wnt signaling in human neural stem cells and brain tumour ... › bitstream › 1807 › ... ·...

95
Wnt Signaling in Human Neural Stem Cells and Brain Tumour Stem Cells by Caroline Brandon A thesis submitted in conformity with the requirements for the degree of Master of Science Laboratory Medicine and Pathobiology University of Toronto © Copyright by Caroline Brandon 2010

Upload: others

Post on 05-Jul-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

Wnt Signaling in Human Neural Stem Cells and Brain Tumour Stem Cells

by

Caroline Brandon

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

Laboratory Medicine and Pathobiology University of Toronto

© Copyright by Caroline Brandon 2010

Page 2: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

ii

Targeting Wnt Signaling in Human Neural Stem Cells and Brain

Tumour Stem Cells

Caroline Brandon

Master of Science

Laboratory Medicine and Pathobiology University of Toronto

2010

Abstract

We sought to determine whether activation of the Wnt signaling pathway altered the function of

hNSCs in vitro. We took three approaches to activate Wnt signaling: Wnt3a, constitutively

stabilized β-catenin (ΔN90), and the GSK3 inhibitor BIO. While Wnt3a and ΔN90 had no effect

on proliferation in both stem cell (+EGF/FGF) and differentiating (-EGF/FGF) conditions, BIO

reduced proliferation in both. All methods of Wnt signaling activation promoted neuronal lineage

commitment during hNSC differentiation. Furthermore, BIO was able to induce mild neuronal

differentiation in stem cell conditions, suggesting that GSK3-inhibition interferes with several

pathways to regulate hNSC fate decisions.

We also probed BTSC function using BIO-mediated GSK3 inhibition. We found that in stem cell

conditions, BIO was able to induce neuronal differentiation, decrease proliferation, and induce

cell cycle arrest. Together this data suggests that GSK3-inhibition, possibly through activation of

Wnt signaling, may offer a novel mechanism for the differentiation treatment of glioblastomas.

Page 3: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

iii

Acknowledgments

Science is obsessed with quantification. How much? How far? How long? What cannot be

quantified is the amount of gratitude I feel for all the support and guidance I have received

throughout my life, especially over the last few years from colleagues, friends, and family. I

thank you all for keeping me sane in trying times, for making me laugh when my first instinct

was to cry, and for stretching my imagination beyond its limited confines. I dedicate this body of

work to my parents. A girl couldn’t ask for bigger and better fans. Without the both of you, goals

would stay goals and dreams would remain dreams. Thank you for helping make them a reality. I

love you both more than science can explain.

Page 4: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

iv

Table of Contents

Acknowledgments.......................................................................................................................... iii

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

List of Tables ................................................................................................................................ vii

List of Figures .............................................................................................................................. viii

Abbreviations ................................................................................................................................. ix

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

1 Introduction to the Literature ..................................................................................................... 1

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

1.1.1 Embryonic Stem Cells ............................................................................................ 1

1.1.2 Adult Stem Cells ..................................................................................................... 1

1.2 Neural Stem Cells ............................................................................................................... 2

1.2.1 Regulation of NSC activity ..................................................................................... 3

1.3 Cancer Stem Cells............................................................................................................... 4

1.3.1 Stochastic Model versus the Cancer Stem Cell Hypothesis ................................... 4

1.3.2 Lessons from Leukemia .......................................................................................... 7

1.4 Brain Tumours .................................................................................................................... 7

1.4.1 Identification of Brain Tumour Stem Cells ............................................................ 8

1.4.2 Intrinsic regulators of Brain Tumours and BTSCs ................................................. 9

1.4.3 Extrinsic Regulation of Brain Tumours and BTSCs............................................. 10

1.5 Introduction to Wnt Signaling .......................................................................................... 11

1.5.1 The Canonical Wnt Signaling Pathway ................................................................ 11

1.5.2 Regulatory mechanisms of the Canonical Wnt Pathway...................................... 12

1.5.3 GSK3: the master of multitasking......................................................................... 13

1.6 Wnt Signaling in Development and Stem Cells ............................................................... 18

Page 5: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

v

1.6.1 Wnt in Neural Development and NSCs ................................................................ 19

1.7 Wnt Signaling in Cancer and CSCs .................................................................................. 20

1.7.1 Wnt in Colorectal Cancers .................................................................................... 20

1.7.2 Wnt in Skin Cancers ............................................................................................. 20

1.7.3 Wnt in Leukemias ................................................................................................. 21

1.8 Wnt in Brain Tumours ...................................................................................................... 22

1.8.1 Wnt in Medulloblastomas ..................................................................................... 22

1.8.2 Wnt in Glioblastoma Multiforme.......................................................................... 22

1.8.3 Brain tumour stem cells in vitro............................................................................ 23

1.9 Thesis Rationale and Aim................................................................................................. 26

1.10Thesis Hypothesis ............................................................................................................. 26

Chapter 2 ....................................................................................................................................... 27

2 Materials and Methods............................................................................................................. 27

2.1 Cell culture and differentiation protocol........................................................................... 27

2.2 Immunoblotting................................................................................................................. 27

2.3 Immunofluorescence......................................................................................................... 28

2.4 Intracellular Flow Cytometry............................................................................................ 28

2.5 Luciferase Assay............................................................................................................... 29

2.6 Transfection of cells.......................................................................................................... 29

2.7 MTT assays ....................................................................................................................... 29

2.8 BrdU labeling.................................................................................................................... 29

Chapter 3 ....................................................................................................................................... 31

3 Results ...................................................................................................................................... 31

3.1 Wnt signaling promotes a neuronal cell fate choice in human fetal neural stem cells ..... 31

3.1.1 Human fetal neural stem cells express key Wnt pathway components and exhibit low baseline TCF/LEF-mediated transcription......................................... 31

Page 6: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

vi

3.1.2 Wnt3a does not alter human fetal neural stem cell proliferation .......................... 35

3.1.3 Wnt3a promotes a neuronal cell fate choice in human fetal neural stem cells under differentiating conditions ............................................................................ 37

3.1.4 Stabilized β-catenin activates Tcf/Lef—mediated transcription........................... 40

3.1.5 Stabilized β-catenin does not alter human neural stem cell proliferation. ............ 42

3.1.6 Stabilized β-catenin promotes neuronal cell fate choice in human fetal neural stem cells under differentiating conditions ........................................................... 44

3.1.7 BIO—mediated inhibition of GSK3 activates TCF/LEF transcription ................ 47

3.1.8 BIO promotes differentiating human fetal neural stem cells to slow proliferation and exit the precursor state .............................................................. 49

3.1.9 BIO promotes a neuronal cell fate choice in differentiating human fetal neural stem cells............................................................................................................... 51

3.1.10 BIO decreases proliferation and induces neuronal differentiation of human neural stem cells in EGF and FGF........................................................................ 54

3.2 GSK3 inhibition induces neuronal differentiation of brain tumour stem cells ................. 56

3.2.1 Brain tumour stem cells express Wnt pathway components and can activate TCF/LEF-transcriptional activity.......................................................................... 56

3.2.2 BIO induces neuronal differentiation of brain tumour stem cells......................... 59

3.2.3 BIO treatment induces brain tumour stem cells to exit the cell cycle and decrease proliferation............................................................................................ 62

Chapter 4 ....................................................................................................................................... 64

4 Discussions and Future Directions........................................................................................... 64

4.1 Wnt signaling in human neural stem cells ........................................................................ 64

4.2 Targeting the Wnt pathway for differentiation therapy of brain tumour stem cells ......... 69

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

Page 7: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

vii

List of Tables

Table 1 .......................................................................................................................................... 25

Page 8: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

viii

List of Figures

Figure 1 – The Stochastic model and the cancer stem cell model. ................................................. 6

Figure 2 – The canonical Wnt Signaling Pathway........................................................................ 17

Figure 3 - HumanNSCsexpresscanonicalWntsignalingpathwaycomponents................... 32

Figure 4 – hNSC lines activate TCF/LEF transcriptional activity in response to Wnt3a............. 34

Figure 5 – Wnt3a does not alter hNSC proliferation. ................................................................... 36

Figure 6 – Wnt3a promotes a neuronal cell fate in differentiating hNSCs................................... 38

Figure 7 – Stabilized β-catenin activates Tcf/Lef transcriptional activity in hNSCs. .................. 41

Figure 8 – Stabilized β-catenin does not affect proliferation of hNSCs in vitro. ......................... 43

Figure 9 – Stabilized β-catenin promotes a neuronal cell fate choice during differentiation. ...... 45

Figure 10 – BIO-mediated inhibition of GSK3 activates Tcf/Lef transcription ........................... 48

Figure 11 – BIO-mediated GSK3 inhibition promotes hNSCs to exit the precursor state. .......... 50

Figure 12 – BIO promotes a neuronal cell fate choice in differentiating hNSCs. ........................ 52

Figure 13 – BIO decrease proliferation and induces mild neuronal differentiation in EGF and

FGF. .............................................................................................................................................. 55

Figure 14 – GliNS1 expresses Wnt pathway components and is BIO-responsive in vitro. ......... 58

Figure 15 – BIO reduces GliNS1 precursor marker expression in EGF and FGF. ...................... 60

Figure 16 – BIO treatment induces neuronal differentiation of GliNS1 in EGF and FGF........... 61

Figure 17 – BIO treatment promotes cell cycle exit and decreased proliferation......................... 63

Page 9: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

ix

Abbreviations

AML Acute myeloid leukemia APC Adenomatous polyposis coli BCR-ABL Breakpoint cluster region-Ableson murine leukemiaviral oncogene homologue BDNF Brain-derived neurotrophic factor bHLH Basic helix loop helix BIO (2’Z,3’E)-6-Bromoindirubin-3’-oxime (a GSK3 inhibitor) BMP4 Bone morphogenic protein 4 BrdU Bromodeoxyuridine BTSC Brain tumour stem cell CK1 Casein Kinase 1 CML Chronic myeloid leukemia CNS Central nervous system CSC Cancer stem cell DAPI 4', 6-diamidino-2-phenylindole, DNA intercalater DAPT N-[N-(3,5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester DKK1-4 Dickkopf 1-4 DVL Dishevelled E Embryonic day ECM Extracellular matrix EDTA ethylenediaminetetra-acetic acid EGF Epidermal growth factor EGFR Epidermal growth factor receptor ERBB2 Human epidermal growth factor receptor 2 ES Embryonic Stem cell FAP Familial adenomatous polyposis FBS Fetal Bovine Serum FGF Fibroblast growth factor fgfr1 Fibroblast growth factor receptor 1 Fzd Frizzled receptor GAPDH Glyceraldehyde 3-phosphate dehydrogenase GBM Glioblastoma Multiforme GFAP Glial fibrillary acidic protein Gli Glioma-associated oncogene homologue 1 GSK3α/β Glycogen synthase kinase-3 α isoform/β isoform GTPase guanosine triphosphate hydrolase enzymes Hh Hedgehog HRP Horseradish peroxidase JAK2 Janus kinase 2 JNK c-Jun N-terminal kinase LEF Lymphoid enhancer-binding factor Li+ Lithium LIF Leukemia inhibitory factor LRP5/6 Low density lipoprotein receptor-related protein 5/6 MAP2 Microtubule-associated protein 2 mRNA Messenger ribonucleic acid

Page 10: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

x

MTT 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide NGS Normal goat serum NH2 Amine NICD Notch intracellular domain NOD/SCID Non-obese diabetic/Severe combined immunodeficiency NSC Neural stem cell PBS Phosphate buffered saline PCP Planar Cell Polarity PFA Paraformaldehyde PDGFRA Platelet-derived growth factor receptor A pHH3 Phospho-histone H3 PI3K Phospoinositide 3-kinase PKB Protein Kinase B (also known as AKT) PLO Poly-L-Ornithine PNET Primitive neuroectodermal tumour PSA-NCAM Poly-sialated neural cell adhesion molecule PTCH Patched gene PTEN Phosphatase and tensin homologue PVDF Polyvinylidene fluoride RMS Rostral migratory stream RTK Receptor tyrosine kinase RYK Related to receptor tyrosine kinase SDS sodium dodecyl sulfate SGZ Subgranular zone SL-IC SCID leukemia initiating cell STAT1/3 Signal Transducers and Activators of Transcription-1/3 SVZ Subventricular zone TBST Tris-Buffered Saline Tween-20 TCF T-cell factor TK Thymidine Kinase TS Turcot syndrome WIF Wnt inhibitory factor Wnt Wingless integration-1

Page 11: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

1

Chapter 1

1 Introduction to the Literature

1.1 Introduction to Stem Cells

The homeostatic replenishment and repair of tissues is one of the most fundamental processes in

cellular biology. These processes are due in large part to the persistence of stem cells throughout

an organism’s lifetime. Stem cells are what allow blood, bone, muscle, epithelia, and gametes to

turn over at astonishing rates throughout an organism’s lifetime. For instance, the mammalian

intestinal lining is turned over every two to seven days1. Stem cells are defined by their unique

functional properties: self-renewal—the ability to regenerate itself, and differentiation—their

ability to mature into multiple cell lineages that comprise the tissue in which they reside.2 These

functions are tightly regulated by intrinsic cues as well as extrinsic cues that are found in the

stem cell microenvironment, known as the niche3. There are two major types of stem cells:

pluripotent embryonic stem (ES) cells and multipotent somatic stem cells (embryonic and adult).

1.1.1 Embryonic Stem Cells

The inner cell mass of the developing embryo contains a population of pluripotent cells that can

be characterized in vitro as Embryonic stem (ES) cells. ES cells in vitro are characterized by

their capacity to self-renew indefinitely as well as their ability to differentiate into all the somatic

cell types comprising all three germ layers of the embryo and adult tissues, as well as the germ

cells4. This differentiation profile of ES cells defines the pluripotent state, a less restricted state

than their adult counterparts, which demonstrate tissue-specific multilineage differentiation

potentiality5.

1.1.2 Adult Stem Cells

Adult stem cells are tissue-specific, self-renewing, and exhibit multipotency. These stem cells

can regenerate the various cell types specific to the tissue from which it resides5. These cells are

responsible for replenishing tissues under homeostatic conditions as well as after injury.

Different tissues have varying rates of turnover. Tissues with a relatively slow turnover rate

include skeletal muscle and the brain, while tissues with high turnover rate include the skin, the

intestine, and the blood, changing over every 4 weeks, 3-5 days, and 1 billion cells per day,

Page 12: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

2

respectively6. What mediates the proliferative, self-renewing, and differentiating processes of

these cells are highly complex combinations of extrinsic signals such as soluble growth factors

and hormones, molecules from adjacent cells or in the extracellular matrix, constituting a

complex niche5. In many pathologies these signals are altered, leading to the deregulation of

stem cells and their signature processes. Furthermore, intrinsic alterations, such as genetic

mutations or epigenetic changes can alter the hereditable programs of these cells and have

catastrophic consequences for their resident tissues. Loss of self-renewing capacity can result in

premature aging, senescence or tissue degeneration. On the other hand, genetic and epigenetic

changes in stem cells that result in increased proliferative and/or self-renewing capacity, or

blocks in terminal differentiation capacity, can lead to cancer.

1.2 Neural Stem Cells

Until recently, it was thought that neurogenesis, the process by which new mature and

specialized neurons are generated, did not persist through adulthood. However, in the last two

decades, seminal studies by several groups have shown that, in fact, neural stem cells (NSCs) do

persist throughout adulthood and neurogenesis can and does occur in at least two regions within

the brain: the subventricular zone (SVZ) along the fourth lateral ventricle and the subgranular

zone (SGZ) of the hippocampus7,8,9,10. Although adult NSCs from both the SVZ and the SGZ

generate neurons, they occupy very different niches with unique anatomical structures. The

NSCs from the SVZ, called type B cells by their morphologic criteria on electron microscopy,

are relatively slow cycling and express Nestin, Sox2, and GFAP, reminiscent of embryonic radial

glial cells. B cells give rise to transit amplifying cells (called C cells) that are a rapidly dividing

GFAP-negative population that ultimately gives rise to type A cells, neuroblasts that will migrate

through the rostral migratory stream (RMS) to the olfactory bulb and generate mature neurons. C

type cells are thought to be EGF-responsive and can reacquire stem cell characteristic (observed

in B type cells) in the presence of EGF11. Type A cells express early neuronal markers such as

doublecortin and PSA-NCAM12,13. Within the SGZ, there are thought to be two NSC

populations, one of which (type 1) is more quiescent and expresses Nestin, Sox2 and GFAP,

while the second type (type 2) of NSC cycles more frequently and express Nestin and Sox2, but

not GFAP10,14. The relationship between the two populations remains unknown. In both

neurogenic regions, NSCs are closely associated with the vasculature, adjacent to a plethora of

neighboring cells that can contribute to NSC regulation, and are in close contact with basal

Page 13: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

3

lamina components rich in extracellular matrix (ECM) components such as laminin, enabling

them to respond to local and systemic signals14.

1.2.1 Regulation of NSC activity

A critical question in the neural stem cell field is what regulates stem cell function, particularly

expansion via self-renewal and directed differentiation of specific target cells, for instance

dopaminergic neurons15. Many extrinsic signals that modulate NSC function have been studied

in depth in an effort to unravel this complex molecular network. Epidermal growth factor (EGF)

has been shown to be a potent mitogen, maintaining multipotentiality and promoting the

expansion of NSCs in vitro and in vivo11,16. Fibroblast growth factor-2 (FGF2) has also been

shown to promote NSC proliferation and suppress neurogenesis in vitro. In fact, NSCs are

typically grown in a serum-free defined medium that includes EGF and FGF, as well as other

survival factors17. However, in vivo, FGF2 has also been shown to increase the number of

newborn neurons in the olfactory bulb, suggesting it may play a role in regulating neurogenesis

in the SVZ18. SGZ neurogenesis was reduced as well as a decrease in BrdU+ cells was observed

in the SGZ when fgfr1 was deleted from the CNS, suggesting it could regulate SGZ neuronal

progenitor proliferation19. Other factors have also been shown to mediate neurogenesis. Brain-

derived neurotrophic growth factor (BDNF), for one, is a major positive regulator of

neurogenesis in both the SVZ as well as the SGZ10. The Notch signaling pathway has been

shown to be necessary for the maintenance of NSC self-renewal in vivo20. Furthermore, it is

thought to be a negative regulator of neurogenesis through the inhibition of a variety of basic

helix-loop-helix (bHLH) transcription factors implicated in neuronal differentiation, including

neurogenin1 and 2, Mash1 and Math121. Alternatively, Leukemia inhibitory factor (LIF), a

positive regulator of pluripotency in ES cells, is a potent inducer of astrocytic differentiation

through activation of signal transducers and activators of transcription 1 and 3 (STAT 1 and 3)

signaling, cooperating with a host of chromatin remodeling complexes to bind the GFAP

promoter, activating astrocyte differentiation22. Interestingly, it seems that the Notch signaling

pathway also contributes to gliogenesis by activating STAT3 through the recruitment of Janus

kinase 2 (JAK2), thereby activating STAT323. The Wnt pathway has also been shown to regulate

NSC proliferation and neurogenesis in vitro and in vivo and will be discussed in greater detail

later. Though this list of extrinsic cues is incomplete, it does hint at the complex network of

Page 14: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

4

signals that regulate neural precursor function within its in situ niche as well as in the culture

dish.

1.3 Cancer Stem Cells

While human cancers have been recognized as abnormal tissues with morphologically

heterogeneous populations of cells for over 100 years, it has only been in the last decade that

functional heterogeneity has been demonstrated in a number of cancers24, 25, 26, 27, 28. In the 1950s

and 1960s, an unethical study where human subjects were injected with increasing numbers of

their own explanted cancer cells was published that suggested that the growth potential of cancer

might reside within rare cell populations29. Results from this experiment showed that although

over a million cells were injected, only 50% of these injections developed into palpable nodules,

suggesting that not all tumour cells are capable of initiating growth30. There are at least two

models of cancer growth that can help explain these results31.

1.3.1 Stochastic Model versus the Cancer Stem Cell Hypothesis

The stochastic model assumes that each cell within a tumour has equal capacity to proliferate and

regenerate a tumour, although the probability of any given cell doing so is random and very low.

