103: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

88

6 Appendix 2. Supplemental Figures

6.1 Quantitative RT-PCR Results of Pluripotency Factor Expression

Figure S1. Glioma stem cell lines express L-Myc and neural stem cell-associated factors,

but do not express Nanog or Oct4.

Quantitative RT-PCR results showing the expression levels of reprogramming-associated

factors in glioma stem cell lines (GNS1, GNS2, and GNS3), p53-/-

NS, and p53+/+

NS in stem

cell conditions (serum-free medium with EGF and bFGF) and differentiation conditions

(with 10% serum). Fold expression is shown relative to J1 (ES) cells. Overexpression of c-

Myc was frequently detected in glioma stem cells and normal NSCs. The data represents

the average from triplicate readings and was generated while supervising Stephanie

Dobson, a rotation student in the lab.

Page 104: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

89

6.2 Alkaline Phosphatase Expression in Mouse Glioma Stem Cell Lines

A B

Figure S2. Glioma stem cells, but not normal neural stem cells, express alkaline

phosphatase in stem cell conditions.

(A) The three mouse glioma stem cell lines express alkaline phosphatase (AP) when grown

in neural stem cell conditions (serum-free medium with EGF and bFGF), as detected by

light microscopy (middle column) or fluorescence microscopy (right column). (B) Normal

neural stem cells grown in stem cell conditions fail to express AP, regardless of p53 status.

J1 mouse ES cells and MEFs were used as a positive and negative control, respectively.

Page 105: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

90

A B

Figure S3. Glioma stem cells and normal neural stem cells fail to express alkaline

phosphatase in differentiation conditions.

(A) The three mouse glioma stem cell lines do not express detectable levels of alkaline

phosphatase (AP) when grown in differentiation conditions (medium with 10% serum), as

detected by light microscopy (middle column) or fluorescence microscopy (right column).

(B) Normal neural stem cells grown in differentiation conditions fail to express AP,

regardless of p53 status. J1 mouse ES cells and MEFs were used as a positive and negative

control, respectively.

Page 106: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

91

6.3 Nanog Expression in MEF-Derived iPS Cell Lines

Figure S4. MEF-derived iPS cell lines express Nanog and 2i treatment enhances Nanog

expression.

Nanog expression was detected by RT-PCR in 6 iPS cell lines derived from the four-factor

infection of CD1 MEFs. Six additional MEF-derived iPS cell lines were treated with

DMSO (D) or 2i. Treatment with 2i was found to enhance Nanog expression in some of the

MEF-derived iPS cell lines, as detected by end-point RT-PCR. J1 ES cells and three-factor

infected MEF-derived iPS cells (3F iPS, obtained from the Ellis Lab) were used as positive

controls, with MEFs as a negative control.

Page 107: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

92

References

1. Fuchs E (2009) The tortoise and the hair: slow-cycling cells in the stem cell race. Cell 137:

811-819.

2. Altman J (1963) Autoradiographic investigation of cell proliferation in the brains of rats and

cats. Anat Rec 145: 573-591.

3. Reynolds BA, Weiss S (1992) Generation of neurons and astrocytes from isolated cells of the

adult mammalian central nervous system. Science 255: 1707-1710.

4. Morshead CM, Reynolds BA, Craig CG, McBurney MW, Staines WA, et al. (1994) Neural

stem cells in the adult mammalian forebrain: a relatively quiescent subpopulation of

subependymal cells. Neuron 13: 1071-1082.

5. Weiss S, Reynolds BA, Vescovi AL, Morshead C, Craig CG, et al. (1996) Is there a neural

stem cell in the mammalian forebrain? Trends Neurosci 19: 387-393.

6. Doetsch F, Caille I, Lim DA, Garcia-Verdugo JM, Alvarez-Buylla A (1999) Subventricular

zone astrocytes are neural stem cells in the adult mammalian brain. Cell 97: 703-716.

7. Eriksson PS, Perfilieva E, Bjork-Eriksson T, Alborn AM, Nordborg C, et al. (1998)

Neurogenesis in the adult human hippocampus. Nat Med 4: 1313-1317.

8. Scadden DT (2006) The stem-cell niche as an entity of action. Nature 441: 1075-1079.

9. Doetsch F, Petreanu L, Caille I, Garcia-Verdugo JM, Alvarez-Buylla A (2002) EGF converts

transit-amplifying neurogenic precursors in the adult brain into multipotent stem cells.

Neuron 36: 1021-1034.

10. Kuhn HG, Winkler J, Kempermann G, Thal LJ, Gage FH (1997) Epidermal growth factor

and fibroblast growth factor-2 have different effects on neural progenitors in the adult rat

brain. J Neurosci 17: 5820-5829.

11. Conti L, Pollard SM, Gorba T, Reitano E, Toselli M, et al. (2005) Niche-independent

symmetrical self-renewal of a mammalian tissue stem cell. PLoS Biol 3: e283.

12. Uchida N, Buck DW, He D, Reitsma MJ, Masek M, et al. (2000) Direct isolation of human

central nervous system stem cells. Proc Natl Acad Sci U S A 97: 14720-14725.

13. Capela A, Temple S (2002) LeX/ssea-1 is expressed by adult mouse CNS stem cells,

identifying them as nonependymal. Neuron 35: 865-875.

14. Evans MJ, Kaufman MH (1981) Establishment in culture of pluripotential cells from mouse

embryos. Nature 292: 154-156.

15. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, et al. (1998)

Embryonic stem cell lines derived from human blastocysts. Science 282: 1145-1147.

Page 108: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

93

16. Ying QL, Nichols J, Chambers I, Smith A (2003) BMP induction of Id proteins suppresses

differentiation and sustains embryonic stem cell self-renewal in collaboration with

STAT3. Cell 115: 281-292.