This model would suggest that prospectively isolating subpopulations of cancer cells on the basis

of functional and phenotypic characteristics would not enrich for tumour-initiating capacity. The

cancer stem cell (CSC) model implies there is functional heterogeneity among tumour cells for

the ability to initiate and maintain tumour growth. In contrast to the stochastic model, one would

then predict that an ability to fractionate subsets of tumour cells based on marker expression or

phenotypic characteristics would define different cell populations with differing capacity to

initiate tumour growth. Tumourigenic capacity would then reside within a subpopulation of the

tumour’s cells and not within the bulk of the tumour mass. The CSC hypothesis also predicts

that treatments targeting the bulk tumour population but sparing CSCs would ultimately fail and

lead to recurrence. This hypothesis has been supported by recent studies that demonstrate that

CSCs are less sensitive to radiation and chemotherapeutic treatments32, 33. Therefore, a key to

further understanding cancer biology lies in a more detailed understanding of the mechanisms of

CSC growth and therapeutic resistance.

Page 15: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

5

CSCs demonstrate the following cardinal functions: tumour initiation, self-renewal, and

differentiation capacity regenerating the phenotypic heterogeneity of the parent tumour34.

Through the development of in vivo functional assays that test tumour-initiating capacity of

discrete cell populations, in conjunction with experimental methods of prospective purification

of tumour cells by surface markers or functional behavior, CSCs have been identified in a

number of human cancers, including blood24, brain25,35, colon27,36, breast26, pancreas37,

melanoma28,38, mesenchymal neoplasms39, neuroblastomas40, and lung41.

Page 16: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

6

Figure 1 – The Stochastic model and the cancer stem cell model.

The stochastic model and the cancer stem cell model. a) The stochastic model implies that

all cells within a tumour are functionally the same and have equal tumourigenic potential.

b) The cancer stem cell model suggests that cells within a tumour exist in a functional

hierarchy where only a subpopulation of cells have tumourigenic potential, while the other

cells are unable to contribute to the tumour bulk. These tumour-initiating cells are termed

cancer stem cells.

Page 17: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

7

1.3.2 Lessons from Leukemia

The first evidence of CSCs came from John Dick’s work in blood system cancers, where a rare

population of CD34+ CD38- cells was isolated from acute myeloid leukemia (AML) patients and

infused back into severe combined immunodeficient (SCID) mice42. This discrete and very rare

population of cells resulted in leukemic blast generation while a more differentiated population

of cells (CD34+ CD38+ and CD34-) did not, thereby identifying the SCIC leukemia-initiating cell

(SL-IC). A subsequent study, published by the same group in 1997, was able to increase

engraftment efficiency by infusing SL-ICs into non-obese diabetes (NOD)/SCID mice24. This

superior repopulation assay allowed them to demonstrate that SL-ICs differentiate in vivo to

reacquire the same leukemic phenotype as seen in the patient while retaining self-renewal

capacity. Furthermore, they were able to show that AML is organized in a hierarchical fashion,

much like the normal hematopoietic system, and that the primitive CD34+ CD38- cell population

(also shown to be the SCID-repopulating cells of the normal human hematopoietic system43) is

the target of transformation. This in vivo model of serial transplantation of cells into NOD/SCID

mice has become the gold standard assay for demonstrating in vivo tumour initiation,

multilineage differentiation potential, and self-renewal capacity of CSCs. By following a similar

logic and experimental method, the identification of CSCs in other blood and solid tumours has

been possible.

1.4 Brain Tumours

Brain tumours refer to a heterogeneous collection of neoplasms that occur in the CNS. The two

most common classes of brain tumours are primitive neuroectodermal tumours (PNETs) and

gliomas44. The most common and aggressive type of PNET is medulloblastoma, a cerebellar

tumour that arises primarily in children, accounting for 20-30% of pediatric tumours.

Astrocytomas, the most common gliomas, are graded on a scale of I-IV, I being benign low

grade and IV being glioblastoma multiforme (GBM), a highly invasive and aggressive tumour

normally found within the cerebral hemisphere, with peak onset at 40 to 70 years of age.

Following treatment, the mean survival time is only 10-12 months due to their inherent

therapeutic resistance and diffuse infiltration of the brain tissue45. Current treatment of both

medulloblastomas and the more lethal malignant gliomas consists of a combination of surgical

resection, radiation and chemotherapy. An additional challenge to treatment is that the blood-

Page 18: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

8

brain barrier restricts access of therapeutic molecules into the brain milieu in order to preserve

the integrity of brain function. Little advancement has been made in the chemotherapeutic

treatment of glioblastomas (grade IV); temozolomide offering only an additional three months of

median survival as the best advance in the past 40 years. This partially explains why it remains

one of the deadliest cancers and why more efforts are necessary to uncover novel drugs that

effectively target the cancer cells.

1.4.1 Identification of Brain Tumour Stem Cells

In 2003, Al-Hajj et al. isolated cells from fresh human breast cancer cells based on the cell

surface marker expression profile CD44+ CD24−/low Lineage− that exhibited potent tumour-

initiating ability when as few as 1 x 103 cells were injected into the mammary fat pad of

NOD/SCID mice26. These cells could be serially transplanted, demonstrating their self-renewal

and extensive proliferative capacity while also recapitulating phenotypic copies of the patient

tumour, all of which are hallmarks of CSCs. This was the first time CSCs were prospectively

isolated in a solid tumour.

In 2000, a study by Uchida et al. showed that multipotent and self-renewing human neural stem

cells could be prospectively isolated and purified based on CD133+ expression46. Under the

premise that brain-tumour initiating cells could originate from a normal stem or progenitor cell,

the Dirks laboratory identified brain-tumour stem cells (BTSCs) in adult glioblastomas and

childhood medulloblastomas based on the cell surface marker CD133+ 25,35. Having initially

identified CD133+ cells as a clonogenic and multipotent population in vitro, they then showed

that CD133+ were the tumour-initiating population in vivo, generating a tumour with as few as 1

x 102 cells, and could self-renew in vivo, as was demonstrated by serial transplantation of

CD133+ cells into the brains of NOD/SCID mice. Several groups, including our own, have since

reported that CD133+ does not universally identify a tumour-initiating population, but it remains

the most reliable marker available47,48. Recently, a study by Fine’s group demonstrated that up to

60% of freshly isolated glioblastomas do not express CD133, but express SSEA-1 (CD15),

which also identifies a tumourigenic subpopulation when CD133 is not expressed. Thus, an

additional marker of BTSCs is SSEA-1, which enriches for self-renewing and multipotent

tumour-initiating cells that could give rise to SSEA-1- as well as SSEA-1+ cells, establishing a

cellular hierarchy49. Needless to say, more efforts are needed to identify marker signatures that

Page 19: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

9

will define BTSC populations. Moreover, it is likely that more than one putative BTSC

population will exist from patient to patient as different brain tumours may have originated from

different cell populations or have undergone different transforming events that will lead to

distinct cell surface profiles.

1.4.2 Intrinsic regulators of Brain Tumours and BTSCs

Cancer has long been considered a disease of genomic alterations: DNA sequence mutations,

copy number changes, chromosomal rearrangements and various other aberrations have all been

shown to contribute to the development and maintenance of many human cancers50. Genome-

wide profiling studies of human glioblastoma confirm decades’ worth of findings that several

core molecular pathways, namely the p53, pRB, p16Ink4/p19Arf and receptor tyrosine kinase

(RTK) pathways, are the main contributors in gliomagenesis and disease progression50,51. The

p53 tumour suppressor—a transcription factor that regulates cell cycle progression and apoptosis

in response to various insults—is mutated or lost in over 60% of sporadic astrocytomas52.

Furthermore, patients with Li-Fraumeni syndrome—a syndrome characterized by the germ-line

mutation of the TP53 gene rendering them susceptible to cancers—are predisposed to various

brain tumours including gliomas53.

PTEN—phosphotase and tensin homologue—is a major inhibitor of the pro-growth PI3K/AKT

pathway and is mutated or lost in over 30% of primary glioblastomas (GBMs)50. Moreover, in

human GBMs harbouring TP53 mutations, 60% of these tumours had concomitant PTEN

alterations including homozygous deletion54. In the same study, it was found that over 60% of

primary GBMs showed loss of heterozygosity (LOH) of chromosome10q, where the PTEN gene

is located. In a genetic study that examined the roles of p53 and Pten in neural and glioma stem

cells, p53-/- pten+/- fetal NSC and tumourspheres showed increased proliferation and resistance to

differentiation cues55. They also showed that p53 and Pten cooperate to regulate Myc—a

transcription factor known to play a role in cell cycle progression and stem cell self-renewal—

showing that elevated levels of Myc impeded stem cell differentiation in NSCs and

tumourspheres, indicating a contribution of p53 and Pten in BTSC regulation. Over 50% of

human GBMs lack a functional Ink4a/Arf locus, thereby altering the p53 and pRb pathways,

both of which have known roles in cell cycle progression and stem cell regulation50,56.

Page 20: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

10

Along with the loss of key tumour suppressors, various RTKs, including EGFR, ERBB2, and

PDGFRA are amplified in 45%, 8%, and 13%, respectively, indicating a role for these pro-

proliferative pathways in gliomagenesis. Altogether, the core pathways that harbour the majority

of genetic abnormalities found in human GBMs are pathways shown to regulate cell cycle

progression and stem cell functions in normal NSCs11,57,58. This fact highlights the importance of

studying these pathways within BTSCs in order to gain insight into tumour initiation and

propagation.

1.4.3 Extrinsic Regulation of Brain Tumours and BTSCs

It is well established that the tumour microenvironment can regulate initiation and maintenance

of the malignancy through extrinsic cues, including a variety of developmental signaling

pathways59. Individuals with germ-line alterations in the PTCH (patched) gene, a member of the

hedgehog signaling pathway, are prone to the development of basal cell carcinoma as well as a

medulloblastomas, a cerebellar malignancy60. Furthermore, studies have shown that blocking

hedgehog signaling with an inhibitor known as cyclopamine can halt proliferation and

clonogenicity of stem-like cells from GBMs, as well as prevent tumour growth in vivo61. The

Bone Morphogenic Protein signaling pathway may also be important for GBMs. In a study by

Piccirillo et al., transient exposure to BMP4 was shown to deplete the BTSC population in vitro

by promoting astrocytic differentiation as well as inhibit tumour initiation in vivo62. Notch

signaling may also be important. Pharmacological inhibition of the γ-secretase complex Notch

processing pathway—necessary for proper signal transduction—has been shown to block

proliferation and decrease the number of CD133+ cells in medulloblastomas63. In another study,

Purow et al. showed that primary glioma tissues overexpress the Notch1 intracellular domain

(NICD), indicating pathway activation, and that down-regulation of Notch1 or its ligand Delta-

like-1 leads to increased apoptosis and decreased proliferation in vitro, as well as prolonged

survival in a murine orthotopic brain tumour model64. Wnt signaling, another developmental

pathway implicated in a number of human cancers, has also been shown to play a role in brain

tumour initiation and maintenance but has not been explored in as much detail as the above

pathways65. This will be discussed in further detail later. Therefore, as is the case with normal

NSCs that are regulated by the extrinsic cues within their niche, so too are BTSCs, which receive

signals from their microenvironment that regulate their ability to self-renew and proliferate,

thereby contributing to tumour initiation and maintenance66.

Page 21: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

11

1.5 Introduction to Wnt Signaling

Multicellular organisms require the precise orchestration of a variety of signaling pathways to

govern the processes necessary for proper development: proliferation, differentiation, cell fate

decisions, and survival67,68. The Wingless-int (Wnt) signaling pathway regulates a large number

of adult and developmental processes, primarily by modulating gene transcription through

several signal transductions69. Wnts, for which mammals have 20, are cysteine-rich secreted

glycoproteins that signal through one of ten Frizzled (Fz) receptors, which are seven-pass

transmembrane proteins with an extracellular N-terminal cysteine-rich domain70. The Wnt-Fz

complex recruits the scaffolding protein Dishevelled (Dvl), bringing together downstream

pathway components for signal transduction71. Downstream of Dvl, the pathway splits into at

least three known branches: the planar cell polarity (PCP) branch, the Wnt/calcium (Wnt/Ca2+)

pathway, and the canonical or Wnt/β-catenin-mediated pathway72. The PCP pathway is mediated

by small GTPases (Rho and Rac1) and the c-jun amino (N)-terminal kinase (JNK), and has been

shown to control cell polarity and orientation within a tissue as well as convergent extension

movements during gastrulation73. In the Wnt/Ca2+, signal transduction through Dvl induces

calcium influx, activating protein kinase C (PKC) and calcium/calmodulin dependent protein

kinase II (CaMKII), and has been shown to play a role in cell migration during gastrulation and

cardiac development72. The most well-characterized or canonical Wnt signaling pathway

involves signaling through the β-catenin and the GSK3-Axin-APC protein complex and

transcriptional activation mediated by Tcf/Lef transcription factors at specific targets.

1.5.1 The Canonical Wnt Signaling Pathway

The canonical pathway—the only Wnt pathway referred to from here in—is mediated by β-

catenin, a cytoplasmic protein whose stability is regulated by the destruction complex72. The

destruction complex comprises several key players: Axin, adenomatous polyposis coli (APC),

glycogen synthase kinase 3 (GSK3), and casein kinase 1 (CK1). In the absence of Wnt

stimulation, β-catenin is phosphorylated by CK1 at serine 45 (S45), which primes it for further

phosphorylation by GSK3 at threonine 41 (T41), S37 and S33, targeting β-catenin for

ubiquitination and subsequent degradation74. However, when Wnt engages its Fz receptor, Dvl is

recruited to the cytoplasmic tail of Fz and thought to recruit Axin and the other complex

components with it, allowing β-catenin to evade phosphorylation and degradation70.

Page 22: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

12

Unphosphorylated stabilized β-catenin is free to translocate to the nucleus where it acts as a

transcriptional co-activator by interacting with a group of transcription factors known as T-cell

Factors/Lymphoid Enhancer Factors (TCF/LEFs) (and various chromatin remodeling proteins)

that together transcribe target genes75.

1.5.2 Regulatory mechanisms of the Canonical Wnt Pathway

Despite the simple picture painted above, the Wnt pathway is extremely complex. For one, along

with the 20 mammalian Wnt ligands, there are 10 mammalian Fz receptors, as well as several

other receptors (e.g. low-density lipoprotein receptor-related protein 5/6—LRP5/6, RYK, Ror2)

that are known to initiate one of three Wnt signaling cascades either in conjunction with Fz

receptors, or on their own72. Not all Wnt-Fz combinations are well understood. Some ligands

have been denoted as “canonical” because they were capable of inducing axis duplication in the

Xenopus following injection of Wnt mRNAs, including Wnt1, Wnt3a, Wnt5a, Wnt7a, Wnt8a,

and Wnt8b76. However, new studies suggest that signal transmission initiated by any of these

ligands may be highly context-dependent and in some cases may act to inhibit TCF/LEF-

mediated transcription77. Wnt inhibitors are very important in regulating signal transmission

throughout development and various cell processes. Inhibitory molecules such as Dickkopf1-4

(Dkk1-4), Wnt inhibitory factors (WIFs), and soluble frizzled-related proteins (sFRPs) act as

Wnt pathway antagonists by either binding directly to the Fz-receptor complex to block Wnt

from binding (i.e. Dkk1), or by binding directly to Wnt ligands (WIFs and sFRPs) through their

cysteine-rich domains, which these inhibitors also possess 78. Thus, at the cell surface level

alone, there is a great deal of complexity in regulating the appropriate signal transduction.

As a means of mediating transcriptional output upon stimulation, the Wnt signaling pathway has

built-in feedback loops that regulate the pathway at several key points. A prime example is the

classic Wnt target genes Axin2 or DKK1. Both proteins are negative regulators of the signaling

pathway and are actively transcribed when TCF/LEF-mediated transcription is on. The negative

feedback loop helps to stabilize β-catenin directly (i.e. Axin2) or indirectly by interfering with

ligand-receptor binding (i.e. Dkk1)70. This complex network of regulatory mechanisms is crucial

for mitigating the risk implicated in a signaling network that has such a broad impact on

fundamental cellular processes throughout an organism’s life. Any deregulation of the pathway

Page 23: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

13

will have profound impacts on the feedback loop, and could in turn perpetuate any resulting

phenotype.

Further downstream, there are internal regulators that modulate β-catenin stability through direct

or indirect interaction with various components of the destruction complex. The most notable

interactions involve GSK3, a key negative regulator of β-catenin and central moderator of the

canonical signaling pathway as a whole. However, GSK3 is a downstream switch for many

signaling pathways including Wnt, growth factors, insulin, RTKs, hedgehog, G-protein coupled

receptors (GPCRs) and is involved in almost every cell function from metabolic regulation, cell

development, cell cycle regulation, gene transcription, cell proliferation and apoptosis79.

1.5.3 GSK3: the master of multitasking

Glycogen synthase kinase-3 (GSK3) is a multifaceted kinase that functions in several distinct

pathways. Originally identified for its phosphorylation of glycogen synthase, the rate-limiting

enzyme of glycogen metabolism, it has since been shown to target over 50 substrates in the

RTK—PI3K, Hh, Notch, and Wnt pathways80. In the Drosophila, the loss of GSK3

(Shaggy/Zeste-white3) results in the accumulation of the full-length active form of Cubitus

interruptus (Ci), the fly orthologue of the Gli3 transcription factor, and the subsequent ectopic

expression of Hh-responsive genes81. Thus, it is thought that GSK3 regulates the phosphorylation

and ensuing proteolytic degradation of Ci. Espinosa et al. found that GSK3β also regulated the

Notch signaling pathway in vitro and in vivo through the phosphorylation of the Notch2 receptor

intracellular domain82. Furthermore, they found that GSK3β inhibition by Wnt1 or its

pharmacological inhibition using LiCl abrogated Notch2 phosphorylation and increased Hes1-

reporter activity, Hes1 being a target gene of Notch signaling. Together this suggests that GSK3β

is a negative regulator of the Notch pathway, and may crosstalk with the Wnt pathway.

Two isoforms, GSK3α and GSK3β, are present in mammals83. While GSK3α (52 kDa) is 5 kDa

larger than GSK3β due to an amino-terminal glycine rich extension of unknown function, there

is tremendous functional overlap between the two isoforms in most instances, as was observed in

the GSK3β knockout mouse84,85. Upon analysis of the GSK3β knockout mouse model, it was

observed that one unique function of GSK3β is its necessity for the proper nuclear function of

NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) in response to TNFα

(Tumour Necrosis Factor-α)-induced apoptosis in hepatocytes. Interestingly, there was no

Page 24: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

14

evidence that Wnt signaling or glucose metabolism was disturbed, suggesting that GSK3α is able

to compensate for the loss of GSK3β in these pathways. However, O’Brien et al. performed a

study comparing Gsk3β+/- mice with lithium-treated mice and found that while Gsk3α protein

levels were not significantly increased in Gsk3β+/- mice brains, there was an increase in

stabilized β-catenin, similar to levels observed in Li+ mice brains86. Therefore, at least in the

brain, the loss of one copy of the Gsk3β allele resulted in the increased stabilization of β-catenin,

suggesting a possible slight preference for GSK3β in the brain. Doble et al. also analyzed the

functional redundancy of GSK3α and GSK3β in Wnt/β-catenin signaling in ES cell lines87. ES

cell lines were generated with 0-4 functional GSK3 alleles and their isoform-specific function in

the canonical Wnt pathway was examined. It was found that GSK3 protein levels did not

increase when either GSK3α or GSK3β was absent, and TCF—mediated transcription was

normal. Only when cells lacked 3 or 4 alleles was gene dosage reflected in TCF activity.

Furthermore, ES cells lacking 3 or 4 of the GSK3 alleles showed impaired differentiation,

particularly down the neuroectoderm lineage, which was restored upon reintroduction of

functional GSK3. While this study validates the redundancy of GSK3α and GSK3β as regulators

of Wnt/β-catenin signaling, as well as their role in ES cell regulation, it does not determine

whether the observed phenotype is Wnt/β-catenin—dependent. This could be addressed by

interfering with β-catenin—mediated signaling downstream of GSK3 to see whether, similar to

with GSK3 re-expression, the neuroectodermal differentiation capacity of GSK3α-/- /β-/- cells can

be rescued.

One peculiar feature of GSK3 is that unlike most kinases, it is highly active in resting cells and is

regulated by inhibition in response to various cellular signals. Such signals include hormone and

growth-factor activation of RTKs leading to the activation of PI3K—mediated activation of PBK

(also known as Akt), and Wnt-induced inactivation of GSK3 via Dishevelled (Dsh), the

mechanism of which is not entirely clear80. Insulin, among other hormone and growth factors,

bind to RTKs at the cell membrane leading to the activation of PI3K, which recruits and

activates the prosurvival factor PKB88. Subsequently, PBK phosphorylates GSK3 on Ser21 (on

the α isoform) or Ser9 (one the β isoform) and inactivates it, resulting in the primary mechanism

of growth-factor inhibition of GSK389. Surprisingly, PKB—mediated inactivation of GSK3 is

not associated with an increase in TCF/LEF transcriptional activity, maintaining a distinction

between the PI3K/PKB pathway and the canonical Wnt signaling pathway90. The inactivation of

Page 25: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

15

GSK3α/β in vivo also requires dephosphorylation of tyr 279/216, respectively, in response to

extracellular signaling91. Unlike most protein kinases that rely on sequence recognition, many

target substrates of GSK3 require a priming phosphorylation event by another kinase in order to

generate a recognition site. The primed molecule can then interact with Arg96 (on the GSK3β

isoform), which is proximal to the substrate-binding site, and bind to GSK392. When GSK3 is

inactivated by hormone and growth factor inhibiting phosphorylation on Ser9 at the amino

terminus (or Ser21 on the GSK3α isoform), the NH2-terminal domain bends back and binds to

Arg96, blocking the substrate-binding site93. Therefore, GSK3 would not be capable of binding

phosphoprimed substrates that rely on Arg96 binding but would be able to target proteins that are

independent of this mechanism, such as β-catenin.

GSK3-mediated phosphorylation is a crucial event leading to the subsequent ubiquitination and

degradation of β-catenin85. It has been shown by several groups that Axin, β-catenin and GSK3

exist in a ternary structure and that Axin promotes GSK3-dependent phosphorylation of β-

catenin94,95. Only Axin-associated GSK3 can display activity toward β-catenin while non-

associated GSK3 showed no activity toward β-catenin and is irrelevant in the Wnt pathway96.

Moreover, Axin shows no preference between either GSK3 isoform, and readily binds to either80.

However, it remains unknown how Axin acts to commandeer an entire pool of Wnt-responsive

GSK3 and whether (and if so, how) Axin acts to shield GSK3 from other regulatory interactions

(i.e. PKB phosphorylation). Axin is not the only molecule affecting GSK3 activity toward β-

catenin. Although the mechanism remains unknown, FRAT (frequently rearranged during

advanced T-cell lymphomas), also known as GBP (GSK3-binding protein), binds to GSK3 in a

mutually exclusive manner with Axin and negatively regulates GSK3 activity toward β-catenin97.

Also, DISC1 (Disrupted in Schizophrenia 1) inhibits GSK3β activity specifically toward β-

catenin through direct interaction, resulting in stability of β-catenin and increased target gene

expression in murine neural precursors98. Therefore, direct interaction with GSK3 is one of the

primary means of regulating its kinase activity toward β-catenin.

Lithium (Li+), long used in the treatment of bipolar disorder, has been shown to inhibit GSK3

kinase activity in a non-competitive manner in vitro and in vivo99,100. The effect is reasonably

specific in that no other kinase is affected but it does interfere with other processes and enzymes,

including IMPase (inositol monophosphatase)99. In corroboration with independent genetic

studies, many GSK3 targets have been identified using Li+ and other chemical inhibitors of

Page 26: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

16

GSK3, including cyclin D101. BIO ((2’Z,3’E)-6-Bromoindirubin-3’-oxime), the synthetic

derivative of 6-bromoindirubin, was also found to be a potent competitive inhibitor of GSK3

activity and has been shown to mimic the canonical Wnt pathway by maintaining self-renewal

and pluripotency of mouse and human ES cells102. Furthermore, Lluis et al. found that activation

of β-catenin through BIO-mediated GSK3 inhibition increases the efficiency with which somatic

cell reprogramming occurs mediated by cell fusion of several types of hybrids103. However, it

was found that concentrations above 0.005 µM are no longer specific to Gsk3α/β kinase activity

alone and can inhibit the closely related CDK1/CyclinB, CDK2/CyclinA, and CDK5/p35 at

concentrations below 1µM104. Also, while it has yet to be documented in the literature, it is likely

that BIO interferes with other signaling pathways such as Hh, Notch, and RTK-PI3K, given that

GSK3 plays a mediating role in all of them. Therefore, it is important to consider other possible

direct targets and downsteam effects of BIO-mediated GSK3 inhibition other than the canonical

Wnt signaling pathway. In any event, GSK3 remains a central mediator of many critical

pathways, and understanding how this kinase can be modulated genetically and chemically will

likely be crucial for understanding the molecular basis of various developmental processes and

diseases.

Page 27: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

17

Figure 2 – The canonical Wnt Signaling Pathway.

A schema of canonical Wnt signaling. a) The signaling pathway is inactive due to the lack

of Wnt ligand. β-catenin is restrained in the Axin/APC/GSK3 destruction complex and

targeted for serial phosphorylation by GSK3 and CK1 on its amino terminal.

Phoosphorylated β-catenin is degraded. TCF/LEF complexes remain in its repressed state.

b) When Wnt ligands bind to the Frizzled-Lrp5/6 receptors on the cell surface, the

destruction complex is recruited to the inner cell membrane and β-catenin can evade

phosphorylation, allowing it to stabilize and accumulate in the cytosol. β-catenin then

translocates to the nucleus and activates TCF/LEF—mediated transcription of target

genes, such as neurogenin1.

Page 28: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

18

1.6 Wnt Signaling in Development and Stem Cells

The canonical Wnt pathway has been shown to be crucial throughout development (Table 1).

Embryos with a null mutation for β-catenin show gastrulation defects such as failure to produce

an A-P (anterior-posterior) Axis or to form mesoderm by E7 that result in embryonic lethality78.

Embryos with constitutive activation of β-catenin seems to result in premature epithelial to

mesenchymal transition in the epiblast105. Furthermore, culturing ES cells in vitro while

maintaining pluripotency seems to require GSK3 inhibition, which can be accomplished by

several small molecule inhibitors106. In tissue-specific development, Wnt has also been shown to

be a crucial pathway. Wnt signaling seems to be important in maintaining self-renewal and

expansion of cardiac progenitors and subsequently needs to be repressed in order for

differentiation of cardiomyocytes or smooth muscle cells to take place107. In the skin, Tcf3 is

exclusively expressed in bulge stem cells and is thought to maintain stem cell activity through

transcriptional repression of Wnt target genes108. Cells destined to become transit-amplifying

cells and differentiate to become hair switch from a Tcf3- to a Lef1-mediated transcriptional

program, resulting in the terminal differentiation of bulge stem cells to hair cells109. In the

intestine, Wnt signaling seems to be the predominant force controlling cell fate along the crypt-

villus axis as nuclear β-catenin is observed at the bottom of the crypts where the stem cells are

thought to reside70. In Tcf4-/- mice, the differentiated epithelial compartments of the neonatal

intestine appear normal, however the crypt progenitor pool is ablated110. A similar phenotype is

also exhibited in mice with transgenic expression of the inhibitor Dkk185. Also, the Wnt target

gene, Lrg5, has been shown to mark the crypt stem cell111. TCF reporters have also been shown

to be active in hematopoietic stem cells (HSCs)70. In a recent study by Zhao et al., β-catenin-

deleted HSCs showed decreased long-term self-renewing capacity upon transplantation into

recipient mice but did not affect lineage differentiation112. Interestingly, an earlier study showed

that deleting β-catenin using an interferon-based method showed no decrease in self-renewal or

reconstitution capacity in vivo113. They hypothesized that γ-catenin (plakoglobin) could be

playing a redundant role in HSCs. However, Koch et al. deleted both β- and γ-catenin in HSCs

and found no impairment of self-renewal in a primary reconstitution assay114. This series of

experiments highlights the controversy and the complexity that remains to be unraveled in the

blood field.

Page 29: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

19

1.6.1 Wnt in Neural Development and NSCs

Not surprisingly, Wnt signaling also plays a crucial role in A-P axis formation of the neural tube

through the specification of cell fates78. Various lines of evidence suggest that Wnts have a

posteriorizing role during head formation while inhibiting Wnt, particularly by Dkk1, allows for

anterior neural structure generation115. Various structures within the embryonic cerebral cortex

have been shown to require the activity of specific ligands for proper development. Most

notably, Wnt3a is required at E10.5 to regulate proliferative expansion of a hippocampal

progenitor pool that leads to normal hippocampal development116. In the adult hippocampus, it

has been shown that secreted Wnt3 regulates hippocampal neurogenesis in vitro and in vivo by

promoting the proliferation of neuronal precursors and their subsequent differentiation117. Wnt7a

was also shown to promote neuronal differentiation of E11.5 cortical neural progenitors in vitro,

even in the presence of FGF-2, a potent mitogen118. However, another study by Israsena et al.

suggests that the status of FGF-2 determines whether β-catenin-mediated signaling promotes

proliferation or neuronal differentiation of NSCs119. They propose that while NSCs are in the

SVZ, they receive FGF-2 stimulation, promoting renewal and expansion. However, when these

cells migrate into the cortex and away from the SVZ, they no longer receive FGF-2 signals, and

the β-catenin-mediated pathway promotes a neuronal cell fate. This theory may also help to

explain findings by Chenn and Walsh, who found that when an activated form of β-catenin was

overexpressed in nestin-expressing cells there was an expansion of cells resulting in an

overgrowth of the cerebral cortex, which was thought to be due to increased reentry into the cell

cycle, 120. However, in another study by Ivaniutsin et al. that examined the role of APC in the

developing cerebral cortex, different results were observed with respect to the regulation of NSC

proliferation121. Whereas Chenn and Walsh expressed stabilized β-catenin in neural precursors

prior to commitment to the cerebral cortical fate (~E9.5), APC is deleted under the Emx1

promoter (~E9.5), a promoter that is active in both proliferating and differentiated cortical

neurons122. When APC is deleted under the Emx1 compartment, there is a decrease in the size of

the precursor pool, which seems to be the result of increased apoptosis and a decrease in the

number of cycling cells as indicated by bromodeoxyuridine (BrdU) uptake. Furthermore, loss of

APC resulted in premature neuronal differentiation and caused cerebral cortical cells to adapt

fates typically associated with more dorsal-posterior regions of the CNS. Therefore, while both

studies use models that result in increased β-catenin-mediated signaling, the spatiotemporal

Page 30: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

20

regulation of β-catenin-mediated signaling is critical for neural cell fate decisions and may differ

dramatically in a cell-autonomous manner. Along the same lines, it is possible that Wnt signaling

may have differing roles at various levels within the functional hierarchy, for example self-

renewal of NSCs as well as neuronal differentiation of more committed progenitors. Until the

hierarchy can be delineated more effectively, these questions remain unanswered. Further

investigation is required to truly understand the effects of β-catenin-mediated signaling on NSC

function in vitro and in vivo.

1.7 Wnt Signaling in Cancer and CSCs

1.7.1 Wnt in Colorectal Cancers

Given the integral roles that Wnt signaling plays in development as well as homeostasis, it is no

surprise that the canonical pathway has been linked to many human cancers, particularly of the

epithelial variety. For example, around 85% of all sporadic and hereditary colorectal tumours

show loss of APC function123. Of the tumours with wild-type APC, 15% show point mutations

on key serine/threonine residues in the amino-terminus of β-catenin, thought to be putative

targets of GSK3 phosphorylation124. Since Tcf4 is crucial for stem cell maintenance in the crypt,

it is likely that stable activation of the β-catenin/Tcf4 pathway causes the expansion of the crypt

compartment, which is seen in early stages of colorectal tumourigenesis125. Also, the hereditary

disease known as familial adenomatous polyposis (FAP) that eventually leads to colorectal

cancer in inflicted individuals is caused by a loss of function mutation in the APC gene124. This

is similar to another disease known as Turcot’s Syndrome (TS), discussed later, in which a

germline mutation in the APC gene leads to colorectal cancer and, infrequently, to primary brain

tumours126.

1.7.2 Wnt in Skin Cancers

In the context of normal skin homeostasis, TCF3-mediated repression of canonical Wnt signaling

is necessary to maintain bulge stem cell activity, while an active LEF1-mediated pathway seems

to promote terminal differentiation127. In human metastatic melanoma as well as a murine model

(B16), it was observed that activated β-catenin signaling correlated with decreased proliferation

and smaller tumour volume128. Furthermore, the study showed that inducing Wnt signaling up-

regulates markers of melanocyte differentiation. Therefore, it seems the loss of Wnt signaling

Page 31: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

21

may be critical for melanoma progression and metastasis. In squamous cell carcinoma (SCC),

however, Malanchi et al. reported the identification of a murine CSC population based on the

expression of CD34 and the exclusion of various lineage markers129. They also report that

cutaneous CSC maintenance is dependent on continuous β-catenin signaling since when β-

catenin was genetically ablated, there was a marked loss of the CD34+ population and CSCs lost

the ability to initiate secondary tumours due to terminal differentiation, indicating a loss of self-

renewal properties. Therefore, in another type of skin cancer, it seems that β-catenin mediated

signaling induces the opposite effect as compared to normal circumstances, signifying the highly

context-dependent nature of Wnt signaling.

1.7.3 Wnt in Leukemias

Wnt has also been shown to be deregulated in various leukemias130. In acute myeloid leukemia

(AML), it has been shown that important Wnt pathway antagonists (sFRPs and DKKs) are

downregulated due to promoter methylation and silencing, potentialliy contributing to the

pathogenesis of AML131. The activation of Wnt/β-catenin signaling has also been implicated in

maintaining chronic myeloid leukemia (CML) stem cell function112. This was demonstrated

using the BCR-ABL leukemia mouse model and crossed into conditionally deleted β-catenin-/-

mice. Results of the study showed that self-renewal of CML stem cells were impaired in BCR-

ABL/β-catenin-/- mice compared to control BCR-ABL mice, as was shown by serial

transplantation assay. It was shown that BCR-ABL protein levels were significantly decreased in

β-catenin-/- mice, but that this observation was specific to BCR-ABL CML and not acute

lymphoid leukemia (ALL). In another study, it was observed that blast crisis (BC) CML stem

cells express a misspliced variant of GSK3β—a negative regulator of β-catenin—that resulted in

the loss of its kinase domain and the activation of canonical Wnt signaling132. Moreover, BC

CML stem cells that expressed this splice variant exhibited enhanced leukemic engraftment that

could be rescued by reintroducing full-length GSK3β. Further investigation is necessary to

uncover novel therapeutic targets specifically within the Wnt pathway.

Page 32: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

22

1.8 Wnt in Brain Tumours

1.8.1 Wnt in Medulloblastomas

The dysregulation of Wnt signaling has been well documented in medulloblastomas. Turcot’s

syndrome, a familial cancer syndrome in which patients have a germline mutation in the APC

gene, results in the development of colonic polyps and has also been linked to the development

of primary brain tumours, most of which are medulloblastomas133. Many studies have also

documented mutations in the genes of key components of the Wnt signaling pathway, including

APC, CTNNB1 (the gene for β-catenin), CDH1 (the gene for the adhesion molecule E-cadherin),

and GSK-3β loss of heterozygosity in sporadic medulloblastomas134. Therefore, evidence

suggests that activated Wnt signaling is implicated in the pathogenesis of at least a subset of

medulloblastomas. Interestingly, however, a study conducted in Britain found that children that

presented with medulloblastomas that were immunoreactive for nuclear β-catenin had a

significantly higher overall (OS) and event-free (EFS) survival compared to children who did not

show nuclear immunoreactivity to β-catenin, and thus β-catenin status can actually be used as a

prognostic marker135,136. While it is clear that the Wnt/β-catenin pathway has been directly linked

to medulloblastomas oncogenesis, it is unknown whether it plays a role as a driving

transformation event in tumour initiation, or whether its dysregulation is a secondary event that

is associated with and contributes to disease progression.

1.8.2 Wnt in Glioblastoma Multiforme

In gliomas, specifically glioblastoma multiforme (GBM), less is known about the role of Wnt in

tumourigenesis, however, a few studies using serum-derived lines have attempted to address this

broad question. In one study it was observed that sFRPs, a group of secreted Wnt inhibitors,

slowed the motility of glioma cells while enhancing proliferation and in vivo tumourigenicity137.

Surprisingly, while β-catenin levels were not altered by sFRP-2-mediated inhibition, tyrosine

phosphorylation, which is thought to regulate β-catenin’s association with cadherins, was

significantly decreased and may help to explain the observed phenotype. A separate study by Pu

et al. observed that knockdown of Wnt2 as well as β-catenin induced apoptosis and decreased

cell growth in vitro and in vivo138. They also observed an associated decrease in PI3K/Akt

activity in Wnt2 and β-catenin knockdown lines, which may help to explain the observed

decrease in proliferation. Overexpression of Wnt5a, thought primarily to be a canonical ligand,

Page 33: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

23

has also been shown promote proliferation of glioma cells in vitro, while in vivo, Wnt5a-

knockdown results in decreased tumour volume upon orthotopic transplantation in comparison to

the control139. Early studies investigating the role of Wnt1 in the development of the CNS

revealed that Wnt1 regulates proliferation of mid/hindbrain precursor cells, and that

overexpression of Wnt1 causes an expansion of the hindbrain region140,141. Although no reports

implicate Wnt1 in gliomagenesis as of yet, it is conceivable that its mitogenic effects may

contribute to neoplastic progression.

While various secreted proteins have been implicated in gliomas, another study looked at

GSK3β, a negative regulator of β-catenin as well as a point of convergence for several signaling

transduction pathways known to modulate cell proliferation (eg. FGF-1, Insulin, PI3K/Akt)142.

When treated with various GSK3 inhibitors, tumour cells showed decreased expression of neural

precursor markers (Sox2 and Nestin) and increased expression markers associated with glial and

neuronal differentiation (GFAP and β-III-tubulin, respectively). In contrast to the previous

findings, this study suggests that an activation of the canonical Wnt signaling pathway through

inactivation of a key modulator—GSK3β—results in growth arrest and increased terminal

differentiation. It is clear from these confounding results that further investigation of Wnt

signaling in gliomas is warranted to truly appreciate its affect on tumourigenesis and

maintenance.

1.8.3 Brain tumour stem cells in vitro

A caveat of the above studies is that experiments were carried out using serum-derived GBM

lines. It has become increasingly clear, however, that traditional serum-derived GBM lines to do

not recapitulate the genotype and phenotype of the original human GBM, making it a poor in

vitro model for drug discovery and preclinical testing17. GBM lines established in defined serum-

free conditions with bFGF and EGF exhibit consistent growth kinetics, retain similar

morphology to NSCs, express known immature markers (Sox2 and Nestin), retain differentiation

capacity, and retained in vivo tumourigenic and self-renewing properties through serial

transplantation, irrespective of passage number. In contrast, the serum-derived lines varied in

growth kinetics depending on the passage number, altered their morphology to resemble

fibroblast-like cells, lost NSC marker expression and showed varied response to differentiation

cues. Also, serum-derived lines were only tumourigenic at late passages, when their proliferation

Page 34: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

24

rate seemed to increase exponentially. When gene expression and cluster analysis were

performed for serum-free, serum-derived, NSCs and parent GBM tissue, results revealed that

gene expression of serum-derived GBM lines did not reflect the gene expression pattern of the

original GBM, nor did it cluster with serum-free GBM lines or NSCs. On the other hand, serum-

free GBM lines retained similar gene expression patterns to the original GBM from which it

originated, regardless of passage, and it clustered with other fresh GBMs, and NSCs143. These

observations reveal that results from studies using serum-derived GBM lines may not report a

biologically relevant phenomenon. What this also suggests is that very little is known about the

role of Wnt on the tumour-initiating population (i.e. BTSCs), which are thought to be the cells

driving the tumour’s progression. Thus, it is important to carry out future investigations in a

culture system that more accurately reflects the in vivo characteristics of human GBMS, such as

serum-free adherent GBM lines144.

Page 35: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

25

Table 1

Page 36: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

26

1.9 Thesis Rationale and Aim

One view of cancer is that it may represent aberrant organogenesis. In the last ten years, there

has been a large amount of evidence suggesting that tumours are arranged in a functionally

hierarchical fashion, with a subpopulation of tumour stem cells driving tumour initiation and

maintenance, much like adult stem cells within normal tissues. Many developmentally

significant pathways, including the canonical Wnt pathway, have been implicated in cancer

development. Thus it is necessary to gain a firm understanding of the molecular events

governing normal development in order to appreciate what is perturbed in tumour initiation and

progression. There remain many gaps in our understanding of canonical Wnt signaling in human

NSCs. Our goal has been to determine the effect of β-catenin-mediated Wnt signaling on normal

human NSC proliferation, survival, and lineage specification in vitro. Additionally, and in

parallel, we also sought to determine whether Wntsignalingmayplayaroleinregulating

cancer stem cells isolated from human glioblastomas (GBMs). Our ultimate goal is to determine

how targeting the canonical Wnt signaling pathway in human brain tumors may affect the

proliferation and/or differentiation of brain tumour stem cells.