17. Williams RL, Hilton DJ, Pease S, Willson TA, Stewart CL, et al. (1988) Myeloid leukaemia

inhibitory factor maintains the developmental potential of embryonic stem cells. Nature

336: 684-687.

18. Smith AG, Heath JK, Donaldson DD, Wong GG, Moreau J, et al. (1988) Inhibition of

pluripotential embryonic stem cell differentiation by purified polypeptides. Nature 336:

688-690.

19. Niwa H, Ogawa K, Shimosato D, Adachi K (2009) A parallel circuit of LIF signalling

pathways maintains pluripotency of mouse ES cells. Nature 460: 118-122.

20. Hall J, Guo G, Wray J, Eyres I, Nichols J, et al. (2009) Oct4 and LIF/Stat3 additively induce

Kruppel factors to sustain embryonic stem cell self-renewal. Cell stem cell 5: 597-609.

21. Zhang P, Andrianakos R, Yang Y, Liu C, Lu W (2010) Kruppel-like factor 4 (Klf4) prevents

embryonic stem (ES) cell differentiation by regulating Nanog gene expression. The

Journal of biological chemistry 285: 9180-9189.

22. Ying QL, Wray J, Nichols J, Batlle-Morera L, Doble B, et al. (2008) The ground state of

embryonic stem cell self-renewal. Nature 453: 519-523.

23. Mitsui K, Tokuzawa Y, Itoh H, Segawa K, Murakami M, et al. (2003) The homeoprotein

Nanog is required for maintenance of pluripotency in mouse epiblast and ES cells. Cell

113: 631-642.

24. Niwa H, Miyazaki J, Smith AG (2000) Quantitative expression of Oct-3/4 defines

differentiation, dedifferentiation or self-renewal of ES cells. Nature genetics 24: 372-376.

25. Boyer LA, Lee TI, Cole MF, Johnstone SE, Levine SS, et al. (2005) Core transcriptional

regulatory circuitry in human embryonic stem cells. Cell 122: 947-956.

26. Chambers I (2004) The molecular basis of pluripotency in mouse embryonic stem cells.

Cloning and stem cells 6: 386-391.

27. Shi W, Wang H, Pan G, Geng Y, Guo Y, et al. (2006) Regulation of the pluripotency marker

Rex-1 by Nanog and Sox2. The Journal of biological chemistry 281: 23319-23325.

28. Thomson M, Liu SJ, Zou LN, Smith Z, Meissner A, et al. (2011) Pluripotency factors in

embryonic stem cells regulate differentiation into germ layers. Cell 145: 875-889.

29. Silva J, Nichols J, Theunissen TW, Guo G, van Oosten AL, et al. (2009) Nanog is the

gateway to the pluripotent ground state. Cell 138: 722-737.

Page 109: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

94

30. Brambrink T, Foreman R, Welstead GG, Lengner CJ, Wernig M, et al. (2008) Sequential

expression of pluripotency markers during direct reprogramming of mouse somatic cells.

Cell stem cell 2: 151-159.

31. Chambers I, Silva J, Colby D, Nichols J, Nijmeijer B, et al. (2007) Nanog safeguards

pluripotency and mediates germline development. Nature 450: 1230-1234.

32. Yamanaka S, Blau HM (2010) Nuclear reprogramming to a pluripotent state by three

approaches. Nature 465: 704-712.

33. Le Lievre CS, Le Douarin NM (1975) Mesenchymal derivatives of the neural crest: analysis

of chimaeric quail and chick embryos. Journal of embryology and experimental

morphology 34: 125-154.

34. Gurdon JB, Wilmut I (2011) Nuclear transfer to eggs and oocytes. Cold Spring Harbor

perspectives in biology 3.

35. Briggs R, King TJ (1952) Transplantation of living nuclei from blastula cells into enucleated

frogs' eggs. Proc Natl Acad Sci USA 38: 455-463.

36. Gurdon JB (1962) Adult frogs derived from the nuclei of single somatic cells. Developmental

biology 4: 256-273.

37. Gurdon JB (1962) The developmental capacity of nuclei taken from intestinal epithelium

cells of feeding tadpoles. Journal of embryology and experimental morphology 10: 622-

640.

38. Hochedlinger K, Jaenisch R (2002) Monoclonal mice generated by nuclear transfer from

mature B and T donor cells. Nature 415: 1035-1038.

39. Harris H, Miller OJ, Klein G, Worst P, Tachibana T (1969) Suppression of malignancy by

cell fusion. Nature 223: 363-368.

40. Blau HM, Chiu CP, Webster C (1983) Cytoplasmic activation of human nuclear genes in

stable heterocaryons. Cell 32: 1171-1180.

41. Blau HM, Pavlath GK, Hardeman EC, Chiu CP, Silberstein L, et al. (1985) Plasticity of the

differentiated state. Science 230: 758-766.

42. Chiu CP, Blau HM (1985) 5-Azacytidine permits gene activation in a previously

noninducible cell type. Cell 40: 417-424.

43. Tada M, Tada T, Lefebvre L, Barton SC, Surani MA (1997) Embryonic germ cells induce

epigenetic reprogramming of somatic nucleus in hybrid cells. The EMBO journal 16:

6510-6520.

44. Tada M, Takahama Y, Abe K, Nakatsuji N, Tada T (2001) Nuclear reprogramming of

somatic cells by in vitro hybridization with ES cells. Current biology : CB 11: 1553-

1558.

Page 110: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

95

45. Pereira CF, Terranova R, Ryan NK, Santos J, Morris KJ, et al. (2008) Heterokaryon-based

reprogramming of human B lymphocytes for pluripotency requires Oct4 but not Sox2.

PLoS genetics 4: e1000170.

46. Bhutani N, Brady JJ, Damian M, Sacco A, Corbel SY, et al. (2010) Reprogramming towards

pluripotency requires AID-dependent DNA demethylation. Nature 463: 1042-1047.

47. Schneuwly S, Klemenz R, Gehring WJ (1987) Redesigning the body plan of Drosophila by

ectopic expression of the homoeotic gene Antennapedia. Nature 325: 816-818.

48. Gehring WJ (1996) The master control gene for morphogenesis and evolution of the eye.

Genes to cells : devoted to molecular & cellular mechanisms 1: 11-15.

49. Davis RL, Weintraub H, Lassar AB (1987) Expression of a single transfected cDNA converts

fibroblasts to myoblasts. Cell 51: 987-1000.

50. Vierbuchen T, Ostermeier A, Pang ZP, Kokubu Y, Sudhof TC, et al. (2010) Direct

conversion of fibroblasts to functional neurons by defined factors. Nature 463: 1035-

1041.

51. Pang ZP, Yang N, Vierbuchen T, Ostermeier A, Fuentes DR, et al. (2011) Induction of

human neuronal cells by defined transcription factors. Nature: doi:10.1038/nature10202.

52. Toma JG, Akhavan M, Fernandes KJ, Barnabe-Heider F, Sadikot A, et al. (2001) Isolation of

multipotent adult stem cells from the dermis of mammalian skin. Nature cell biology 3:

778-784.

53. Takahashi K, Yamanaka S (2006) Induction of pluripotent stem cells from mouse embryonic

and adult fibroblast cultures by defined factors. Cell 126: 663-676.

54. Okita K, Ichisaka T, Yamanaka S (2007) Generation of germline-competent induced

pluripotent stem cells. Nature 448: 313-317.

55. Wernig M, Meissner A, Foreman R, Brambrink T, Ku M, et al. (2007) In vitro

reprogramming of fibroblasts into a pluripotent ES-cell-like state. Nature 448: 318-324.

56. Tokuzawa Y, Kaiho E, Maruyama M, Takahashi K, Mitsui K, et al. (2003) Fbx15 is a novel

target of Oct3/4 but is dispensable for embryonic stem cell self-renewal and mouse

development. Molecular and cellular biology 23: 2699-2708.

57. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, et al. (2007) Induction of

pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131: 861-872.

58. Yamanaka S (2009) A fresh look at iPS cells. Cell 137: 13-17.

59. Wernig M, Lengner CJ, Hanna J, Lodato MA, Steine E, et al. (2008) A drug-inducible

transgenic system for direct reprogramming of multiple somatic cell types. Nature

biotechnology 26: 916-924.

Page 111: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

96

60. Hanna J, Saha K, Pando B, van Zon J, Lengner CJ, et al. (2009) Direct cell reprogramming is

a stochastic process amenable to acceleration. Nature 462: 595-601.

61. Yamanaka S (2009) Elite and stochastic models for induced pluripotent stem cell generation.

Nature 460: 49-52.

62. Lister R, Pelizzola M, Kida YS, Hawkins RD, Nery JR, et al. (2011) Hotspots of aberrant

epigenomic reprogramming in human induced pluripotent stem cells. Nature 471: 68-73.

63. Kim K, Doi A, Wen B, Ng K, Zhao R, et al. (2010) Epigenetic memory in induced

pluripotent stem cells. Nature 467: 285-290.

64. Polo JM, Liu S, Figueroa ME, Kulalert W, Eminli S, et al. (2010) Cell type of origin

influences the molecular and functional properties of mouse induced pluripotent stem

cells. Nature biotechnology 28: 848-855.

65. Esteban MA, Wang T, Qin B, Yang J, Qin D, et al. (2010) Vitamin C enhances the

generation of mouse and human induced pluripotent stem cells. Cell stem cell 6: 71-79.

66. Yoshida Y, Takahashi K, Okita K, Ichisaka T, Yamanaka S (2009) Hypoxia enhances the

generation of induced pluripotent stem cells. Cell stem cell 5: 237-241.

67. Huangfu D, Maehr R, Guo W, Eijkelenboom A, Snitow M, et al. (2008) Induction of

pluripotent stem cells by defined factors is greatly improved by small-molecule

compounds. Nature biotechnology 26: 795-797.

68. Huangfu D, Osafune K, Maehr R, Guo W, Eijkelenboom A, et al. (2008) Induction of

pluripotent stem cells from primary human fibroblasts with only Oct4 and Sox2. Nature

biotechnology 26: 1269-1275.

69. Utikal J, Polo JM, Stadtfeld M, Maherali N, Kulalert W, et al. (2009) Immortalization

eliminates a roadblock during cellular reprogramming into iPS cells. Nature 460: 1145-

1148.

70. Kawamura T, Suzuki J, Wang YV, Menendez S, Morera LB, et al. (2009) Linking the p53

tumour suppressor pathway to somatic cell reprogramming. Nature 460: 1140-1144.

71. Marion RM, Strati K, Li H, Murga M, Blanco R, et al. (2009) A p53-mediated DNA damage

response limits reprogramming to ensure iPS cell genomic integrity. Nature 460: 1149-

1153.

72. Hong H, Takahashi K, Ichisaka T, Aoi T, Kanagawa O, et al. (2009) Suppression of induced

pluripotent stem cell generation by the p53-p21 pathway. Nature 460: 1132-1135.

73. Li H, Collado M, Villasante A, Strati K, Ortega S, et al. (2009) The Ink4/Arf locus is a

barrier for iPS cell reprogramming. Nature 460: 1136-1139.

Page 112: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

97

74. Woltjen K, Michael IP, Mohseni P, Desai R, Mileikovsky M, et al. (2009) piggyBac

transposition reprograms fibroblasts to induced pluripotent stem cells. Nature 458: 766-

770.