1.10 Thesis Hypothesis

Canonical Wnt signaling plays a role in lineage specification of normal human NSCs. By

extension, we hypothesize that brain tumor stem cells may also retain responsiveness to Wnt

signaling by altering cell fate decisions, and thus may offer a novel target for therapy of GBMs.

Page 37: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

27

Chapter 2

2 Materials and Methods

2.1 Cell culture and differentiation protocol

Human fetal brains (5205: Gestational Week (GW) 11; 5281: GW12.5) were mechanically

dissociated in Neurobasal medium (Invitrogen) and made into single cell suspensions with

Accutase (Sigma) treatment. Primary cells were plated onto laminin (10 mg/L, Sigma)-coated

dishes (Primaria) in expansion media made up of NeuroCult Basal Human medium (Stem Cell

Technologies), modified N2 supplement (Ying and Smith, 2003), B27 (20ml/L final, Invitrogen),

penicillin-streptomycin-fungizome (10 ml/L final, Sigma), Heparin (2 µg/ml final, Sigma),

recombinant mouse EGF (10 ng/ml final, Sigma), and recombinant human FGF-2 (Stem Cell

Technologies). Cells were incubated in a 37°C incubator containing 5% CO2, fed every 2-3 days

and passaged 1:2-1:3 when cultures became confluent.

Brain tumour samples were obtained from patients treated in Toronto hospitals following local

ethical board approval. GliNS1 (unknown, M), G377 (19 yr, F), G179 (52 yr, M) were all

diagnosed as glioblastoma multiforme (GBM). GNS cells were cultured as described above with

human NS cells.

In order to differentiate NSCs and BTSCs cells, 0.5-2×106 human NS cells were plated onto

poly-L-ornithine (Sigma) and laminin 10 cm dishes in expansion medium for 24 hours then

switched to expansion medium without EGF. After 7 days, medium was changed to the mix

NeuralCult basal-Neurobasal (1:1), supplemented with N2 (0.5×), Heparin (2 µg/ml final), and

B27 (1×) for 14 days. Recombinant mouse Wnt3a (R&D laboratories) and BIO (sigma) were

used in some cell culturing experiments.

2.2 Immunoblotting

Cells were lifted off the tissue culture plates with Accutase (Sigma), washed with PBS and lysed

in lysis buffer (50 mM Tris pH=7.4, 1% NP-40, 0.25% NaDeoxycholate, 0.15 M NaCl, 1mM

EDTA, 1 mM Na3VO4, 1mM NaF, 1% SDS) to obtain total protein. Cellular lysate was loaded

with 4X SDS loading buffer (125 mM Tris pH 6.8, 4% SDS, 10% glycerol, 0.006%

bromophenol blue, 18 µg/ml β-mercaptoethanol) onto 7.5% and 15% SDS gels and separated by

Page 38: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

28

electrophoresis. Proteins were transferred to Imobilon-P PVDF membranes (Millipore) using a

wet transfer apparatus (BioRad) and blocked in 5% non-fat Milk 1×TBST for an hour at room

temperature. The primary antibodies β-catenin (1:1000, Cell Signaling), β-III-tubulin (1:1000,

Chemicon), GFAP (1:1000, Dako), Nestin (1:1000, Chemicon, and β-actin (1:5000, Sigma) were

incubated in 1% non-fat milk 1×TBST either overnight at 4°C or at room temperature for one

hour. Membranes were then probed with the HRP conjugated secondary antibody in 1% non-fat

milk 1×TBST for one hour at room temperature and detected with enhanced chemiluminescence

(ECL) (GE health) and exposed to Amersham hyperfilm ECL film. Blots were stripped with

Restore stripping buffer (Thermo Scientific) for 10-15 minutes at room temperature and reprobed

if necessary.

2.3 Immunofluorescence

Cells were plated at required density (stated in results text and figures) on poly-L-ornithine and

laminin-coated glass coverslips. Cells were fixed in 4% PFA at room temperature for 10-20

minutes, permeabilized with 0.3%-Triton-100X PBS for 5 minutes, and blocked in 1X PBS with

10% FBS for one hour at room temperature. Cells were stained with primary antibody (1:500; β-

III-tubulin, 1:500; GFAP, 1:1000; MAP2, 1:500) in 1X PBS with 10% FBS either for one hour at

room temperature or overnight at 4°C. Cells were then washed in 1XPBS and incubated with

secondary antibodies (Alexa-488; Alexa-568) for one hour at room temperature. Following three

washes, converslips were mounted onto glass slides with DAPI staining. All analysis was

performed on the Zeiss Axiovert 200M microscope.

2.4 Intracellular Flow Cytometry

Cells were dissociated from the tissue culture plate and suspended in 1 ml of 1X PBS. Cells were

fixed with 50 µl of 32% PFA for 10 minutes on ice. Cells were washed with 1X PBS and

permeabilized with 100% ice-cold methanol for 30 minutes on ice, followed by two more washes

with 1X PBS. Cells were blocked in staining buffer (5% NGS, 4mM EDTA, 15mM HEPES,

PBS) for at least one hour at 4°C and then incubated with primary antibody (β-III-tubulin, 1:500;

GFAP, 1:8000) in staining buffer for 30-60 minutes at 4°C. Cells were washed in fresh staining

buffer and incubated in fresh staining buffer with secondary antibodies (Alexa-488; Alexa-405;

Alexa-350; Alexa-633) for 30-60 minutes at 4°C. Cells were washed in fresh staining buffer and

Page 39: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

29

left overnight at 4°C to be analyzed the next morning. Cells were analyzed on the BD LSRII-SC

analyzer.

2.5 Luciferase Assay

Luciferase assays were performed as outlined in the Promega Dual-Luciferase Reporter Assay

System (Cat. No. E1910) in 24-well tissue culture plates coated in PLO and laminin. The Renilla

luciferase plasmid was used as a transfection and loading control for every assay. SuperTOPflash

(and FOPflash) luciferase reporter constructs were used to measure TCF/LEF transcriptional

activity and were generous gifts from R.T. Moon.

2.6 Transfection of cells

Transfections were performed according to protocol of the Amaxa mouse Neural Stem Cell

Nucleofector Kit (Lonza). Transfected cells were always replated in full media with EGF and

FGF for a minimum of 24 hours prior to any assay to ensure full recovery post-transfection. Each

plasmid was transfected in the following amounts: TOPflash (or FOPflash): 3µg, Renilla

luciferase: 0.06µg, ΔN90 (or pcDNA empty vector): 5µg. The ΔN90 cDNA expression vector

was a generous gift from B. Alman.

2.7 MTT assays

Cells were seeded in a poly-L-ornithine and laminin-coated 96-well tissue culture plate at

required density (stated in results text and figures). Proliferation was measured on required days

(stated in results text and figures) by adding MTT reagent (1:10, Roche) to the final volume in

each well. Cells and MTT reagent were incubated for 4 hours at 37°C. Equal volume of MTT

solubilizaton buffer to total volume was added to each well and incubated overnight at 37°C to

allow for full lysis of cells. Quantification of viable cells through reading of ultraviolet

absorption spectrum at 575 nm was performed the next day on a Versamax microplate reader

(Molecular Devices, Sunnyvale, CA) equipped with SoftMax Pro software (Molecular Devices,

Sunnyvale, CA).

2.8 BrdU labeling

Cells grown on glass cover slips were pulsed with BrdU (10µM) for 24 hours and then washed

twice with PBS. Cells were then fixed with 4% PFA for 20 minutes at room temperature. Cells

Page 40: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

30

were washed with PBS and then treated with 2M HCl for 20 minutes at room temperature. Cells

were then treated with 0.1M sodium borate (NaB4O7) (pH8.5) for 2 minutes at room temperature,

followed by 2 washes with PBS. Cells were stained for BrdU (1:250) according to the previously

described immunocytochemistry protocol.

Page 41: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

31

Chapter 3

3 Results

3.1 Wnt signaling promotes a neuronal cell fate choice in human fetal neural stem cells

3.1.1 Human fetal neural stem cells express key Wnt pathway components and exhibit low baseline TCF/LEF-mediated transcription

In order to obtain an understanding of whether Wnt signaling was active in normal human neural

stem cells (hNSCs), we first sought to determine whether key signaling components were

expressed in serum free cultures of human neural stem cells that we had isolated from human

fetal brain. Reverse transcriptase polymerase chain reaction (RT-PCR) was used to determine the

mRNA expression of various members of the pathway, including Frizzled receptors Fz1, Fz3,

Fz6, Fz7, Fz8, Fz9 and Low density lipoprotein related-receptor protein 5 (Lrp5), the soluble

pathway inhibitor and target gene DKK1, and the central signaling regulators Gsk3βandβ‐

catenin(Figure3a).The mRNA expression patterns of these receptors demonstrated variable

transcript levels and heterogeneity among Wnt pathway components and between hNSC lines

(Figure 3a). We performed western blot analysis on these two hNSC lines to confirm protein

expression of β-catenin, the central mediator of the canonical pathway (Figure 3b). Given that

key components for β-catenin-mediated Wnt signaling are expressed in hNSCs, these cells will

likely respond to experimental manipulation of pathway activity.

Page 42: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

32

Figure 3 - HumanNSCsexpresscanonicalWntsignalingpathwaycomponents

hNSCs express Wnt signaling pathway components. a) RT-PCR for various components

reveal that hNSC lines express various Frizzled receptors (1, 3, 6, 7, 8. 9), Lrp5, Dkk1, and

the central components β-catenin and Gsk3β at the mRNA level. Gapdh was used as a

loading control. b) β–catenin was also detected at the protein level by western blotting. β-

actin was used as a loading control.

Page 43: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

33

Canonical Wnt signaling is a cascade of molecular interactions converging on the stabilization of

β-catenin and culminating in TCF/LEF—mediated transcription of target genes (Figure 4a).

Wnt3a is one of 19 Wnt ligands identified in the mammalian system. We then tested whether

exogenous stimulation of Wnt signaling resulted in alteration in expression and activity of β-

catenin to assess whether the Wnt signaling cascade is intact in our hNSCs. Within one hour of

exogenous Wnt3a stimulation, β-catenin levels were increased compared to baseline levels in

both hNSC lines, indicating that Wnt3a stimulation resulted in the stabilization and accumulation

of β-catenin (Figure 4b).

The luciferase reporter assay, TOPflash, measures TCF/LEF-mediated transcriptional activity,

which is a measure of canonical Wnt signaling. Eight TCF/LEFconsensus DNA-binding sites lie

upstream of a minimal TK promoter and the firefly luciferase gene (Figure 4c).The negative

control, FOPflash, has eight mutated TCF/LEFbinding sites. When Wnt signaling is activated,

luciferase is transcribed and levels correlate directly to signaling output. As a means of

transfection control, the Renilla luciferase gene is co-transfected into cells to give an estimate of

total transcriptional activity within the bulk transfected cells, serving as a transcriptional

baseline. TOP/FOPflash levels are determined by normalizing it to baseline transcriptional

activity. We used the TOPflash assay to investigate whether the observed stabilization of β-

catenin protein levels correlates with TCF/LEFtranscriptional activity. We found that hNSCs

exhibit very low basal Wnt signaling levels in serum free EGF/FGF conditions in vitro and were

able to respond to Wnt3a stimulation, demonstrating 30-55 fold increase in luciferase levels after

24 hours. Therefore, while hNSCs maintain low basal TCF/LEF transcriptional activity in vitro,

they are capable of responding to Wnt signaling, which may have implications regarding their

functional characteristics.

Page 44: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

34

Figure 4 – hNSC lines activate TCF/LEF transcriptional activity in response to Wnt3a.

Human fetal neural stem cells are Wnt-responsive and activate TCF/LEF transcription. a)

A schema depicting the sequence of events that begins with a Wnt ligand binding to a

Frizzled receptor complex, inhibiting the GSK3α/β-APC-Axin destruction complex,

stabilizing β-catenin and activating TCF/LEF transcription of target genes. b) Cells treated

with recombinant mouse Wnt3a (100 ng/ml) for 2 hours respond to Wnt3a stimulation

through the accumulation β-catenin to varying degrees relative to baseline levels. c) The

TOPFlash construct is depicted with 8X TCF/LEFconsensus-binding sites upstream of a

minimal TK promoter and the Firefly luciferase gene. d) hNSCs transfected with TOPflash

(or the negative control FOP) and the Renilla luciferase construct. After 24 hours of

recovery, hNSCs were treated with Wnt3a (100 ng/ml) for 24 hours (or fresh media) and

their TOP/FOP levels were compared in the above bar graph. All TOP and FOP readings

are normalized to the Renilla luciferase output for transfection control.

Page 45: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

35

3.1.2 Wnt3a does not alter human fetal neural stem cell proliferation

Within the mammalian CNS, Wnt3a signaling has been shown to be critical for proper

hippocampal development by regulating hippocampal precursor expansion116. Wnt3a is

considered to be a canonical Wnt ligand that activates the β-catenin-mediated pathway. The

current model has Wnt3a forming a receptor-ligand complex with Frizzled and Lrp5/6 that

triggers a series of intracellular events leading to the destabilization of the destruction complex

and the accumulation of β-catenin (Figure 2a).

Exogenous stimulation of hNSCs with Wnt3a is a physiologically relevant approach to activating

β-catenin—mediated signaling and induces a 30 to 55-fold increase in TCF/LEF transcriptional

activity (Figure 4d). We wanted to know whether exogenous Wnt3a stimulation resulted in any

functional consequences in hNSCs. Using the MTT assay to assess proliferation and survival, we

found no significant changes in hNSCs treated with Wnt3a analyzed over 13 days in the presence

of EGF and FGF compared to their untreated counterparts (Figure 5a). Therefore, it seems that

Wnt signaling does not cooperate with EGF and FGF signaling to promote proliferation further.

We were concerned that the potent mitogenic effects of EGF and FGF were masking any

possible negative affects exerted by Wnt3a signaling. Therefore, we performed MTT assays on

Wnt3a-treated hNSCs under differentiating conditions—in the absence of EGF and FGF (-

EGF/FGF)—over 13 days, and no significant change in proliferation of hNSCs treated with

Wnt3a was observed (Figure 5b). Although it is possible that Wnt signaling is not a major

regulator of hNSC function, this is likely not the case. Another explanation is that Wnt signaling

does not directly regulate hNSC proliferation, but may affect other cell fate decisions, such as

lineage fate choice.

Page 46: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

36

Figure 5 – Wnt3a does not alter hNSC proliferation.

Wnt3a does not significantly alter hNSCs proliferation. a) hNSCs were seeded at 2000

cells/well (96-well plate), grown for 13 days in stem cell conditions (+EGF/FGF) and in the

presence or absence of recombinant mouse Wnt3a (50 ng/ml). No significant difference in

proliferation, as measured by the MTT assay, was observed between the two groups (n=3).

b) hNSCs were seeded at 2000 cells/well (96-well plate), grown for 13 days under

differentiating conditions (-EGF/FGF) and in the presence or absence of recombinant

mouse Wnt3a (50 ng/ml). No significant difference in proliferation was observed between

the two groups (n=4; unpaired student’s T-test).

Page 47: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

37

3.1.3 Wnt3a promotes a neuronal cell fate choice in human fetal neural stem cells under differentiating conditions

Several canonical Wnt species, including Wnt3 and Wnt7a, have been shown to regulate

neuronal differentiation in the murine brain3, 118. Therefore, we hypothesized that Wnt3a may

promote a neuronal fate choice in differentiating hNSCs. In order to test this hypothesis, we

differentiated hNSCs in the absence or presence of exogenous Wnt3a, and analyzed the

expression of lineage-specific markers using two methods of analysis (Figure 6a).

Immunofluorescence revealed that Wnt3a-treated hNSCs differentiated to produce a greater

proportion of βIII-tubulin-positive neurons compared to untreated differentiated hNSCs (Figure

6b). Furthermore, the proportion of GFAP-positive cells was greatly reduced in Wnt3a-treated

hNSCs, suggesting Wnt3a may have an inhibitory effect on astrogenesis (Figure 6b). In order to

quantify this phenomenon, we performed intracellular flow cytometry for the same markers

(Figure 6c). Results indicate a 2.6 fold (p=0.0235; n=4) increase in βIII-tubulin-positive neurons

and a 0.59-fold decrease (p=0.0124; n=4) in GFAP-positive astrocytes in the presence of Wnt3a,

suggesting that Wnt3a activation of the canonical pathway promotes a neuronal fate choice in

cultured hNSCs at the expense of the glial lineage.

Page 48: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

38

Figure 6 – Wnt3a promotes a neuronal cell fate in differentiating hNSCs.

Exogenous Wnt3a promotes a neuronal cell fate in differentiating hNSCs. a) Cells were

differentiated for 21 days in the absence or presence of Wnt3a (100 ng/ml) and analyzed for

Page 49: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

39

lineages specific markers using immunofluorescence and intracellular flow cytometry. Blue

arrows indicate when media was changed and fresh media and Wnt3a were added to cells.

b) hNSCs differentiated in the presence of Wnt3a significantly increased the number of

βIII-tubulin-expressing cells (red) compared to the untreated differentiated hNSCs.

Furthermore, Wnt3a-treated hNSCs also produced significantly fewer GFAP-expressing

cells (green) compared to untreated hNSCs (DAPI, blue). c) Intracellular flow cytometry

histograms reveal that the number βIII-tubulin-expressing cells increases by 2.598-fold and

a 0.588-fold decrease in the amount of GFAP-positive cells in the presence of Wnt3a

relative to untreated hNSCs. Unstained, red; untreated, green; Wnt3a-treated, blue (n=4;

paired student’s T-test).

Page 50: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

40

3.1.4 Stabilized β-catenin activates Tcf/Lef—mediated transcription

The stabilization of β-catenin is critical for its translocation to the nucleus and subsequent

coactivation of Tcf/Lef transcription factors. Cytosolic β-catenin levels are modulated by CK1’s

and GSK3’s constitutive phosphorylation on several conserved amino acid residues (S45, T41,

S37, and S33) located at the amino terminus of the protein. These serial phosphorylation events

prime the protein for ubiquitination and proteasomal degradation. Therefore, stabilization

requires that β-catenin evades phosphorylation and subsequent degradation. In order to

circumvent degradation and the need for extracellular stimulation, a truncated mutant of β-

catenin was produced where the first 90 amino acids are removed from the amino terminus of the

protein, ΔN90 β-catenin (or just ΔN90). This truncation results in the loss of the key

phosphorylation substrates of CK1 and GSK while conserving critical domains integral to its

function, such as the transactivating domain and other protein-protein binding domains also

present in the wild-type (Figure 7a)145.

Previous studies have shown that the expression of ΔN90 within the neural precursor

compartment of the developing murine CNS in vivo results in active Wnt signaling, increased

cell cycle reentry, increased proliferation, and thus an expansion of the precursor pool120.

Therefore, in parallel to our other methods, we set out to understand how ΔN90 might affect

hNSCs in vitro. First, we confirmed that the protein product of the ΔN90 plasmid is detectable

by immunoblotting 48 hours after the transfection. Indeed, when we probe with a carboxy-

terminal recognizing antibody, we do see a second band at around 78 kDa as well as the

endogenous protein located at 92 kDa (Figure 7b). Next, we hoped to confirm that ΔN90

increases Tcf/Lef-mediated transcription, thereby mimicking activated Wnt signaling. Using the

TOPFlash assay we were able to confirm that ΔN90 results in a dramatic increase (167-fold) in

signaling activity relative to endogenous signaling levels (pcDNA transfection control vector)

(Figure 7c).

Page 51: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

41

Figure 7 – Stabilized β-catenin activates Tcf/Lef transcriptional activity in hNSCs.

ΔN90-β-catenin activates Tcf/Lef transcription. a) Schema of wildtype (WT) and the

stabilized mutant form of β-catenin (ΔN90) illustrate the 13 armadillo repeats, the Axin,

APC and TCF/LEF binding domains. ΔN90 has an amino terminal truncation of the

protein, which removes key phosphorylation substrates targets by CK1 and GSK3 kinase

activity. b) hNSCs were transfected with either ΔN90 or the control empty vector (pcDNA).

Cell lysates were collected 48 hours post transfection and an immunoblot was performed

for total β-catenin levels. Only ΔN90-transfected cells expressed ΔN90-β-catenin at ~78kDa.

c) hNSCs were transfected with the TOPflash (or FOPflash) and Renilla construct in

conjunction with either the ΔN90 construct (or pcDNA). The TOPFlash assay indicates that

ΔN90 dramatically elevates (~167-fold) Tcf/Lef activity relative to baseline levels (pcDNA)

(n=2).

Page 52: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

42

3.1.5 Stabilized β-catenin does not alter human neural stem cell proliferation.

In light of previously reported findings as well as the substantial induction of TCF/LEF activity,

we predicted that expression of ΔN90 in hNSCs would result in increased proliferation. We

performed an MTT assay comparing ΔN90-transfected cells to pcDNA-transfected cells in stem

cell conditions (+EGF/FGF). Surprisingly, there was no significant difference in proliferation

between ΔN90-expressing cells and the control-transfected hNSCs in the presence of EGF and

FGF (Figure 8a). One possibility is that β-catenin does not act synergistically or additively with

the potent mitogenic effects of EGF and FGF in the culturing media, and its effects are masked.

Therefore, we sought to investigate the effects of stabilized β-catenin in conditions where hNSCs

are induced to differentiate (-EGF and FGF). We performed an MTT assay comparing ΔN90-

transfected cells to the control-transfected control cells at days 4 and 8 post-transfection under

differentiating conditions and found no significant difference in proliferation (Figure 8b). These

results indicate that, contrary to findings observed in embryonic mouse neural precursors in vivo,

stabilized β-catenin does not increase the proliferation and/or survival of hNSCs in vitro.

Page 53: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

43

Figure 8 – Stabilized β-catenin does not affect proliferation of hNSCs in vitro.

Stabilized β-catenin does not significantly alter proliferation rate of hNSCs. a) hNSCs were

transiently transfected with either pcDNA (transfection control) or ΔN90, seeded at 2000

cells/well (96-well plate) in EGF and FGF and grown for 8 days. Proliferation, as measured

by the MTT assay, was not significantly altered by stabilized β-catenin (n=3). b) hNSCs

were transiently transfected with either pcDNA (transfection control) or ΔN90, seeded at

2000 cells/well (96-well plate) in differentiating conditions (-EGF/FGF) and grown for 8

days. Proliferation, as measured by the MTT assay, was not significantly altered by

stabilized β-catenin (n=3; unpaired student’s T-test).

Page 54: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

44

3.1.6 Stabilized β-catenin promotes neuronal cell fate choice in human fetal neural stem cells under differentiating conditions

Several groups have observed that Wnt signaling also plays a role in cell fate determination of

neural precursors117,118. Therefore, we wanted to investigate whether stabilized β-catenin

affected the cell fate choice of hNSCs during differentiation. In order to interrogate this, we

transfected hNSCs with ΔN90 or pcDNA (transfection control) and differentiated them in the

absence of EGF and FGF (Figure 9a)146. We confirmed the presence of the ΔN90 mutant at the

protein level two days post-transfection (Figure 7b). After 21 days, we performed

immunofluorescence for βIII-tubulin and GFAP to identify both a neuronal and astrocyte

population, respectively. We found that constitutively active Wnt signaling in ΔN90-transfected

hNSCs prior to differentiation mildly increased the number of βIII-tubulin-positive neurons but

showed no dramatic change in GFAP-expressing cells (Figure 9b). An immunoblot confirmed

the mild pro-neuronal trend and showed a decrease in GFAP protein levels, indicating a mild

block in astrogenesis in the ΔN90-differentiated population (Figure 9c). Furthermore, nestin

protein levels were decreased in ΔN90-transfected differentiated populations, inferring a more

terminally differentiated population compared to the control differentiated population (Figure

9c). We used intracellular flow cytometry to quantify neuronal and astrocytic populations within

each sample using βIII-tubulin and GFAP, respectively, and found a 1.697-fold increase in the

number of βIII-tubulin expressing cells in differentiated hNSCs transfected with ΔN90 compared

to the pcDNA-transfected control group (Figure 9d). Only a slight decrease was detected in the

number of GFAP-positive cells present in the ΔN90-transfected sample (0.987-fold) (figure 9d).

While these results are preliminary and must be repeated several times to confirm the observed

pro-neuronal trend, they do suggest that activation of the canonical Wnt signaling pathway helps

to promote a neuronal cell fate choice in differentiating hNSCs. Furthermore, while previous

reports focus on the pro-proliferative affects of ΔN90 in the precursor pool of the murine nervous

system, no such affects were observed in hNSCs in vitro (Figure 8).

Page 55: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

45

Figure 9 – Stabilized β-catenin promotes a neuronal cell fate choice during differentiation.

Stabilized β-catenin promotes a neuronal cell fate under differentiating conditions. a)

hNSCs were transiently transfected with either pcDNA or ΔN90 and differentiated by

sequential removal of EGF and FGF for a period of 21 days. b) Differentiated hNSCs were

Page 56: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

46

stained for neuronal (βIII-tubulin, red) and glial (GFAP, green) markers. Cells transfected

with ΔN90 produced a greater proportion of βIII-tubulin—expressing cells and no change

in GFAP—positive cells numbers compared to the transfection control (pcDNA). c)

Western blots for βIII-tubulin and GFAP also confirmed that stabilized β-catenin (ΔN90)

promoted a neuronal cell fate choice in differentiating hNSCs as well as a decrease in

GFAP. Furthermore, immunoblotting for the precursor marker nestin was drastically

reduced in ΔN90-transfected hNSCs. d) Intracellular flow cytometry histograms for βIII-

tubulin and GFAP reveal a 1.697-fold increase in the proportion of βIII-tubulin-positive

cells and a slight decrease (0.987-fold) in the number of GFAP-positive cells detected in

ΔN90-transfected cells compared to the control. Unstained, red; pcDNA control, green; and

ΔN90, blue. (n=1).

Page 57: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

47

3.1.7 BIO—mediated inhibition of GSK3 activates TCF/LEF transcription

Concurrently, a third strategy to perturb canonical Wnt signaling is by treating with GSK3 small

molecule inhibitors since chemical inhibitors represent a clinically relevant method of altering

Wnt signaling. These are thought to have potent effects on Wnt signaling, but may also interfere

with other pathways. We decided to use the small molecule GSK3 inhibitor, BIO, to further

interrogate cell fate choices of hNSCs because it is easily accessible and other independent

groups have published functional studies using BIO as a method of Wnt stimulation104. BIO

((2’Z,3’E)-6-Bromoindirubin-3’-oxime), is a reversible ATP competitive inhibitor of GSK3α/β

that can serve as a Wnt signaling agonist (Figure 10a)102. While BIO is specific to GSK3α/β,

concentrations above 0.005 µM have also been shown to inhibit the closely related cyclin-

dependent kinases (CDKs) 1, 2, and 5, which may influence proliferation rates104.

GSK3 was initially identified as a kinase involved in glucose metabolism, but was later

discovered to play an integral role as a negative regulator of the canonical Wnt pathway147.

GSK3 participates as a key member of the destruction complex by phosphorylating β-catenin,

priming it for ubiquitination and subsequent proteasomal degradation. When GSK3 kinase

activity is disrupted, β-catenin can evade phosphorylation and translocate to the nucleus to

interact with the TCF/LEF transcription factor complex, mediating target gene transcription

(Figure 10a). BIO has been shown to stabilize β-catenin and mimic Wnt signaling104. To

validate that BIO acts as a Wnt signaling agonist in our system, we performed a TOPFlash assay

on hNSCs treated with BIO for 24 hours (Figure 10b). We used 1µM of BIO because

concentrations below 0.5µM showed almost no TCF/LEF activation after 24 hours (data not

shown) and its only off-target substrates were the CDKs mentioned above. Results indicate that

1µM of BIO significantly activates TCF/LEF transcription by approximately 26.4-fold relative to

baseline transcriptional activity, thereby mimicking canonical Wnt activation. Although higher

concentrations (2 µM) illicited a higher TCF/LEF-mediated signal as measured by TOPFlash

activity, this concentration was accompanied by increased cell sloughing, suggesting cytotoxic

effects (data not shown). Therefore, chemical activation of Wnt signaling by GSK3 inhibition

offers another strategy for assessing the function of hNSCs in vitro.

Page 58: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

48

Figure 10 – BIO-mediated inhibition of GSK3 activates TCF/LEF transcription

a) BIO ((2’Z,3’E)-6-Bromoindirubin-3’-oxime) inhibits both GSK3α and GSK3β kinase

activity, resulting in the stabilization and accumulation of β-catenin and transcriptional

activation of the TCF/LEF complex. b) hNSCs were transfected with the TOPflash

construct (or FOP) and the Renilla luciferase construct. After 24 hours of recovery, hNSCs

were treated with BIO (1µM) for 24 hours (or fresh media for endogenous levels) and their

TOP/FOP levels were compared in the above bar graph. All TOP and FOP readings are

normalized to the Renilla luciferase output for transfection control. BIO was able to induce

a 26.4-fold increase in TCF/LEF activity relative to endogenous TCF/LEF activity (*:

p<0.01; n=3; unpaired student’s T-test).

Page 59: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

49

3.1.8 BIO promotes differentiating human fetal neural stem cells to slow proliferation and exit the precursor state

Activation of Wnt signaling by ΔN90 and Wnt3a did not show any affect on proliferation of

hNSCs under differentiating conditions. However, when we performed an MTT assay for hNSCs

treated with BIO under differentiating conditions, we found a significant reduction in

proliferation of hNSCs treated with BIO (Day 4: p≤0.001; Day 10: p≤0.0001, Figure 11a). This

observation may be due to a GSK3-mediated pathway that does not converge solely on Wnt

signaling. Also described earlier, BIO may also inhibit key CDKs at the concentration used.

Therefore, the affect on proliferation may be attributed to CDK inhibition, rather than GSK3

inhibition. Further investigation is required to determine which target molecule is responsible for

the observed decrease in proliferation.

HNSCs were differentiated in the absence of EGF and FGF for 21 days and then analyzed for

precursor marker expression by intracellular flow cytometry. BIO-treated hNSCs showed a

significant decrease in the number of Nestin-positive (0.334-fold) and Sox2-positive (0.41-fold)

cells compared to untreated differentiated hNSCs (Figure 11b). Together, the decrease in

proliferation paired with the loss of immature marker expression suggests that BIO-mediated

inhibition of GSK3 may promote hNSCs to exit the precursor state toward a more differentiated

cell type.

Page 60: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

50

Figure 11 – BIO-mediated GSK3 inhibition promotes hNSCs to exit the precursor state.

a) hNSCs were seeded at a density of 2000 cells/well (96-well plate) and grown under

differentiating conditions (-EGF/FGF) for 10 days either in the presence or absence of BIO

(1µM). Treatment with BIO resulted in a significant decrease in hNSC proliferation (*:

p≤0.001, **: p≤0.0001) (n=3; unpaired student’s T-test). b) Intracellular flow cytometry

histograms reveal that BIO-treated differentiated hNSCs exhibit a significant decrease in

then proportion of Nestin-positive and Sox2-positive cells compared to their untreated

counterparts. Unstained, red; untreated, green; BIO-treated, blue (n=3; paired student’s

T-test).

Page 61: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

51

3.1.9 BIO promotes a neuronal cell fate choice in differentiating human fetal neural stem cells

Since activation of canonical Wnt signaling by other approaches results in a neuronal fate choice

during hNSC differentiation, and that BIO causes a loss in precursor marker expression, we

predicted that BIO treatment in the absence of growth factors would result in an increased

proportion of neurons within the differentiated population. In order to validate our predictions,

we differentiated our hNSCs for 3 weeks in the absence or presence of BIO and then analyzed

our cells for expression of lineage-specific markers (Figure 12a). hNSCs differentiated in the

presence of BIO showed a substantial increase in βIII-tubulin-positive cells compared to

untreated differentiated hNSCs when analyzed by immunofluorescence (Figure 12b).

Simultaneously, there was a drastic reduction in the number of GFAP-positive cells when

differentiated in the presence of BIO (Figure 12b). Intracellular flow cytometry for βIII-tubulin

and GFAP was performed in order to quantify these changes. hNSCs showed a significant

increase in neuronal differentiation (1.9-fold) at the expense of glial differentiation (0.4-fold)

when GSK3 is inhibited by BIO in differentiating hNSCs (Figure 12c). Together with earlier

data, our results seem to suggest that BIO-mediated GSK3 inhibition enhances differentiation

and specifically promotes a commitment to the neuronal lineage at the expense of an astrocytic

lineage in the absence of growth factors.

Page 62: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

52

Figure 12 – BIO promotes a neuronal cell fate choice in differentiating hNSCs.

BIO promotes a neuronal cell fate choice in differentiating hNSCs. a) Cells were

differentiated for 21 days in the absence or presence of BIO (1µM) and analyzed for

Page 63: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

53

lineages specific markers using immunofluorescence and intracellular flow cytometry. Blue

arrows indicate when media was changed and fresh media and BIO were added to cells. b)

hNSCs differentiated in the presence of BIO (1µM) (-EGF/FGF + BIO) showed a drastic

increase in βIII-tubulin—expressing (green) cells and a substantial decrease in GFAP—

expressing (red) cells compared to untreated differentiated (-EGF/FGF) hNSCs, as

determined by immunofluorescence. c) Intracellular flow cytometry histograms show that

the proportion of βIII-tubulin-positive cells increases by 1.9-fold and the number of GFAP-

positive cells decreases to 0.4-fold of the untreated proportion. Unstained, red; untreated,

green; BIO-treated, blue (n=3; paired student’s T-test).

Page 64: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

54

3.1.10 BIO decreases proliferation and induces neuronal differentiation of human neural stem cells in EGF and FGF

In vitro, NSCs are bathed in a pool of mitogens, including EGF and FGF, which are known to

promote progenitor expansion148. Since BIO significantly reduced proliferation in the absence of

growth factors, we wanted to know whether BIO-mediated GSK3 inhibition could overcome the

mitogenic effects of EGF and FGF to decrease proliferation of hNSCs under stem cell

conditions. We performed MTT assays on hNSCs treated with BIO (and untreated control

hNSCs) for 10 days in EGF and FGF, and found that BIO treatment resulted in a significant

decrease in proliferation of hNSCs (Day 4, p=0.0082; Day 10, p=0.0003, Figure 13a). One

explanation for the observed decrease in proliferation is that cells are undergoing differentiation

to produce a more quiescent population. Given how important EGF and/or FGF are in

maintaining self-renewal and proliferation of stem cells in vitro7, we wanted to know whether

BIO was potent enough to overcome these mitogenic signals and induce neuronal differentiation.

To address this possibility, we treated hNSCs with BIO for 14 days in the presence of EGF and

FGF and then performed intracellular flow cytometry for the known neural progenitor marker

nestin as well as neuronal- and astrocyte-specific markers βIII-tubulin and GFAP, respectively

(Figure 13b). Given that cells slowed down when treated with BIO in the presence of EGF and

FGF, we suspected that hNSCs were exiting the progenitor state, thereby losing nestin

expression. However, over four independent experiments, we found no significant change in

nestin expression in our hNSCs after treating with BIO. Likewise, we found no consistent trend

in the fold-change of GFAP-expressing cells when hNSCs are treated with BIO in the presence

of EGF and FGF. However, we do see a 1.8-fold increase in βIII-tubulin expression, suggesting a

trend toward induction of neurogenesis when hNSCs are treated with BIO in the presence of

EGF and FGF. Therefore, it seems that a GSK3-mediated decrease in proliferation may reflect

some change in metabolic and/or cell cycle rate, hNSC neuronal differentiation in stem cell

conditions, or both effects. It is possible that GSK3 plays a role in both proliferation and

neuronal commitment, as is suggested from earlier data in differentiating hNSCs, but that the

pathways responsible for regulating each function are not necessarily one and the same and may

exert different effects depending on the larger molecular context. In sum, a trend is seen where

GSK3-inhibition in EGF and FGF is able to overcome EGF and FGF to induce differentiation.

Page 65: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

55

Figure 13 – BIO decrease proliferation and induces mild neuronal differentiation in EGF

and FGF.

BIO decreases proliferation and promotes a proportion of hNSCs to exit from the

progenitor state. a) hNSCs were seeded at 2000 cells/well (96-well plate) in EGF and FGF,

and grown in the absence or presence of BIO (1µM) for 10 days. BIO-treated hNSCs

showed a significant decrease in proliferation as observed by MTT assay (*: p≤0.01, **:

p≤0.001) (n=3; unpaired student’s T-test). b) The bar graph represents the average fold

changes in marker expression when hNSCs are treated with BIO (1µM) for 14 days in stem

cell conditions (+EGF/FGF) (n=4; paired student’s T-test).

Page 66: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

56

3.2 GSK3 inhibition induces neuronal differentiation of brain tumour stem cells

3.2.1 Brain tumour stem cells express Wnt pathway components and can activate TCF/LEF-transcriptional activity.

Glioblastoma multiforme (GBMs) is an aggressive type of glial brain tumour that is

characterized by its cellular heterogeneity, a high mitotic index, invasiveness, and necrotic foci45.

GBM tumours demonstrate cell populations that stain for both precursor and differentiated

markers. These precursor phenotype cells are thought to contain the tumour-initiating population

(BTSCs) responsible for the growth and maintenance of the bulk tumour25. Given that BIO-

mediated inhibition of GSK3 activity was able to attenuate proliferation and promote neuronal

lineage commitment under specific signaling contexts in normal hNSCs, we wanted to determine

whether BIO could have similar functional affects on BTSCs. Since hNSCs and BTSCs are

functionally similar in several respects (i.e. proliferation, multilineage differentiation, and self-

renewal) we predict that GSK3 inhibition will lead to increased BTSC neuronal differentiation.

First, we wanted to confirm that our BTSC lines (G377, G179, and GliNS1) express Wnt

pathway components, similar to our hNSC lines. RT-PCR revealed that BTSC lines express key

components at the mRNA level, including the receptors frizzled Fz1, 3,6,7,8,9, and Lrp5, the

central mediators β-catenin and Gsk3β, and the canonical secreted inhibitor Dkk1 (Figure 14a).

As with our normal hNSC lines, BTSC lines also show unique heterogeneous expression patterns

of Wnt pathway components, suggesting that our in vitro conditions preserve in vivo expression

signatures. We then confirmed protein-level expression of β-catenin in our BTSC lines by

western blot and found that while all BTSCs expressed β-catenin to varying degrees, GliNS1

possessed the greatest amount of β-catenin compared to two other BTSC lines (Figure 14b).

Interestingly, however, GliNS1 also showed the highest level of active GSK3 isoforms (GSK3β

Y216 more so than GSK3α Y279) of all BTSC lines, making GliNS1 a sensible cell line for

BIO-mediated GSK3-inactivation. SW480 is an adenocarcinoma cell lines with an APC mutation

that results in the stabilization of β-catenin, and therefore makes for a suitable positive control

for β-catenin. It too possesses high levels of active GSK3β, but this has little bearing on β-

catenin levels due to the functional loss of APC. From this point on we chose to carry out our

preliminary investigations using GliNS1 for this and several reasons. First, GliNS1 is a cell line

Page 67: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

57

derived from a glioblastoma that has been confirmed by clinical pathology. Moreover, with the

foresight to eventually move to in vivo studies, GliNS1 has been shown to be highly

tumourigenic in orthotopic tumour engraftment models by other members of the Dirks

laboratory. And furthermore, it has also been included in our microarray studies and therefore

makes gene expression data readily available to gain further insight into the function of this cell

line.

In order to determine whether GliNS1 was BIO-responsive through the β-catenin TCF/LEF

pathway, we performed the TOPFlash assay and found that 1µM of BIO for 24 hours was

sufficient to induce a 44-fold increase in signaling activity (145.5 versus 3.32 Relative

Luciferase Units—RLU) over baseline TCF/LEF transcriptional levels (Figure 14c). Taken

together, this suggests that GliNS1 expresses active forms of GSK3 and is also BIO-responsive.

Therefore, BIO-mediated GSK3 inhibition may represent one strategy for perturbing various

signaling pathways to gain functional insight into BTSC regulation and function.

Page 68: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

58

Figure 14 – GliNS1 expresses Wnt pathway components and is BIO-responsive in vitro.

Brain tumour stem cells express Wnt pathway components and activate TCF/LEF—

mediated transcription in response to GSK3 inhibition. a) RT-PCR for various components

reveal that BTSC lines express various Frizzled receptors (1, 3, 6, 7, 8. 9), Low density

lipoprotein related-receptor protein 5 (Lrp5), Dickkopf 1 (Dkk1), and the central

components β-catenin and glycogen synthase kinase 3β (GSK3β) at the mRNA level.