75. Zhou H, Wu S, Joo JY, Zhu S, Han DW, et al. (2009) Generation of induced pluripotent stem

cells using recombinant proteins. Cell stem cell 4: 381-384.

76. Okita K, Nakagawa M, Hyenjong H, Ichisaka T, Yamanaka S (2008) Generation of mouse

induced pluripotent stem cells without viral vectors. Science 322: 949-953.

77. Yu J, Hu K, Smuga-Otto K, Tian S, Stewart R, et al. (2009) Human induced pluripotent stem

cells free of vector and transgene sequences. Science 324: 797-801.

78. Warren L, Manos PD, Ahfeldt T, Loh YH, Li H, et al. (2010) Highly efficient

reprogramming to pluripotency and directed differentiation of human cells with synthetic

modified mRNA. Cell stem cell 7: 618-630.

79. Anokye-Danso F, Trivedi CM, Juhr D, Gupta M, Cui Z, et al. (2011) Highly Efficient

miRNA-Mediated Reprogramming of Mouse and Human Somatic Cells to Pluripotency.

Cell stem cell 8: 376-388.

80. Gore A, Li Z, Fung HL, Young JE, Agarwal S, et al. (2011) Somatic coding mutations in

human induced pluripotent stem cells. Nature 471: 63-67.

81. Hussein SM, Batada NN, Vuoristo S, Ching RW, Autio R, et al. (2011) Copy number

variation and selection during reprogramming to pluripotency. Nature 471: 58-62.

82. Laurent LC, Ulitsky I, Slavin I, Tran H, Schork A, et al. (2011) Dynamic changes in the copy

number of pluripotency and cell proliferation genes in human ESCs and iPSCs during

reprogramming and time in culture. Cell stem cell 8: 106-118.

83. Nakagawa M, Koyanagi M, Tanabe K, Takahashi K, Ichisaka T, et al. (2008) Generation of

induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nature

biotechnology 26: 101-106.

84. Nakagawa M, Takizawa N, Narita M, Ichisaka T, Yamanaka S (2010) Promotion of direct

reprogramming by transformation-deficient Myc. Proceedings of the National Academy

of Sciences of the United States of America 107: 14152-14157.

85. Maekawa M, Yamaguchi K, Nakamura T, Shibukawa R, Kodanaka I, et al. (2011) Direct

reprogramming of somatic cells is promoted by maternal transcription factor Glis1.

Nature 474: 225-229.

86. Kim JB, Zaehres H, Wu G, Gentile L, Ko K, et al. (2008) Pluripotent stem cells induced

from adult neural stem cells by reprogramming with two factors. Nature 454: 646-650.

87. Hester ME, Song S, Miranda CJ, Eagle A, Schwartz PH, et al. (2009) Two factor

reprogramming of human neural stem cells into pluripotency. PloS one 4: e7044.

Page 113: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

98

88. Kim JB, Greber B, Arauzo-Bravo MJ, Meyer J, Park KI, et al. (2009) Direct reprogramming

of human neural stem cells by OCT4. Nature 461: 649-643.

89. Kim JB, Sebastiano V, Wu G, Arauzo-Bravo MJ, Sasse P, et al. (2009) Oct4-induced

pluripotency in adult neural stem cells. Cell 136: 411-419.

90. Shi Y, Do JT, Desponts C, Hahm HS, Scholer HR, et al. (2008) A combined chemical and

genetic approach for the generation of induced pluripotent stem cells. Cell stem cell 2:

525-528.

91. Knudson AG, Jr. (1971) Mutation and cancer: statistical study of retinoblastoma.

Proceedings of the National Academy of Sciences of the United States of America 68:

820-823.

92. Knudson AG, Jr. (1977) Mutation and cancer in man. Cancer 39: 1882-1886.

93. Esteller M (2008) Epigenetics in cancer. The New England journal of medicine 358: 1148-

1159.

94. Gal-Yam EN, Saito Y, Egger G, Jones PA (2008) Cancer epigenetics: modifications,

screening, and therapy. Annual review of medicine 59: 267-280.

95. Jones PA, Laird PW (1999) Cancer epigenetics comes of age. Nature genetics 21: 163-167.

96. Jones PA (1996) DNA methylation errors and cancer. Cancer research 56: 2463-2467.

97. Nan X, Ng HH, Johnson CA, Laherty CD, Turner BM, et al. (1998) Transcriptional

repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase

complex. Nature 393: 386-389.

98. Jones PL, Veenstra GJ, Wade PA, Vermaak D, Kass SU, et al. (1998) Methylated DNA and

MeCP2 recruit histone deacetylase to repress transcription. Nature genetics 19: 187-191.

99. Kouzarides T (2007) Chromatin modifications and their function. Cell 128: 693-705.

100. Li E, Bestor TH, Jaenisch R (1992) Targeted mutation of the DNA methyltransferase gene

results in embryonic lethality. Cell 69: 915-926.

101. Okano M, Xie S, Li E (1998) Cloning and characterization of a family of novel mammalian

DNA (cytosine-5) methyltransferases. Nature genetics 19: 219-220.

102. Okano M, Bell DW, Haber DA, Li E (1999) DNA methyltransferases Dnmt3a and Dnmt3b

are essential for de novo methylation and mammalian development. Cell 99: 247-257.

103. Feinberg AP, Vogelstein B (1983) Hypomethylation distinguishes genes of some human

cancers from their normal counterparts. Nature 301: 89-92.

104. Coulondre C, Miller JH, Farabaugh PJ, Gilbert W (1978) Molecular basis of base

substitution hotspots in Escherichia coli. Nature 274: 775-780.