GAPDH was used as a loading control. b) An immunoblot of three BTSC lines for activated

forms of GSK3α and/or GSK3β (Y279 and Y216, respectively) and total β-catenin. The

adenocarcinoma line SW480 was used as a control for β-catenin. β-actin was used as a

loading control. c) TOPFlash assay for GliNS1 BIO-responsiveness. GliNS1 cells were

treated with BIO (1µM) for 24 hours (or fresh media for endogenous levels) and TOP/FOP

levels were compared in the above bar graph. All TOP and FOP readings are normalized

to the Renilla luciferase output for transfection control.

Page 69: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

59

3.2.2 BIO induces neuronal differentiation of brain tumour stem cells

Having confirmed that GliNS1 responds to BIO through the activation of TCF/LEF transcription

similarly to hNSCs, we wanted to determine whether BIO also induces neuronal differentiation

in a highly mitogenic environment. We cultured GliNS1 in EGF and FGF in the presence or

absence of BIO for 14 days and analyzed each population for progenitor and lineage-specific

markers using both immunofluorescence and intracellular flow cytometry. Immunofluorescence

for precursor markers revealed that there was a noticeable decrease in nestin and sox2 expression

in BIO-treated GliNS1 relative to untreated cultures (Figure 15a). These changes were quantified

using intracellular flow cytometry, confirming that BIO-treatment led to a 0.637-fold and 0.627-

fold decrease in the number of nestin and sox2 expressing cells compared to untreated GliNS1

grown under identical conditions (Figure 15b). What this may imply is that BIO is inducing

GliNS1 cells to exit the precursor state and differentiate, which can be characterized by the

expression of more mature lineage-specific markers. Given this possibility, we also analyzed

BIO-treated and untreated cells for neuronal and astrocytic markers using both

immunofluorescence and intracellular flow cytometry. Immunofluorescence for βIII-tubulin

revealed that in the presence of EGF and FGF, BIO-treated GliNS1 showed a marked increase in

neuronal differentiation relative to the untreated GliNS1 population, as evidenced by the increase

in βIII-tubulin-positive cells (Figure 16a). Furthermore, intracellular flow cytometry was used to

quantify the change in neuronal marker expression and found that the number of GliNS1 cells

expressing βIII-tubulin significantly increased by 4.797-fold with BIO treatment (Figure 16b).

Furthermore, while only a few GliNS1 cells express GFAP in the presence of EGF and FGF,

BIO treatment reduced the number of GFAP-positive cells present in culture to 0.236-fold of the

untreated levels (Figure 16a,b). Together, this data supports our prediction that BIO-mediated

GSK3 inhibition leads to the depletion of the Sox2+/Nestin+ precursor population and the

simultaneous induction of neuronal differentiation of GliNS1 BTSCs, even in highly mitogenic

stem cell conditions.

Page 70: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

60

Figure 15 – BIO reduces GliNS1 precursor marker expression in EGF and FGF.

GliNS1 exhibit decreased precursor marker expression with BIO treatment. a)

Immunofluoescence for nestin (Red) and sox2 (green). GliNS1 treated with BIO (1µM) for

14 days in the presence of EGF and FGF produced fewer nestin- and sox2-expressing cells

relative to untreated GliNS1, as revealed by immunofluorescence. b) Intracellular flow

histograms confirmed the change in nestin and sox2 expression with BIO treatment,

revealing a 0.637- and 0.627-fold decrease in the number of nestin-positive and sox2-

positive cells, respectively, relative to untreated cells. Unstained, red; untreated, green;

BIO-treated, blue (n=3; paired student’s T-test).

Page 71: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

61

Figure 16 – BIO treatment induces neuronal differentiation of GliNS1 in EGF and FGF.

BIO induces neuronal differentiation of GliNS1. a) GliNS1 treated with BIO (1µM) for 14

days in the presence of EGF and FGF produced a greater proportion of βIII-tubulin+ (red)

cells and fewer GFAP+ (green) cells compared to their untreated control, as assessed by

immunofluorescence. b) Intracellular flow cytometry histograms confirmed that BIO-

treated GliNS1 produced 4.797-fold more βIII-tubulin+ cells and a 0.236-fold decrease in

GFAP-expressing cell. Unstained, red; untreated, green; BIO-treated, blue (n=3; paired

student’s T-test).

Page 72: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

62

3.2.3 BIO treatment induces brain tumour stem cells to exit the cell cycle and decrease proliferation

The ultimate goal of any chemotherapy is to kill or arrest the growth of the tumour cells.

Therefore, we wanted to know whether BIO-induced lineage commitment was potentially

associated with terminal differentiation, which would be marked by cell cycle exit. Forcing

neuronal lineage commitment could be a viable treatment strategy if these cells terminally exited

the cell cycle and could therefore no longer contribute to clonal expansion and tumor bulk. In

order to address the affect of BIO on bulk proliferation rate of GliNS1, we performed an MTT

assay over a period of 14 days in EFG and FGF and in the presence or absence of BIO. While no

significant difference was detected at earlier time points, proliferation differed significantly by

Day 14, suggesting a greater proportion of cells are exiting the cell cycle when exposed to BIO-

mediated GSK3-inactivation long-term (Figure 17a). To confirm that cells were exiting the cell

cycle by day 14, we treated GliNS1 with BIO and performed a BrdU pulse for 24-hours 6 days

and 13 days after treatment began to mark cells going through S-phase of the cell cycle in that

time period. We then stained for BrdU and found that after 7 days of BIO treatment, there was no

difference in the total number of BrdU+ cells between the BIO-treated and untreated BTSCs

(BIO: 77.39% versus untreated: 78.56%), confirming what we observe in our MTT assay (Figure

17b). After 14 days of BIO treatment, we observed a 22% reduction in the number of BrdU+

cells compared to our untreated GliNS1 cultures (BIO: 32.53% versus Untreated: 54.11%)

(Figure 17b), revealing that prolonged treatment of BIO results in increased proportion of cells

undergoing cell cycle arrest, contributing to an observed decrease in proliferation.

Together with earlier findings that BIO treatment for a period of 14 days results in a reduction in

precursor marker expression and an increase in the neuronal lineage marker, βIII-tubulin, results

suggest that a proportion of GliNS1 exits the cell cycle and terminally differentiate down the

neuronal lineage to produce a more quiescent population of cells. Therefore, the inhibition of

GSK3, possibly through the promotion of canonical Wnt signaling, may offer a novel

mechanism for differentiation therapy of GBMs.

Page 73: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

63

Figure 17 – BIO treatment promotes cell cycle exit and decreased proliferation.

Long-term BIO treatment promotes BTSCs to exit the cell cycle. a) MTT assay performed

over a period of 14 days for GliNS1 grown in EGF and FGF with/without BIO (1µM) (***:

p≤0.001) (unpaired student’s T-test). b) GliNS1 grown in EGF and FGF with/without BIO

(1µM) for 7 and 14 days, then pulsed with BrdU (10µg/ml) for 24 hours prior to analysis.

There was a 22% reduction in the number of BrdU+ cells at day 14, indicating a trend

toward cell cycle exit when GliNS1 is treated with BIO (unpaired student’s T-test).

Page 74: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

64

Chapter 4

4 Discussions and Future Directions

4.1 Wnt signaling in human neural stem cells

Several groups have reported an in vivo expansion of the murine neural precursor pool at the

expense of neuronal differentiation in response to constitutive or targeted activation of β-

catenin—mediated signaling120,149,150. However, other studies also in mice have shown that Wnt

signaling directs neuronal differentiation of neural progenitors, both in vitro and in vivo117,118.

The lack of consensus likely stems in part from the fact that Wnt is highly context-specific,

exerting varied effects depending on the receiving cell, developmental timing, and the other

confounding signals present within the microenvironment. Several questions remain unresolved

that could help reconcile the inconsistencies observed within the field. For one, it remains

unclear where Wnt is acting within the cellular hierarchy. Likely, Wnt regulates most cellular

populations along the hierarchy, but in context-specific ways. For example, it is possible that

Wnt promotes self-renewal of the true stem cell under the appropriate circumstances. However,

committed progenitors within the same environment may interpret this signal as an instruction to

differentiate into a neuron. Moreover, if the microenvironment changes, Wnt may cooperate with

other signals to alter the cell fate decisions of the receiving cell. For example, Israsena et al.

found that the presence of FGF2 determined whether β-catenin effects proliferation or whether it

promotes neuronal differentiation119.

A second reason for the lack of consensus is that the parameters of the experiment dictate the

results we observe. For example, the seminal study by Chenn and Walsh (2002) involves the

expression of constitutively active β-catenin (ΔN90) under the control of the nestin second intron

enhancer, which has been shown to be active by E10 in a diverse group of cells that exhibit

heterogeneity in their transcription factor repertoire120,151. However, nestin expression is

detected as early as E7.75 in many proliferative regions of the developing mouse CNS152.

Therefore, ΔN90 is likely introduced into a stem cell compartment that is already

developmentally locked in a proliferative expansion program that precedes the neurogenic burst

beginning around E10.5153. A similar explanation can be attributed to the findings reported in the

study by Kim et al. (2009), GSK3 deleted under the same nestin promoter resulted in

Page 75: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

65

hyperproliferation of neural progenitors and suppressed neuronal differentiation149. One possible

explanation for both these findings is that stabilized β-catenin is introduced into a compartment

of cells already predisposed to proliferation or symmetrical expansion. A study by Hirabayashi et

al. show this by dissecting NPCs from the cortices of E10.5 and E13.5 mice and infecting them

with stabilized β-catenin (S33Y β-catenin)118. After 2 days in the presence of FGF2, E10.5 NPCs

showed a decrease in Tuj1+ (βIII tubulin) cells relative to the control-infected NPCs, while E13.5

NPCs infected with S33Y β-catenin resulted in an almost 4-fold increase in Tuj1+ cells.

Therefore, the developmental age of the NPCs exposed to stabilized β-catenin greatly affects

how they respond154. In other words, the parameters of the experiments dictate the observed

phenotypes. A complete in vivo time course is necessary to understand the role of β-catenin—

mediated signaling at each developmental stage. One way to address this is to create an inducible

system where Wnt signaling can be perturbed in a controlled and specified manner in vivo. Both

the Estrogen Receptor (ER)-tamoxifen and tetracycline (tet)-inducible systems are commonly

used to control spatial and temporal targeting of specific perturbations155,156. This would provide

greater insight into how Wnt signaling regulates CNS development in a spatial-temporal manner.

While a consensus remains to be reached in the murine system, less is known about how these

trends hold up in the human CNS, and what role canonical Wnt signaling plays in hNSCs. The

data from mice may not hold up in humans because of marked differences in development, such

as cell cycle control and sheer differences in generation of numbers of mature cell types. For

example, the murine neocortex is formed over a 6-day period and requires about 11 cell cycles

whereas neurogenesis in the human neocortex requires about 34 cell cycles and occurs over a

period of approximately 120 days, representing the exponential expansion of the human cerebral

cortex157. Several limitations have hindered our ability to interrogate the functional regulation of

hNSCs. For one, access to human CNS tissue can be challenging, particularly obtaining growth

of precursors from postnatal brain. Second, in vivo experiments are virtually impossible for

obvious reasons. Third, neural development in the human CNS does not occur in a linear fashion,

nor is it as well characterized as the murine system (although progress is being made)158. Much

of what is known is inferred from studies in the mouse, or from some studies of nonhuman

primates159. Therefore, very few studies focus on Wnt in human neural precursors.

In this particular study, we hoped to gain insight into the role of β-catenin—mediated signaling

in hNSCs, particularly how it regulates cell fate choices. Using cell biology, molecular genetics,

Page 76: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

66

and chemical biology, we activated canonical Wnt signaling and interrogated the functional

implications of this activation on hNSCs. By all three methods, our data suggests that activating

the canonical Wnt signaling pathway results in a neuronal cell fate choice during differentiation.

Interestingly, however, ΔN90 and Wnt3a were only able to promote a neuronal lineage in

differentiating conditions, but were unable to induce neuronal differentiation under stem cell

conditions (data not shown). Wnt signaling did affect lineage choice in the absence of EGF and

FGF, confirming that Wnt signaling does play a role in neuronal cell fate choice. The context

may be reminiscent of how Wnt signaling might regulate neural precursors in vivo, promoting a

pro-neuronal program in progenitors that migrate away from the stem cell niche and the grips of

potent mitogenic signals. BIO-mediated GSK3 inhibition was able to induce mild neuronal

differentiation in stem cell conditions. However, nestin showed no dramatic change, indicating

that these neurons likely have not terminally differentiated. This suggests that Wnt signaling may

not be sufficient to override EGF and FGF, but GSK3-inhibition may interfere with these and

several other pathways in cooperation with Wnt activation, enabling a switch in cell fate, even in

stem cell conditions. Therefore, the effects of β-catenin-mediated signaling probably depend

heavily on other factors simultaneously present within the microenvironment.

This can also be said about the role of Wnt signaling in proliferation in our study. Unlike what

was reported in the literature for murine NSCs120,160, neither stabilized β-catenin nor Wnt3a was

able to alter proliferation in stem cell and differentiating conditions. Perhaps Wnt must cooperate

with other signals from the niche in vivo to promote proliferation, and the loss of a niche in vitro

removes a context that favours proliferation. A second possibility may be that hNSC

proliferation is not governed primarily by canonical Wnt signaling. The pro-proliferative

transcription factor c-Myc, a known target of Wnt signaling, is also a target of FGF/PI3K

signaling149. It is conceivable that the FGF activation of c-Myc transcription saturates c-Myc

levels, and TCF/LEF—transcriptional activation of c-Myc results in minimal contribution to

overall c-Myc transcription in the cells, resulting in little change in proliferation. Or finally,

perhaps the bulk proliferation rate for the entire population did not change, but rather a shift

occurred where committed neuronal progenitors (instead of multipotent stem cells) expanded in

response to Wnt activation, giving rise to the post-mitotic neurons seen at day 21. This may

explain why we see no change in overall proliferation for the first two weeks (when MTTs were

Page 77: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

67

performed), but after 21 days we have a more quiescent terminally differentiated neuronal

population relative to the control population.

BIO-mediated inhibition of GSK3 resulted in a significant decrease in proliferation of hNSCs

both in differentiating or stem cell conditions. While BIO does mimic Wnt signaling by

inactivating GSK3, it also affects other pathways that converge on this multi-tasking kinase,

including Notch, Shh, and PI3K/Akt pathways, all of which have been shown to influence NSC

function79,161,162. It is quite likely that a concerted effort among these pathways is required to

regulate hNSC proliferation. To address the extent to which other pathways contribute to control

proliferation, rescue experiments targeting each independent pathway should be performed to

identify cooperating signaling mechanisms involved in the process. Another reason may be some

off target effects of BIO on various CDK/cyclin pairs that may down-regulate cell cycle

reentry104, however this has yet to be confirmed as well.

What is interesting is that while GSK3-inhibition was able to significantly decrease proliferation

in stem cells conditions, it was only able to induce a mild neuronal differentiation in hNSCs.

Therefore, it is probable that Wnt plays a role in specifying a neuronal fate, but may require the

cooperation of several other signals to exert its effects. Similarly, while Wnt may promote

neuronal fate choice, it may not directly regulate proliferation, again pointing to the possibility

that although these processes are often considered to be coupled functionally, various and

differing signals may govern each process somewhat exclusively. Nevertheless, results indicate

that GSK3 plays a key role in regulating the balance between hNSC proliferation and neuronal

differentiation.

Though our study suggests that GSK3 is a negative regulator of neuronal differentiation, elegant

work by Kim et al. showed that elimination of GSK3α/β in the murine CNS results in the

expansion of the neural precursor pool and a decrease in neurogenesis in vivo149. One

explanation for the observed discrepancy is that perhaps the role for GSK3 in hNSCs differs

from its role in mNSCs at various stages in development. Also, while we did not extend our

investigation beyond canonical Wnt signaling, Kim et al. show that Notch, Shh, and FGF/PI3K

signaling also contribute to the observed neural precursor expansion, suggesting that other major

players converge upon GSK3 to regulate neural precursor activity in vivo. When we treat hNSCs

with a gamma secretase inhibitor (DAPT) that inhibits Notch signaling in combination with BIO,

Page 78: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

68

we observe synergistic cooperation resulting in increased neuronal differentiation and a decrease

in BrdU incorporation (data not shown). Therefore, while our findings concerning Notch

signaling agree with Kim et al.’s findings, we find that chemical inactivation of GSK3 induces

the opposite effect, promoting neuronal differentiation. Discrepancies may also lie in the fact that

we are employing a small molecule inhibitor to target GSK3 while Kim et al. have deleted GSK3

genetically under the second enhancer of the human nestin gene149,152,163. This particular

regulatory element targets neuroepithelial (NE) cells in a developmental stage when they are

predisposed to proliferative expansion to develop the SVZ. In humans, neurogenesis begins

around E33 and unlike rodents, continues throughout fetal development only to subside

perinatally153. Our in vitro hNSC lines are established from GW 8-11 (E56-E77) fetal CNS, a

gestational period characterized by the formation of a distinct SVZ, widespread cortical

neurogenesis and histological organization of the CNS structures164. While in culture our hNSC

lines are multipotent, it is possible that these cells were not derived from the quiescent stem cell

population in vivo, but rather were neurogenic precursors that reacquired multipotency in vitro

due to EGF exposure, a phenomenon previously observed with with murine transit-amplifying

cells in culture11. These EGF-responsive transit-amplifying neurogenic precursors have been

localized to the SVZ in mice, and are likely derived from the SVZ in humans as well165.

Therefore, one can presume that our hNSC lines are comprised of a heterogeneous population of

quiescent NSCs and converted multipotent neurogenic transit-amplifying neurogenic progenitors

derived from the expanded SVZ found in the human fetal CNS. Therefore, unlike previous

studies, our populations are likely developmentally more mature and are no longer locked in the

expansion phase when they are perturbed for Wnt signaling, possibly explaining why other

observed expansion of the VZ/SVZ zones, while we observe neurogenesis in response to GSK3-

inhibition149.

Results suggest that Wnt acts in concert with other signaling pathways to tightly regulate hNSCs

function, likely in a spatial-temporal specific manner. Harnessing our knowledge of these

intricate regulatory mechanisms enables us to develop novel pharmacologic strategies, such as in

vivo neuronal replacement therapy through the forced differentiation of hNSCs to treat various

neurodegenerative diseases. Chemical inhibition of GSK3/β-catenin stabilization in ventral

midbrain murine neural precursors results in the production of TH+ dopaminergic neurons,

indicating that Wnt signaling can induce neuronal differentiation of specific classes of

Page 79: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

69

neurons166. Further investigation is required to determine whether Wnt stimulates the production

of specific neuronal subtypes or whether it selects for the survival of specific subtypes, not to

mention how it plays a role in specifying human neuronal subclasses. Neuronal specificity is of

great importance as different neuronal subtypes are implicated in different psychiatric

diseases167. For example, serotonergic neurons have been implicated in depression while

dopaminergic neurons have been widely implicated in the pathobiology of schizophrenia.

Furthermore, the canonical Wnt signaling pathway has been implicated in Alzheimer’s Disease

(AD). Several in vivo studies in mice showed that activation of Wnt by GSK3 inhibition showed

a reduction in β-amyloid (Aβ) peptide production, the leading neuropathological characteristic of

AD168,169. Therefore, addressing these outstanding questions in future studies can lead to novel

therapeutic potential in the form of cell-specific replacement therapy. It also offers new tools for

in vitro drug screens to uncover new drugs that target specific neuronal subtypes for the

treatment of various neurological disorders and diseases.

4.2 Targeting the Wnt pathway for differentiation therapy of brain tumour stem cells

Following pioneering work by the Dick and Clarke laboratories identifying leukemic24 and breast

cancer stem cells26, respectively, Singh et al. prospectively identified and isolated brain tumour

stem cells (BTSCs) by the cell surface antigen CD13325. It was shown that cells within the bulk

of the brain tumour were functionally heterogeneous, and that a subpopulation of the cells, the

CD133+ population, was enriched for tumourigenic potential while the remaining CD133- cells

were unable to initiate tumour formation in vivo. BTSCs are characterized by their ability to self-

renew, exhibit a differentiation capacity phenotypically identical to the original patient tumour,

and in vivo tumour initiating potential. While CD133 may mark the CSC population of a subset

of brain tumours, other groups have also identified SSEA-1/CD15 as tumour-initiation enriching

marker in brain tumours, hinting that each brain tumour may possess phenotypically and

functionally unique tumour-initiating populations49. Tied into BTSC heterogeneity is the

molecular heterogeneity of GBM subclasses. Using integrated genomic analysis has unraveled

four distinct molecular subclasses of GBMs that harbour distinct genomic alterations and

respond differently to clinical treatment170,171. Given the heterogeneity observed within tumours

as well as among them, in vitro drug screens of patient-specific BTSC lines and personalized

Page 80: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

70

medicines seem like the only possible avenue for true therapeutic advancement in the treatment

of gliomas.