Page 114: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

99

105. Li E, Beard C, Jaenisch R (1993) Role for DNA methylation in genomic imprinting. Nature

366: 362-365.

106. Laird PW, Jackson-Grusby L, Fazeli A, Dickinson SL, Jung WE, et al. (1995) Suppression

of intestinal neoplasia by DNA hypomethylation. Cell 81: 197-205.

107. Feinberg AP (1993) Genomic imprinting and gene activation in cancer. Nature genetics 4:

110-113.

108. Beaulieu N, Morin S, Chute IC, Robert MF, Nguyen H, et al. (2002) An essential role for

DNA methyltransferase DNMT3B in cancer cell survival. The Journal of biological

chemistry 277: 28176-28181.

109. Esteller M, Hamilton SR, Burger PC, Baylin SB, Herman JG (1999) Inactivation of the

DNA repair gene O6-methylguanine-DNA methyltransferase by promoter

hypermethylation is a common event in primary human neoplasia. Cancer research 59:

793-797.

110. Esteller M (2006) Epigenetics provides a new generation of oncogenes and tumour-

suppressor genes. British journal of cancer 94: 179-183.

111. Jacobs JJ, Kieboom K, Marino S, DePinho RA, van Lohuizen M (1999) The oncogene and

Polycomb-group gene bmi-1 regulates cell proliferation and senescence through the ink4a

locus. Nature 397: 164-168.

112. Bruggeman SW, Hulsman D, Tanger E, Buckle T, Blom M, et al. (2007) Bmi1 controls

tumor development in an Ink4a/Arf-independent manner in a mouse model for glioma.

Cancer Cell 12: 328-341.

113. Lessard J, Sauvageau G (2003) Bmi-1 determines the proliferative capacity of normal and

leukaemic stem cells. Nature 423: 255-260.

114. Lee TI, Jenner RG, Boyer LA, Guenther MG, Levine SS, et al. (2006) Control of

developmental regulators by Polycomb in human embryonic stem cells. Cell 125: 301-

313.

115. Widschwendter M, Fiegl H, Egle D, Mueller-Holzner E, Spizzo G, et al. (2007) Epigenetic

stem cell signature in cancer. Nature genetics 39: 157-158.

116. Kaminskas E, Farrell A, Abraham S, Baird A, Hsieh LS, et al. (2005) Approval summary:

azacitidine for treatment of myelodysplastic syndrome subtypes. Clinical cancer research

: an official journal of the American Association for Cancer Research 11: 3604-3608.

117. Dey P (2006) Chromatin remodeling, cancer and chemotherapy. Current medicinal

chemistry 13: 2909-2919.

118. Lane AA, Chabner BA (2009) Histone deacetylase inhibitors in cancer therapy. Journal of

clinical oncology : official journal of the American Society of Clinical Oncology 27:

5459-5468.

Page 115: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

100

119. Ben-Porath I, Thomson MW, Carey VJ, Ge R, Bell GW, et al. (2008) An embryonic stem

cell-like gene expression signature in poorly differentiated aggressive human tumors.

Nature genetics 40: 499-507.

120. Kim J, Woo AJ, Chu J, Snow JW, Fujiwara Y, et al. (2010) A Myc network accounts for

similarities between embryonic stem and cancer cell transcription programs. Cell 143:

313-324.

121. Tlsty TD (2011) Phenotypic plasticity in human somatic cells. Plenary Talk, ISSCR 9th

Annual Meeting, Toronto, Canada.

122. Hendrix MJ, Seftor EA, Hess AR, Seftor RE (2003) Vasculogenic mimicry and tumour-cell

plasticity: lessons from melanoma. Nature reviews Cancer 3: 411-421.

123. Ricci-Vitiani L, Pallini R, Biffoni M, Todaro M, Invernici G, et al. (2010) Tumour

vascularization via endothelial differentiation of glioblastoma stem-like cells. Nature 468:

824-828.

124. Wang R, Chadalavada K, Wilshire J, Kowalik U, Hovinga KE, et al. (2010) Glioblastoma

stem-like cells give rise to tumour endothelium. Nature 468: 829-833.

125. Kleinsmith LJ, Pierce GB, Jr. (1964) Multipotentiality of Single Embryonal Carcinoma

Cells. Cancer research 24: 1544-1551.

126. Mintz B, Illmensee K (1975) Normal genetically mosaic mice produced from malignant

teratocarcinoma cells. Proceedings of the National Academy of Sciences of the United

States of America 72: 3585-3589.

127. Papaioannou VE, McBurney MW, Gardner RL, Evans MJ (1975) Fate of teratocarcinoma

cells injected into early mouse embryos. Nature 258: 70-73.

128. Rosner MH, Vigano MA, Ozato K, Timmons PM, Poirier F, et al. (1990) A POU-domain

transcription factor in early stem cells and germ cells of the mammalian embryo. Nature

345: 686-692.

129. Gidekel S, Pizov G, Bergman Y, Pikarsky E (2003) Oct-3/4 is a dose-dependent oncogenic

fate determinant. Cancer cell 4: 361-370.

130. Looijenga LH, Stoop H, de Leeuw HP, de Gouveia Brazao CA, Gillis AJ, et al. (2003)

POU5F1 (OCT3/4) identifies cells with pluripotent potential in human germ cell tumors.

Cancer research 63: 2244-2250.

131. Ezeh UI, Turek PJ, Reijo RA, Clark AT (2005) Human embryonic stem cell genes OCT4,

NANOG, STELLAR, and GDF3 are expressed in both seminoma and breast carcinoma.

Cancer 104: 2255-2265.

132. Clement V, Sanchez P, de Tribolet N, Radovanovic I, Ruiz i Altaba A (2007)

HEDGEHOG-GLI1 signaling regulates human glioma growth, cancer stem cell self-

renewal, and tumorigenicity. Curr Biol 17: 165-172.

Page 116: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

101

133. Holmberg J, He X, Peredo I, Orrego A, Hesselager G, et al. (2011) Activation of neural and

pluripotent stem cell signatures correlates with increased malignancy in human glioma.

PloS one 6: e18454.

134. Ji J, Werbowetski-Ogilvie TE, Zhong B, Hong SH, Bhatia M (2009) Pluripotent

transcription factors possess distinct roles in normal versus transformed human stem

cells. PloS one 4: e8065.

135. McKinnell RG, Deggins BA, Labat DD (1969) Transplantation of pluripotential nuclei from

triploid frog tumors. Science 165: 394-396.

136. Li L, Connelly MC, Wetmore C, Curran T, Morgan JI (2003) Mouse embryos cloned from

brain tumors. Cancer research 63: 2733-2736.

137. Hochedlinger K, Blelloch R, Brennan C, Yamada Y, Kim M, et al. (2004) Reprogramming

of a melanoma genome by nuclear transplantation. Genes & development 18: 1875-1885.

138. Blelloch RH, Hochedlinger K, Yamada Y, Brennan C, Kim M, et al. (2004) Nuclear cloning

of embryonal carcinoma cells. Proceedings of the National Academy of Sciences of the

United States of America 101: 13985-13990.

139. Miyoshi N, Ishii H, Nagai K, Hoshino H, Mimori K, et al. (2010) Defined factors induce

reprogramming of gastrointestinal cancer cells. Proceedings of the National Academy of

Sciences of the United States of America 107: 40-45.

140. Carette JE, Pruszak J, Varadarajan M, Blomen VA, Gokhale S, et al. (2010) Generation of

iPSCs from cultured human malignant cells. Blood 115: 4039-4042.

141. Bonnet D, Dick JE (1997) Human acute myeloid leukemia is organized as a hierarchy that

originates from a primitive hematopoietic cell. Nat Med 3: 730-737.

142. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF (2003) Prospective

identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 100: 3983-

3988.

143. Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, et al. (2003) Identification of a

cancer stem cell in human brain tumors. Cancer Res 63: 5821-5828.

144. Gibbs CP, Kukekov VG, Reith JD, Tchigrinova O, Suslov ON, et al. (2005) Stem-like cells

in bone sarcomas: implications for tumorigenesis. Neoplasia 7: 967-976.

145. Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ (2005) Prospective identification

of tumorigenic prostate cancer stem cells. Cancer Res 65: 10946-10951.

146. Fang D, Nguyen TK, Leishear K, Finko R, Kulp AN, et al. (2005) A tumorigenic

subpopulation with stem cell properties in melanomas. Cancer Res 65: 9328-9337.

147. Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, et al. (2007) Identification

and expansion of human colon-cancer-initiating cells. Nature 445: 111-115.

Page 117: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

102

148. Eramo A, Lotti F, Sette G, Pilozzi E, Biffoni M, et al. (2007) Identification and expansion

of the tumorigenic lung cancer stem cell population. Cell Death Differ.

149. Prince ME, Sivanandan R, Kaczorowski A, Wolf GT, Kaplan MJ, et al. (2007)

Identification of a subpopulation of cells with cancer stem cell properties in head and

neck squamous cell carcinoma. Proc Natl Acad Sci U S A 104: 973-978.

150. Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, et al. (2007) Identification of pancreatic

cancer stem cells. Cancer Res 67: 1030-1037.

151. Wu C, Wei Q, Utomo V, Nadesan P, Whetstone H, et al. (2007) Side population cells

isolated from mesenchymal neoplasms have tumor initiating potential. Cancer Res 67:

8216-8222.

152. Lerner BH (2004) Sins of omission--cancer research without informed consent. N Engl J

Med 351: 628-630.

153. Moore AE, Rhoads CP, Southam CM (1957) Homotransplantation of human cell lines.

Science 125: 158-160.

154. Brunschwig A, Southam CM, Levin AG (1965) Host resistance to cancer. Clinical

experiments by homotransplants, autotransplants and admixture of autologous leucocytes.

Ann Surg 162: 416-425.

155. Wang JC, Dick JE (2005) Cancer stem cells: lessons from leukemia. Trends Cell Biol 15:

494-501.

156. Donnenberg VS, Donnenberg AD (2005) Multiple drug resistance in cancer revisited: the

cancer stem cell hypothesis. J Clin Pharmacol 45: 872-877.

157. Dean M, Fojo T, Bates S (2005) Tumour stem cells and drug resistance. Nat Rev Cancer 5:

275-284.

158. Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, et al. (2006) Glioma stem cells promote

radioresistance by preferential activation of the DNA damage response. Nature 444: 756-

760.

159. Notta F, Mullighan CG, Wang JC, Poeppl A, Doulatov S, et al. (2011) Evolution of human

BCR-ABL1 lymphoblastic leukaemia-initiating cells. Nature 469: 362-367.

160. Merchant TE, Pollack IF, Loeffler JS (2010) Brain tumors across the age spectrum: biology,

therapy, and late effects. Semin Radiat Oncol 20: 58-66.

161. Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, et al. (2007) The 2007 WHO

classification of tumours of the central nervous system. Acta Neuropathol 114: 97-109.

162. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, et al. (2005) Radiotherapy plus

concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352: 987-996.

Page 118: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

103

163. Ohgaki H, Kleihues P (2007) Genetic pathways to primary and secondary glioblastoma. The

American journal of pathology 170: 1445-1453.

164. McLendon R, Friedman A, Bigner D, Van Meir EG, Brat DJ, et al. (2008) Comprehensive

genomic characterization defines human glioblastoma genes and core pathways. Nature

455: 1061-1068.