BTSC lines have been established in serum-free conditions that mimic the culturing conditions

of hNSC lines established within the Dirks laboratory172,144. Along with the ease of manipulation,

these lines retain patient-specific phenotypes, making them a valuable and reliable model of the

human disease for genetic and chemical screens. Moreover, BTSCs and hNSCs show remarkable

similarities. Both populations can be propagated in vitro as monolayers (or neurospheres) in the

presence of growth factors, both possess the ability to self-renew, and both exhibit a range of

multipotent differentiation. Furthermore, microarray and principle component analysis reveals

that hNSCs and BTSCs cluster tightly together with a stem cell signature, but differ significantly

from human normal cortex. Given the similarities, understanding the basic biology of hNSCs can

provide insight into the biology of BTSCs and targetable mechanisms that represent therapeutic

avenues for treating brain tumours. Along this line of logic, we attempted to translate the

knowledge gained in interrogating Wnt signaling in hNSCs and investigate the potential for

GSK3 as a novel target for differentiation therapy for a subset of gliomas.

While this study is preliminary and restricted to the GliNS1 BTSC line, data suggest that GSK3-

inhibition results in the depletion of Sox2+/Nestin+ cells and the production of βIII-tubulin+

neurons. This is encouraging as neurons are thought to be a more quiescent cell type compared to

mature astrocytes, a cell type commonly found in gliomas157,45. Furthermore, over a 2-week

period of BIO-treatment, BTSCs showed decreased proliferation and 22% less BrdU

incorporation relative to their untreated counterparts, suggesting that BIO treatment resulted in

fewer cells undergoing mitosis. Importantly, BrdU has also been shown to incorporate into cells

undergoing DNA synthesis during DNA damage repair processes173. Therefore, another

interpretation is that BrdU is marking cells undergoing DNA damage repair and possibly cell

death, implying that BIO treatment may have function as a pro-survival treatment given the

decrease in cells undergoing DNA damage and apoptosis. In order to rule out this possibility, it

will be important to perform co-staining with BrdU and other markers of cell division, such as

Ki67 and pHH3. Inducing cell cycle arrest or terminal differentiation may be an important

feature of chemotherapy since intracranial pressure due expansion of tumour bulk is one the most

detrimental features of brain cancer.

Page 81: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

71

While the Wnt pathway has been implicated in the pathobiology of medulloblastomas related to

Turcot’s syndrome174, other studies show that activated Wnt signaling predicts a favourable

outcome in sporadic childhood medulloblastomas175. However, little has been reported about the

Wnt signaling pathway in gliomas. GSK3, a central mediator of the Wnt pathway, has received

increasing attention in the brain tumour field. A recent study, investigates Bmi and GSK3 in

serum-derived glioma lines and found that inhibiting GSK3β by siRNA or chemical inhibition

using LiCl and SB216763 resulted in a reduction in clonogenicity as well as the depletion of

stem cell markers and an increase GFAP+, CNPase+ and βIII-tubulin expressing cells176. It was

also found that down-regulation of Bmi1, a member of the polycomb group of proteins involved

in stem cell maintenance, seemed to coincide with a decrease in GSK3β expression, suggesting a

link between these two important molecules. These findings suggest that Bmi1, which has

already been shown to maintain BTSC self-renewal, may operate in a GSK3β-dependent

manner177. Furthermore, GSK3-inhibition has been shown to induce cell death of serum-derived

tumour lines, implicating GSK3 in the regulation of glioma cell survival178.

PTEN regulates many cellular processes, including proliferation and survival, primarily by

inhibiting the PI3K pathway, and is commonly deleted or down-regulated in glioblastomas179.

Recently it has been shown that GSK3—mediated phosphorylation on threonine 366 of the

PTEN protein results in its destabilization and functional inhibition, and that inhibition of GSK3

can stabilize PTEN levels in vitro161. This provides another link between GSK3 and another

major culprit often implicated in the pathobiology of glioblastomas.

Just as it is possible that Wnt signaling must act in concert with other pathways to exert cell

cycle arrest and subsequent neuronal cell fate choice, one can surmise that BIO-mediated GSK3

inhibition may exert its differentiation effects simultaneously through various pathways in

BTSCs. Understanding the molecular mechanisms governing this phenomenon will be

paramount in identifying possible novel treatments intended to induce terminal differentiation

and cell cycle exit of BTSCs. Future studies should focus on the possible interaction between

GSK3 and classic pathways altered in glioblastomas (i.e. PTEN, pRb, EGFRvIII, etc).

Furthermore, GSK3-inhibiting agents should be included in chemical and genetic screens

performed on patient-specific BTSC lines in order to assess the therapeutic potential of targeting

this kinase. Preliminary screens are currently underway in the Dirks laboratory. And finally,

GSK3 inhibitors such as BIO should be tested in vivo in mouse models that present with gliomas

Page 82: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

72

to determine whether GSK3 is a realistic therapeutic target for treatment. Interestingly, lithium

use—a known GSK3 inhibitor often used to treat mood disorders—correlates with decreased

brain tumour incidence, suggesting that preemptive targeting of GSK3 may protect against brain

tumour initiation180. Together, our findings and other studies suggest that GSK3 is implicated in

the regulation of glioma stem cell biology, and may reveal a novel target for chemotherapy for a

subset of glioblastomas.

Page 83: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

73

References

1Crosnier, C., Stamataki, D., & Lewis, J. (2006). Organizing cell renewal in the intestine: stem cell, signals, and combinatorial control. Nat. Rev. Gen. 7, 349-359. 2 Reya, T., Morisson, S.J., Clarke, M.F., & Weissman, I.L. (2001). Stem cells, cancer, and cancer stem cells. Nature 414, 105-111. 3 Jones, D.L., & Wagers, A.J. Wagers (2008). No place like home: anatomy and function of the stem cell niche. Nat. Rev. Mol. Cell Biol. 9, 11-21. 4 Rossant, J. (2008) Stem cells and early lineage development. Cell 132, 527-531. 5 Mimeault, M., Hauke, R., Batra, S.K., (2007). Stem cells: a revolution in therapeutics—recent advances in stem cell biology and their therapeutic applications in regenerative medicine and cancer therapies. Clin. Phar. & Ther. 18(3), 252-264. 6 Fuchs, E. (2009). The tortoise and the hair: slow-cycling cells in the stem cell race. Cell 137, 811-819. 7 Reynolds, B.A., & Weiss, S. (1992). Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system. Science, 225, 1707-1710. 8 Weiss, S., Reynolds, B.A., Vescovi, A.L., Morshead, C., Craig, C.G., van der Kooy, D. (1996). Trends Neurosci. 19, 387-393. 9 Lledo, P.-M. , Alonso, M., Grubb, M. S. (2006). Adult neurogenesis and functional plasticity in neuronal circuits. Nat. Rev. Neurosci. 7, 179-193. 10 Zhao, C., Deng, W., & Gage, F. H. (2008). Mechanisms and functional implications of adult neurogenesis. Cell 132, 645-660. 11 Doetsch, F., Petreanu, L., Caille, I., Garcia-Verdugo, J.-M. & Alvarez-Buylla, A. (2002). EGF converts transit-amplifying neurogenic precursors in the adult brain into multipotent stem cells. Neuron 36, 1021-1034. 12 Doetsch, F., Caille, I., Lim, D.A., Garcia-Verdugo, J.M., & Alvarez-Buylla, A. (1999). Subventricular zone astrocytes are neural stem cells in the adult mammalian brain. Cell 97, 703-716. 13 Mirzadeh, Z., Merkle, F.T., Soriano-Navarro, M., Garcia-Verdugo, J.M., & Alvarez-Buylla, A. (2008) Neural stem cells confer unique pinwheel architecture to the ventricular surface in neurogenic regions of the adult brain. Cell Stem Cell 3, 265-278. 14 Miller, F.D. & Gauthier-Fisher, A. (2009). Home at last: neural stem cell niches defined. Cell Stem Cell 4, 507-510. 15 Temple, S. (2001). The development of neural stem cells. Nature 414, 112-117.

Page 84: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

74

16 Reynolds, B.A., Tetzlaff, W. & Weiss, S. (1992). A multipotent EGF-responsive Striatal embryonic progenitor cell produces neurons and astrocytes. J. Neurosci. 12(11), 4565-4574. 17 Lee, J., Kotliarova, S., Kotliarov, Y., Li, A., Su, Q., Donin, N.M., Pastorino, S., Purow, B.W., Christopher, N., Zhang, W., Park, J.A. & Fine, H.A. (2006). Tumour stem cells derived from glioblstomas cultured in bFGF and EGF more closely mirror the phenotype and genotype of primary tumors than do serum-cultured cell lines. Cancer Cell 9, 391-403. 18 Kuhn, H.G., Winkler, J., Kempermann, G., Thal, L.J., & Gage, F.H. (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. 19 Zhoa, M., Li, D., Shimazu, K., Zhou, Y.X., Lu, B., & Deng, C.X. (2007). Fibroblast growth factor receptor-1 is required for long term proliferation, memory consolidation, and neurogenesis. Biol. Psychiatry 62, 381-390. 20 Hitoshi, S., Alexson, T., Tropepe, V., Donoviel, D., Elia, A.J., Nye, J.S., Conlon, R.A., Mak, T.W., Bernstein, A., & van der Kooy, D. (2002). Notch pathway molecules are essential for the maintenance, but not the generation, of mammalian neural stem cells. Genes & Dev. 16, 846-858. 21 Hirabayashi, Y. & Gotoh, Y. (2005). Stage-dependent fate determination of neural precursor cells in mouse forebrain. Neurosci. Res. 51, 331-336. 22 Sun, Y., Nadal-Vicens, M., Misono, S., Lin, M.Z., Zubiaga, A., Hua, X., Fan, G., Greenberg, M.E. (2001). Neurogenin promotes neurogenesis and inhibits glial differentiation by independent mechanisms. Cell 104, 365-376. 23 Kamakura, S., Oishi, K., Yoshimatsu, T., Nakafuku, M., Masuyama, N., & Gotoh, Y. (2004). Hes binding to STAT3 mediates crosstalk between Notch and JAK-STAT signaling. Nat. Cell Biol. 6(6), 547-554. 24 Bonnet, D. & Dick, J.E. (1997). Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat. Med. 3, 730-37. 25 Singh, S.K., Hawkins, C., Clarke, I.D.,Squire, J.A., Bayani, J., Hide, T., Henkelman, R.M., Cusimano, M.D. & Dirks, P.B. (2004). Identification of human brain tumours initiating cells. Nature, 432, 396-401. 26 Al-Hajj, M., Wicha, M.S.m Benito-Hernandez, A., Morrison, S.J., & Clarke, M.F. (2003). Prospective identification of tumourigenic breast cancer cell. Proc. Natl. Acad. Sci. USA. 100, 3983-3988. 27 Ricci-Vitiani, L., et al. (2006). Identification and expansion of human colon-cancer-initiating cells. Nature. 445, 111-115. 28 Schatton, T., et al. (2008). Identification of cells initiating human melanomas. Nature. 451, 345-349.

Page 85: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

75

29 Lerner, B.H. (2004). Sins of omission—cancer research without informed consent. N. Engl. J. Med. 351, 628-630. 30 Brunschwig, A., Southam, C.M., & Levin, A.G. (1965). Host resistance to cancer. Clinical experiments by homotransplants, autotransplants, and admixture of autologous leukocytes. Ann. Surg. 162, 416-425. 31 Ward, R.J. & Dirks, P.B. (2007). Cancer stem cells: at the headwaters of tumour development. Annu. Rev. Pathol. Mech. Dis. 2, 175-189. 32 Donnenberg, V.S, Donnenberg, A.D. (2005). Multiple drug resistance in cancer revisited: the cancer stem cell hypothesis. J. Clin. Pharmacol. 45, 872–877. 33 Boa, S., Wu, Q., McLendon, R.E., Hao, Y., Shi, Q., Hjelmeland, A.B., Dewhirst, M.W., Bigner, D.D. & Rich, J.N. (2006). Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 444, 756-760. 34 Dalerba, P., Cho, R.W., Clarke, M.F. (2007). Cancer stem cells: models and concepts. Annu. Rev. Med. 58, 267-284. 35 Singh, S.K., Clarke, I.D., Hide, T. & Dirks, P. (2004a). Cancer stem cells in nervous system tumors. Oncogene 23, 7267-7273. 36 O’Brien, C.A., Pollett, A., Gallinger, S., & Dick, J.E. (2006). A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature. 445, 106-110. 37 Hermann, P.C., et al. (2007). Distinct populations of cancer stem cells determine tumour growth and metastatic activity in human pancreatic cancer. Cell Stem Cell. 1, 313-323. 38Boiko, A. D., et al. (2010). Human melanoma-initiating cells express neural crest nerve growth factor receptor CD271. Nature 466, 133-137. 39 Wu, C., et al. (2007). Side population cells isolated from mesenchymal neoplasms have tumour initiating potential. Cancer Res. 67, 8216-8222. 40 Hansforth, L.M., et al. (2007). Neuroblastoma cells isolated from bone marrow metastases contain a naturally enriched tumour-initiating cell. Cancer Res. 67, 11234-11243. 41 Kim, C.F., et al. (2005). Identification of bronchioalveolar stem cells in normal lung and lung cancer. Cell. 121, 823-835. 42 Lapidot, T., Sidard, C., Vormoor, J., Murdoch, B., Hoang, T., Caceres-Cortes, J., Minden, M., Paterson, B., Caligiuri, M.A. & Dick, J.E. (1994). A cell initiating human acute myeloid leukemia after transplantation into SCID mice. Nature 367, 645-648. 43 Bhatia, M., Wang, J.C., Kapp, U., Bonnet, D. & Dick, J.E. (1997). Purification of primitive human hematopoietic cells capable of repopulating immune-deficient mice. Proc. Natl. Acad. Sci. USA 94, 5320-5325. 44 Fogarty, M.P., Kessler, J.D., & Wechler-Reya, R.J. (2005). Morphing into cancer: the role of developmental signaling pathways in brain tumour formation. J. Neurobiol 64, 458-475.

Page 86: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

76

45 DeAndelis, L.M. (2001). Brain Tumors. N. Engl. J. Med. 344, 114-123. 46 Uchida, N., Buck, D.W., He, D., Reitsma, M.J., Masek, M., Phan, T.V., Tsukamoto, A.S., Gage, F.H. & Weissman, I. (2000). Direct isolation of human central nervous system stem cells. Proc. Natl. Acad Sci. USA 97, 14720-14725. 47 Beier, D., Hau, P., Proescholdt, M., Lohmeier, A., Wischhusen, J., Oefner, P.J., Aigner, L., Brawanski, A., Bogdahn, U., & Beier, C.P. (2007). CD133+ and CD133- glioblastoma-derived cancer stem cells show differential growth characteristics and molecular profiles. Cancer Res. 67, 4010–4015.

48 Ogden, A.T., Waziri, A.E., Lochhead, R.A., Fusco, D., Lopez, K., Ellis, J.A.,

Kang, J., Assanah, M., McKhann, G.M., Sisti, M.B., McCormick, P.C., Canoll, P. & Bruce, J.N. (2008). Identification of A2B5+CD133- tumor-initiating cells in adult human gliomas. Neurosurg. 62, 505–514. 49 Son, M.J., Woolard, K., Nam, D., Lee, J. & Fine, H.A. (2009). SSEA-1 Is an Enrichment Marker for Tumor-Initiating Cells in Human Glioblastoma. Cell Stem Cell 4, 440-452. 50 The Cancer Genome Atlas Research Network (2008). Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455, 1061-1068. 51 Parsons, D.W., et al. (2008). An integrated analysis of human glioblastoma multiforme. Science 321, 1807-1812. 52 Zhu, Y. & Parada, L.F. (2002). The molecular and genetic basis of neurological tumours. Nat. Rev. Can. 2, 616-626. 53 Srivastava, S., Zou, Z. Q., Pirollo, K., Blattner, W. & Chang, E. H. (1990). Germ-line transmission of a mutated p53 gene in a cancer-prone family with Li–Fraumeni syndrome. Nature 348, 747–749. 54 Ohgaki, H., Dessen, P., Jourde, B., Horstmann, S., Nishikawa, T., Di Patre, P., Burkhard, C., Schuler, D., Probst-Hensch, N.M., Maiorka, P.C., Baez, N., Pisani, P., Yonekawa, Y., Yasargil, M.G., Mutolf, U.M. & Kleihues, P. (2004). Genetic pathways to glioblastoma: a population-based study. Cancer Res. 64, 6892-6899. 55 Zheng, H., et al. (2008). p53 and Pten control neural and glioma stem/progenitor self-renewal and differentiation. Nature 455, 1129-1134. 56 Ivanchuk, S.M., Mondal, S., Dirks, P.B. & Rutka, J.T. (2001). The INK4A/ARF locus: role in cell cycle control and apoptosis and implications for glioma growth. J. Neuro.-Oncol. 51, 219-229. 57 Meletis, K., Wirta, V., Hede, S., Nister, M., Lundeberg, J. & Frisen, J. (2005). p53 suppresses the self-renewal of adult neural stem cells. Development 133, 363-369. 58 Groszer, M., Erickson, R., Scripture-Adams, D., Dougherty, J.D., Le Belle, J., Zack, J.A., Geschwind, D.H., Lui, X., Kornblum, H.I. & Wu, H. (2006). PTEN negatively regulates neural

Page 87: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

77

stem cell self-renewal by modulating G0-G1 cell cycle entry. Proc. Natl. Acad. Sci. USA 103, 111-116. 59 Malanchi, I. & Huelsken, J. (2008). Cancer stem cells: never Wnt away from the niche. Curr. Opin. Oncol. 21, 41-46. 60 Gilbertson, R. (2004). Medulloblastoma: signaling a change in treatment. Lancet Oncol. 5, 209-218. 61 Bar, E.E., Chaudhry, A., Lin, A., Fan, X., Schreck, K., Matsui, W., Piccirillo, S., Vescovi, A., DiMeco, F., Olivi, A. & Eberhart, C.G. (2007). Cyclomamine-mediated hedgehog pathway inhibition depletes stem-like cancer cells in glioblastoma. Stem Cells 25(2), 2524-2533. 62 Piccirillo, S., Reynolds, B.A., Zanetti, N., Lamorte, G., Binda, E., Broggi, G., Brem, H., Olivi, A., Dimeco, F. & Vescovi, A. (2006). Bone morphogenetic proteins inhibit the tumourigenicity potential of human brain tumour-initiating cells. Nature 444, 761-765. 63 Fan, X., Matsui, W., Khaki, L., Stearns, D., Chun, J., Li, Y.M. & Eberhart, C.G. (2006). Notch pathway inhibition depletes stem-like cells and blocks engraftment in embryonal brain tumours. Cancer Res. 66, 7445-7452. 64 Purow, B.W., Haque, R.M., Noel, M.W., Su., Q., Burdick, M.J., Lee, J., Sundaresan, T., Pastorino, S., Park, J.K., Mikolaenko, I., Maric, D., Eberhart, C.G. & Fine, H.A. (2005). Expressionof Notch-1 and its ligands, Delta-like-1 and jagged-1, is critical for glioma cell survival and proliferation. Cancer Res. 65, 2353-2363. 65 Reya, T. & Clevers, H. (2005). Wnt signaling in stem cells and cancer. Nature 434, 843-850. 66 Jandial, R., U, H., Levy, M.L. & Snyder, E.Y. (2008). Brain tumour stem cells and the tumour microenvironment. Neurosurg. Focus 24, 1-6. 67 Moon, R.T., Bowerman, B., Boutros, M., & Perrimon, N. (2002). The promise and perils of wnt signaling through β-catenin. Science. 296, 1644-1646. 68 Van Amerongen, R. & and Berns, A. (2006) knockout mouse models to study Wnt signal transduction. Trends in Genetics 22, 678-689. 69 Mikels, A.J. & Nusse, R. (2006). Purified Wnt5a protein activates or inhibits β-catenin-TCF signaling depending on receptor context. PLoS Biol. 4, e115. 70 Clevers, H. (2006). Wnt/β-catenin signaling in development and disease. Cell 127, 469-480. 71 Zeng, X., Huang, H., Tamai, K., Zhang, X., Harada, Y., Yokoto, C., Almeida, K., Wang, J., Doble, B., Woodgett, J., Wynshaw-Boris, A., Hseih, J. & He, X. (2008). Initiation of Wnt signaling: control of Wnt coreceptor Lrp6 phosphorylation/activation via frizzled, dishevelled, and axin functions. Development 135, 367-375. 72 Salinas, P.C. & Ciani, L. (2005). Wnts in the vertebrate nervous system: from patterning to neuronal connectivity. Nat. Rev. Neurosci. 6, 351-362.