165. Parsons DW, Jones S, Zhang X, Lin JC, Leary RJ, et al. (2008) An integrated genomic

analysis of human glioblastoma multiforme. Science 321: 1807-1812.

166. Esteller M, Corn PG, Baylin SB, Herman JG (2001) A gene hypermethylation profile of

human cancer. Cancer research 61: 3225-3229.

167. Martinez R, Schackert G (2007) Epigenetic aberrations in malignant gliomas: an open door

leading to better understanding and treatment. Epigenetics : official journal of the DNA

Methylation Society 2: 147-150.

168. Martinez R, Martin-Subero JI, Rohde V, Kirsch M, Alaminos M, et al. (2009) A

microarray-based DNA methylation study of glioblastoma multiforme. Epigenetics :

official journal of the DNA Methylation Society 4: 255-264.

169. Abdouh M, Facchino S, Chatoo W, Balasingam V, Ferreira J, et al. (2009) BMI1 sustains

human glioblastoma multiforme stem cell renewal. The Journal of neuroscience : the

official journal of the Society for Neuroscience 29: 8884-8896.

170. Facchino S, Abdouh M, Chatoo W, Bernier G (2010) BMI1 confers radioresistance to

normal and cancerous neural stem cells through recruitment of the DNA damage

response machinery. The Journal of neuroscience : the official journal of the Society for

Neuroscience 30: 10096-10111.

171. Suva ML, Riggi N, Janiszewska M, Radovanovic I, Provero P, et al. (2009) EZH2 is

essential for glioblastoma cancer stem cell maintenance. Cancer research 69: 9211-9218.

172. Nakamura M, Watanabe T, Yonekawa Y, Kleihues P, Ohgaki H (2001) Promoter

methylation of the DNA repair gene MGMT in astrocytomas is frequently associated

with G:C --> A:T mutations of the TP53 tumor suppressor gene. Carcinogenesis 22:

1715-1719.

173. Esteller M, Garcia-Foncillas J, Andion E, Goodman SN, Hidalgo OF, et al. (2000)

Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to

alkylating agents. The New England journal of medicine 343: 1350-1354.

174. Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, et al. (2005) MGMT gene

silencing and benefit from temozolomide in glioblastoma. The New England journal of

medicine 352: 997-1003.

175. Martinez R, Setien F, Voelter C, Casado S, Quesada MP, et al. (2007) CpG island promoter

hypermethylation of the pro-apoptotic gene caspase-8 is a common hallmark of relapsed

glioblastoma multiforme. Carcinogenesis 28: 1264-1268.

Page 119: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

104

176. Jacks T, Remington L, Williams BO, Schmitt EM, Halachmi S, et al. (1994) Tumor

spectrum analysis in p53-mutant mice. Curr Biol 4: 1-7.

177. Wang Y, Yang J, Zheng H, Tomasek GJ, Zhang P, et al. (2009) Expression of mutant p53

proteins implicates a lineage relationship between neural stem cells and malignant

astrocytic glioma in a murine model. Cancer Cell 15: 514-526.

178. Nolan PM, Hugill A, Cox RD (2002) ENU mutagenesis in the mouse: application to human

genetic disease. Briefings in functional genomics & proteomics 1: 278-289.

179. Oda H, Zhang S, Tsurutani N, Shimizu S, Nakatsuru Y, et al. (1997) Loss of p53 is an early

event in induction of brain tumors in mice by transplacental carcinogen exposure. Cancer

Res 57: 646-650.

180. Ward RJ, Dirks PB (2010) Unpublished.

181. Pollard SM, Yoshikawa K, Clarke ID, Danovi D, Stricker S, et al. (2009) Glioma stem cell

lines expanded in adherent culture have tumor-specific phenotypes and are suitable for

chemical and genetic screens. Cell Stem Cell 4: 568-580.

182. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, et al. (2004) Identification of human

brain tumour initiating cells. Nature 432: 396-401.

183. Ignatova TN, Kukekov VG, Laywell ED, Suslov ON, Vrionis FD, et al. (2002) Human

cortical glial tumors contain neural stem-like cells expressing astroglial and neuronal

markers in vitro. Glia 39: 193-206.

184. Hemmati HD, Nakano I, Lazareff JA, Masterman-Smith M, Geschwind DH, et al. (2003)

Cancerous stem cells can arise from pediatric brain tumors. Proc Natl Acad Sci U S A

100: 15178-15183.

185. Galli R, Binda E, Orfanelli U, Cipelletti B, Gritti A, et al. (2004) Isolation and

characterization of tumorigenic, stem-like neural precursors from human glioblastoma.

Cancer Res 64: 7011-7021.

186. Read TA, Fogarty MP, Markant SL, McLendon RE, Wei Z, et al. (2009) Identification of

CD15 as a marker for tumor-propagating cells in a mouse model of medulloblastoma.

Cancer Cell 15: 135-147.

187. Ward RJ, Lee L, Graham K, Satkunendran T, Yoshikawa K, et al. (2009) Multipotent

CD15+ cancer stem cells in patched-1-deficient mouse medulloblastoma. Cancer Res 69:

4682-4690.

188. Son MJ, Woolard K, Nam DH, Lee J, Fine HA (2009) SSEA-1 is an enrichment marker for

tumor-initiating cells in human glioblastoma. Cell Stem Cell 4: 440-452.

189. Clement V, Marino D, Cudalbu C, Hamou MF, Mlynarik V, et al. (2010) Marker-

independent identification of glioma-initiating cells. Nat Methods 7: 224-228.

Page 120: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

105

190. Piccirillo SG, Reynolds BA, Zanetti N, Lamorte G, Binda E, et al. (2006) Bone

morphogenetic proteins inhibit the tumorigenic potential of human brain tumour-

initiating cells. Nature 444: 761-765.

191. Fan X, Khaki L, Zhu TS, Soules ME, Talsma CE, et al. (2010) NOTCH pathway blockade

depletes CD133-positive glioblastoma cells and inhibits growth of tumor neurospheres

and xenografts. Stem Cells 28: 5-16.

192. Hotta A, Cheung AY, Farra N, Vijayaragavan K, Seguin CA, et al. (2009) Isolation of

human iPS cells using EOS lentiviral vectors to select for pluripotency. Nature methods

6: 370-376.

193. Cox WG, Singer VL (1999) A high-resolution, fluorescence-based method for localization

of endogenous alkaline phosphatase activity. The journal of histochemistry and

cytochemistry : official journal of the Histochemistry Society 47: 1443-1456.

194. Hotta A, Cheung AY, Farra N, Garcha K, Chang WY, et al. (2009) EOS lentiviral vector

selection system for human induced pluripotent stem cells. Nature protocols 4: 1828-

1844.

195. Silva J, Barrandon O, Nichols J, Kawaguchi J, Theunissen TW, et al. (2008) Promotion of

reprogramming to ground state pluripotency by signal inhibition. PLoS biology 6: e253.

196. Ying QL, Stavridis M, Griffiths D, Li M, Smith A (2003) Conversion of embryonic stem

cells into neuroectodermal precursors in adherent monoculture. Nature biotechnology 21:

183-186.

197. Ying QL, Smith AG (2003) Defined conditions for neural commitment and differentiation.

Methods in enzymology 365: 327-341.

198. Hotta A, Ellis J (2008) Retroviral vector silencing during iPS cell induction: an epigenetic

beacon that signals distinct pluripotent states. Journal of cellular biochemistry 105: 940-

948.

199. Ron-Bigger S, Bar-Nur O, Isaac S, Bocker M, Lyko F, et al. (2010) Aberrant epigenetic

silencing of tumor suppressor genes is reversed by direct reprogramming. Stem cells 28:

1349-1354.

200. Fehling HJ, Lacaud G, Kubo A, Kennedy M, Robertson S, et al. (2003) Tracking mesoderm

induction and its specification to the hemangioblast during embryonic stem cell

differentiation. Development 130: 4217-4227.

201. Smith KN, Singh AM, Dalton S (2010) Myc represses primitive endoderm differentiation in

pluripotent stem cells. Cell stem cell 7: 343-354.

202. Muller FJ, Goldmann J, Loser P, Loring JF (2010) A call to standardize teratoma assays

used to define human pluripotent cell lines. Cell stem cell 6: 412-414.

Page 121: Reprogramming Mouse Glioma Stem Cells with Defined · PDF fileReprogramming Mouse Glioma Stem Cells with Defined Factors ... Sick Children for advice and support throughout my MSc

106

203. Muller FJ, Schuldt BM, Williams R, Mason D, Altun G, et al. (2011) A bioinformatic assay

for pluripotency in human cells. Nature methods 8: 315-317.

204. Moon JH, Heo JS, Kim JS, Jun EK, Lee JH, et al. (2011) Reprogramming fibroblasts into

induced pluripotent stem cells with Bmi1. Cell research.

205. Wong DJ, Liu H, Ridky TW, Cassarino D, Segal E, et al. (2008) Module map of stem cell

genes guides creation of epithelial cancer stem cells. Cell stem cell 2: 333-344.

206. Heddleston JM, Li Z, McLendon RE, Hjelmeland AB, Rich JN (2009) The hypoxic

microenvironment maintains glioblastoma stem cells and promotes reprogramming

towards a cancer stem cell phenotype. Cell cycle 8: 3274-3284.

207. Du Z, Jia D, Liu S, Wang F, Li G, et al. (2009) Oct4 is expressed in human gliomas and

promotes colony formation in glioma cells. Glia 57: 724-733.

208. Zbinden M, Duquet A, Lorente-Trigos A, Ngwabyt SN, Borges I, et al. (2010) NANOG

regulates glioma stem cells and is essential in vivo acting in a cross-functional network

with GLI1 and p53. The EMBO journal 29: 2659-2674.

209. Gallo M, Dirks PB (2011) Unpublished.

210. Mathieu J, Zhang Z, Zhou W, Wang AJ, Heddleston JM, et al. (2011) HIF Induces Human

Embryonic Stem Cell Markers in Cancer Cells. Cancer research 71: 4640-4652.

211. Zhao S, Yuan Q, Hao H, Guo Y, Liu S, et al. (2011) Expression of OCT4 pseudogenes in

human tumours: lessons from glioma and breast carcinoma. The Journal of pathology

223: 672-682.

212. Suo G, Han J, Wang X, Zhang J, Zhao Y, et al. (2005) Oct4 pseudogenes are transcribed in

cancers. Biochemical and biophysical research communications 337: 1047-1051.

213. Cauffman G, Liebaers I, Van Steirteghem A, Van de Velde H (2006) POU5F1 isoforms

show different expression patterns in human embryonic stem cells and preimplantation

embryos. Stem cells 24: 2685-2691.

214. Atlasi Y, Mowla SJ, Ziaee SA, Gokhale PJ, Andrews PW (2008) OCT4 spliced variants are

differentially expressed in human pluripotent and nonpluripotent cells. Stem cells 26:

3068-3074.

215. Liedtke S, Stephan M, Kogler G (2008) Oct4 expression revisited: potential pitfalls for data

misinterpretation in stem cell research. Biological chemistry 389: 845-850.

216. Monk M, Holding C (2001) Human embryonic genes re-expressed in cancer cells.

Oncogene 20: 8085-8091.

217. Booth HA, Holland PW (2004) Eleven daughters of NANOG. Genomics 84: 229-238.