Page 88: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

78

73 Seto, E.S., Bellen, H.J. (2004). The ins and outs of wingless signaling. Trends in Cell Biol. 14, 45-53. 74 Hart, M.J., de los Santos, R., Albert, I.N., Rubinfeld, B. & Polakis, P. (1998). Downregulation of β-catenin by human axin and its association with APC, β-catenin and GSK3β. Curr. Biol. 8, 573-581. 75 Staal, F.J.L., van Noort, M., Strous, G.J. & Clevers, H.C. (2002). Wnt signals are transmitted through N-terminally dephosphorylated β-catenin. EMBO 3, 63-68. 76 The Wnt Homepage: http://www.stanford.edu/~rnusse/wntwindow.html. 77 Mikels, A.J. & Nusse, R. (2006). Purified Wnt5a protein activates or inhibits β-catenin-TCF signaling depending on receptor context. PLoS Biol. 4, 570-582. 78 Grigoriyan, T., Wend, P., Klaus, A. & Birchmeier, W. (2008). Deciphering the function of canonical Wnt signals in development and disease: conditional loss- and gain- of-function mutations of β-catenin mice. Genes & Dev. 22, 2308-2341. 79 Doble, B.W. & Woodgett, J.R. (2003). GSK-3: tricks of the trade for a multi-tasking kinase. J. Cell Sci. 116, 1175-1186. 80 Woodgett, J.R. (2001) Judging a protein by more than just its name: GSK-3. Science’s STKE, September 18, 2001. 81 Jia et al. (2002). Shaggy/GSK3 antagonizes hedgehog signaling by regulating Cubitus interruptus. Nature 416, 548-552. 82 Espinosa, L., Ingles-Esteve, J., Aquilera, C. & Bigas, A. (2003). Phosphorylation by glycogen synthase kinase-3β down-regulates Notch activity, a link for Notch and Wnt pathways. J. Biol. Chem. 278, 32227-32235. 83 Woodgett, J.R. (1991). cDNA cloning and properties of glycogen synthase kinase-3. Methods Enzymol. 200, 546-577. 84 Woodgett, J.R. (1990). Molecular cloning and expression of glycogen synthase kinase-3/factorA. EMBO J. 9, 2431-2438. 85 Hoeflich, K.P., Lu, J., Rubie, E.A., Tsao, M.S., Jin, O. & Woodgett, J.R. (2000). Requirement for glycogen synthase kinase-3beta in cell survival and NF-κB activation. Nature 406, 86-90. 86 O’Brien, W.T., Harper, A.D., Jove, F., Woodgett, J.R., Maretto, S., Piccolo, S. & klein, P.S. (2004). Glycogen Synthase Kinase-3β Haploinsufficiency mimics behavioral and molecular effects of lithium. J. Neurosci 24, 6791-6798. 87 Doble, B.W., Patel, S., Wood, G.A., Kockeritz, L.K. & Woodgett, J.R. (2007). Functional redundancy of Gsk3α and GSK3β in Wnt/β-catenin signaling shown by using an allelic series of embryonic stem cells lines. Dev. Cell 12, 857-971. 88 Cantley, L.C., PI3K Pathway. Science’s STKE (Connections Map, as seen April 2010), http://stke.sciencemag.org.myaccess.library.utoronto.ca/cgi/cm/stkecm;CMP_6557.

Page 89: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

79

89 Cross, D.A.E., Alessi, D.R., Cohen, P., Andjelkovich, M. & Hemmings, B.A. (1995). Inhibition of glycogen synthase kinase-3 by insulin mediated protein kinase B. Nature 378, 785-789. 90 Yuan, H., Mao, J., Li, L. & Wu, D. (1999). Suppression of glycogen synthase kinase activity is not sufficient for leukemia enhancer factor-1 activation. J. Biol. Chem. 274, 30419-20423. 91 Murai, H., Okazaki, M. & Kikuchi, A. (1996). Tyrosine dephosphorylation of glycogen synthase kinase-3 is involved in its extracellular signal-dependent inactivation. FEBS Lett. 392, 13-160. 92 Ter Haar, E., Coll, J.T., Austen, D.A., Hsiao, H.-M., Swenson, L. & Jain, J. (2001). Structure of GSK3beta reveals a primed phosphorylation mechanism. Nat. Struct. Biol. 8, 593-596. 93 Dajani, R., Fraser, E., Roe, S.M., Young, N., Good, V., Dale, T.C., Pearl, L.H. (2001). Crystal structure of glycogen synthase kinase 3 beta: structural basis for phosphate-primed substrate specificity and autoinhibition. Cell 105, 721-723. 94 Ikeda, S., Kishida, S., Yamamoto, H., Murai, H., Koyama, S. & Kikuchi, A. (1998). Axin, a negative regulator of the wnt signaling pathway, forsms a complex with GSK-3β-dependent phosphorylation of β-catenin. EMBO J. 17, 1371-1384. 95 Behrens, J., Jerchow, B.-A., Wurtele, M., Grimm, J., Asbrand, C., Wirtz, R., Kuhl, M., Wedlich, D. & Birchmeier, W. (1998). Functional interaction of an Axin homolog, conductin, with β-catenin, APC and GSK3β. Science 280, 596-599. 96 Sakanaka, C., Weiss, J.B. & Williams, L.T. (1998). Bridging of β-catenin and glycogen synthase kinase-3β by axin and inhibition of β-catenin-mediated transcription. Proc. Natl. Acad. Sci. USA 95, 3020-3023. 97 Yost, C., Farr, G.H., Pierce, S.B., Ferkey, D.M., Chen, M.M. & Kimmelman, D. (1998). GBP, an inhibitor of GSK-3, is implicated in Xenopus development and oncogenesis. Cell 93, 1031-1041. 98 Mao, Y., Ge., X., Frank, C.L., Madison, J.M., Koehler, A.N., Doud, M.K., Tassa, C., Berry, E.M., Soda, T., Singh, K.K., Biechele, T., Petryshen, T.L., Moon, R.T., Haggarty, S.J. & Tsai, L.-H. (2009). Disrupted in Schizophrenia 1 regulates neuronal progenitor proliferation via modulation of GSK3β/β-catenin signaling. Cell 136, 1017-1031. 99 Klein, P. & Melton, D. A. (1996). A molecular mechanism for the effect of lithium on development. Proc. Natl. Sci. USA 93, 8455-8459. 100 Stambolic, V., Ruel, L. & Woodgett, J.R. (1996). Lithium inhibits glycogen synthase kinase-3 activity and mimics wingless signaling in intact cells. Curr. Biol. 6, 1664-1668. 101 Diehl, J.A., Cheng, M., Roussel, M.F. & Sherr, C.J. (1998). Glycogen synthase kinase-3β regulates cyclinD1 proteolysis and subcellular localization. Genes Dev. 12, 3499-2511.

Page 90: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

80

102 Sato, N., Meijer, L., Skaltsounis, L., Greengard, P. & Brivanlou, A.H. (2004). Maintenance of pluripotency in human and mouse embryonic stem cells through activation of Wnt signaling by a pharmacological GSK-3-specific inhibitor. Nat. Med. 10, 55-63. 103 Lluis, F., Pedone, E., Pepe, S. & Cosma, M.P. (2008). Periodic activation of Wnt/β-catenin signaling enhances somatic cell reprogramming mediated by cell fusion. Cell Stem Cell 3, 493-507. 104 Meijer, L., Skaltsounis, A.-L., Magiatis, P., Polychronopoulos, P., Knockaert, M., Leost, M., Ryan, X.P., Vonica, C.A., Brivanlou, A., Dajani, R., Crovace, C., Taricone, C., Musacchio, A., Roe, S.M., Pearl, L. & Greengard, P. (2003). GSK-3-Selective inhibitors derived from Tyrian Purple Indirubins. Chem. Biol. 10, 1255-1266. 105 Kemler, R., Hierholzer, A., Kanzler, B., Kuppig, S., Hansen, K., Tekato, M.M., de Vries, W.N., Knowles B.B. & Solter, D. (2004). Stabilization of β-catenin in the mouse zygote leads to premature epithelial-mesenchymal transition in the epiblast. Development 131, 5817-5824. 106 Ying, Q., Wray, J., Nichols, J., Batlle-Morera, L., Doble, B., Woodgett, J., Cohen, P. & Smith, A. (2008). The ground state of embryonic stem cell self-renewal. Nature 453, 519-524. 107 Lin, L., Cui, L., Zhou, W., Dufort, D., Zhang, X., Cai., C.L., Bu, L., Yng, L., Martin, J., Kemler, R. et al. (2007). β-catenin directly regulates Islet1 expression in cardiovascular progenitors and is required for multiple aspects of cardiogenesis. Proc. Natl. Acad. Sci. 104, 9313-9318. 108 DasGupta, R. & Fuchs, E. (1999). Multiple roles for activated LEF/TCF transcription complexes during follicle development and differentiation. Development 126, 4557-4568. 109 Merrill, B.J., Gat, U., DasGupta, R. & Fuchs, E. (2001). Tcf3 and Lef1regulate lineage differentiation of multipotent stem cells in skin. Genes & Dev. 15, 1688-1705. 110 Korinek, V., Barker, N., Moerer, P., van Donselaar, E., Huls, G., Peters, P.J. & Clevers, H. (1998). Depletion of epithelia stem-cell compartments in the small intestine of mice lacking Tcf-4. Nat. Genet. 19, 1-5. 111 Barker, N., van Es, J.H., Kuipers, J., Kujala, P., van den Born, M., Cozijnsen, M., Haegebarth, A., Korving, J., Begthel, H., Peters, P.J. & Clevers, H. (2007). Identification of stem cells in the small intestine and colon by marker gene Lgr5. Nature 449, 1003-1008. 112 Zhao, C., Blum, J., Chen, A., Kwon, H.Y., Jung, S.H., Cook, J.M., Lagoo, A. & Reya, T. (2007). Loss of β-catenin impairs the self renewal of normal and CML stem cells in vivo. Cancer Cell 12, 528-541. 113 Cobas, M., Wilson, A., Ernst, B., Mancini, S.C., MacDonald, H.R., Kemler, R. & Radtke, F. (2004). β‐cateninisdispensableforhematopoiesisandlymphopoiesis.J.Exp.Med.199,221‐229.

Page 91: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

81

114 Koch, U., Wilson, A., Cobas, M., Kemler, R., MacDonald, H.R., & Radtke, F. (2008). Simultaneous loss of β- and γ-catenin does not perturb hematopoiesis or lymphopoiesis. Blood 111, 160–164. 115 Braun, M.M., Etheridge, A., Bernard, A., Robertson, C.P. & Roelink, H. (2003). Wnt signaling is required at distinct stages of development for the induction of the posterior brain. Development 130, 5579-5587. 116 Lee, M.K.S., Tole, S., Grove, E. & McMahon, A.P. (2000). A local Wnt3a signal is required for development of the mammalian hippocampus. Development 127, 457-467. 117 Lie, D., Colamarino, S.A., Song, H., Desire, L., Mira, H., Consiglio, A., Lein, E.S., Jessberger, S., Lansford, H., Dearie, A.R. & Gage, F.H. (2005). Wnt signaling regulates adult hippocampal neurogenesis. Nature 437, 1370-1375. 118 Hirabayashi, Y., Itoh, Y., Tabata, H., Nakajima, K., Akiyama, T., Masuyama, N. & Gotoh, Y. (2004). The Wnt/β/catenin pathway directs neuronal differentiation of cortical neural precursor cells. Development 131, 2791-2801. 119 Israsena, N., Hu, M., Fu, W., Kan, L. & Kessler, J.A. (2004). The presence of FGF2 signaling determines whether β-catenin exerts effects on proliferation or neuronal differentiation of neural stem cells. Dev. Biol. 268, 220-231. 120 Chenn, A. & Walsh, C.A. (2002). Regulation of cerebral cortical size by control of cell cycle exit in neural precursors. Science 297, 365-369. 121 Ivaniutsin, U., Chen, Y., Mason, J.O., Price, D.J. & Pratt, T. (2009). Adenomatous polyposis coli is required for early events in the normal growth and differentiation of the developing cerebral cortex. Neur. Dev. 4:3 doi: 10.1186/1749-8104-4-3. 122 Gulisano, M., Broccoli, V., Pardini, C., Boncinelli, E. (1996). Emx1 and Emx2 show different patterns of expression during proliferation and differentiation of the developing cerebral cortex in the mouse. Eur. J. Neurosci. 8, 1037-1050. 123 Kinzler, K.W. & Vogelstein, B. (1996). Lessons from hereditary colorectal cancer. Cell 87, 159-170. 124 Bienz, M & Clevers, H. (2000). Linking colorectal cancer to Wnt signaling. Cell 103, 311-320. 125 Harada, N., Tamai, Y., Ishikawa, T., Sauer, B., Takaku, K., Oshima, M & Taketo, M.M. (1999). Intestinal polyposis in mice with a dominant stable mutation of the β-catenin gene. EMBO J. 18, 5931-5942. 126 Nikuseva-Martic, T., Beros, V., Pecina-Slaus, N., Pecina, H.I. & Bulic-Jakus, F. (2007). Genetic changes of CDH1, APC, and CTNNB1 found in human brain tumours. Path. Res. Prac. 203, 779-787. 127 Alonso, L. & Fuchs, E. (2003). Stem cells in the skin: waste not, Wnt not. Genes & Dev. 17, 1189-1200.

Page 92: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

82

128 Chien, A.J., Moore, E.C., Lonsdorf, A.S., Kulikauskas, R.M., Rothberg, B.G., Berger, A.J., Major, M.B., Hwang, S.T., Rimm, D.L. & Moon, R.T. (2009). Activated Wnt/ß-catenin signaling in melanoma is associated with decreased proliferation in patient tumors and a murine melanoma model. Proc. Natl. Acad. Sci. 106, 1193-1198. 129 Malanchi, I., Peinade, H., Kassen, D., Hussenet, T., Metzger, D., Chambon, P., Huber, M., Hohl, D., Cano, A., Birchmeier, W. & Huelksen, J. (2008). Cutaneous cancer stem cell maintenance is dependent on β-catenin signaling. Nature 452, 650-654. 130 Majeti, R., Becker, M.W., Tian, Q., Lee, T.M., Yan, X., Lui, R., Chiang, J., Hood, L., Clarke, M.F. & Weissman, I.L. (2009). Dysregulated gene expression networks in human acute myelogenous leukemia stem cells. Proc. Natl. Acad. Sci. 106, 3396-3401. 131 Valencia, A., Roman-Gomez, J., Cervera, J., Such, E., Barragan, E., Bolufer, P., Moscardo, F., Sanz, G.F. & Sanz, M.A. (2009). Wnt signaling pathway is epigenetically regulated by methylation of Wnt antagonists in acute myeloid leukemia. Leukemia 23, 1658-1666. 132 Abrahamssona, A.E, Geron, I., Gotlib, J., Dao, K.T., Barroga, C.F., Newton, I.G., Giles, F.J., Durocher, J., Creusot, R.S., Karimi, M., Jones, C., Zehnder, J.L., Keating, A., Negrin, R.S., Weissman, I.L., and Jamieson, C.H.M. (2009). Glycogen synthase kinase 3β missplicing contributes to leukemia stem cell generation. Proc. Natl. Acad. Sci. 106, 3925-3929. 133 Zurawel, R.H., Chiappa, S.A., Allen, C. & Raffel, C. (1998). Sporadic medulloblastomas contain oncogenic β-catenin mutations. Cancer Res. 58. 896-899. 134 Nikuseva-Martic, T., Beros, V., Pecina-Slaus, N., Pecina, H.I. & Bulic-Jakus, F. (2007). Genetic changes of CDH1, APC, and CTNNB1 found in human brain tumors. Path. Res. & Prac. 203, 779-787. 135 Ellison, D.W., Onilude, O.E., Lindsey, J.C., Lusher, M.E., Weston, C.L., Taylor, R.E., Peasrson, A.D. & Clifford, S.C. (2005). β‐cateninstatuspredictsafavorableoutcomeinchildhoodmedulloblastomas:theUnitedKingdomchildren’scancerstudygroupbraintumourcommittee.J.Clin.Oncol.23,7951‐7957. 136 Rogers, H.A., Miller, S., Lowe, J., Brundler, M.A., Coyle, B., Grundy, R.G. (2009). An investigation of Wnt pathway activation and associated survival in central nervous system primitive neuroectoderm tumours (CNS PNET). Brit. J. Can. 1-11. 137 Roth, W., Wild-Bode, C., Platten, M., Grimmel, C., Melkonyan, H.S., Dichgans, J. & Weller, M. (2000). Secreted Frizzled-related proteins inhibit motility and promote growth of human malignant glioma cells. Oncogene 19, 4210-4220. 138 Pu, P., Zhang, Z., Kang, C., Jiang, R., Jia, Z., Wang, G. & Jiang, H. (2008). Downregulation of Wnt2 and β-catenin by siRNA suppresses malignant glioma cell growth. Can. Gene Therapy, 1-11. 139 Yu, J.M., Jun, E.S., Jung, J.S., Suh, S.Y., Han, J.Y., Kim, J.Y., Kim, K.W. & Jung, J.S. (2007). Role of Wnt5a in the proliferation of human glioblastoma cells. Cancer Let. 257, 172-181.

Page 93: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

83

140 McMahon, A.P. & Bradley, A. (1990). The Wnt-1 (int-1) proto-oncogene is required for development of a large region of the mouse brain. Cell 62, 1073-1085. 141 Dickinson, M.E., Krumlauf, R. & McMahon, A.P. (1994). Evidence for a mitogenic effect of Wnt-1 in the developing mammalian central nervous system. Development 12, 1453-1471. 142 Korur, S., Huber, R.M., Sivasankaran, B., Petrich, M., Morin Jr, P., Hemmings, B.A., Merio, A. & Lino, M.M. (2009). GSK3β regulates differentiation and growth arrest in glioblastoma. PLoS One 4, e7443. 143 Li, A., Walling, J., Kotliarov, Y., Center, A., Steed, M.E., Ahn, S.J., Rosenblum, M., Mikkelsen, T., Zenklusen, J.C. & Fine, H.A. (2008). Genomic changes and gene expression profiles reveal that established glioma cell lines are poorly representative of primary human gliomas. Mol. Cancer Res. 6, 21-29. 144 Pollard, S.M., Yoshikawa, K., Clarke, I.D., Danovi, D., Stricker, S., Russell, R., Bayani, J., Head, R., Lee, M., Bernstein, M., Squire, J.A., Smith, A., Dirks, P. (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. 145 Munemitsu, S., Albert, I., Rubinfeld, B. & Polakis, P. (1996). Deletion of the amino-terminal sequence stabilizes β-catenin in vivo and promotes hyperphosphorylaton of the adenomatous polyposis coli tumour suppressor protein. Mol. Cell. Biol. 16, 4088-4094. 146 Conti, L., Pollard, S.M., Gorba, T., Reitano, E., Toselli, M., Biella, G., Sun, Y., Sanzone, S., Ying, Q.L., Cattaneo, E. & Smith A. (2005). Niche-independent symmetrical self-renewal of a mammalian tissue stem cell. PLoS Biol. 3(9), e283. 147 Bhat, R.V., Budd Haeberlein, S.L. & Avila, J. (2004). Glycogen synthase kinase 3: a drug target for CNS therapies. J. Neurochem. 89, 1313-1317. 148 Jin, K & Galvan, V. (2007). Endogenous neural stem cells in the adult brain. J. Neuroimmune Pharmacol 2, 236-242. 149 Kim, W., Wang, X., Yaohong, W., Doble, B.W., Patel, S., Woodgett, J.R. & Snider, W.S. (2009). GSK-3 is a master regulator of neural progenitor homeostasis. Nat. Neurosci. 12, 1386-1395. 150 Woodhead, G.J., Mutch, C.A., Olsen, E.C. & Chenn, A. (2006). Cell-autonomous β-catenin signaling regulates cortical precursor proliferation. J. Neurosci. 26, 12620-12630. 151 Yaworsky, P.J. & Kappen, C. (1999). Heterogeneity of neural progenitor cells revealed by enhancers in the nestin gene. Dev. Biol. 205, 309-321. 152 Dahlstrand, J., Lardellu, M. & Lendahl, U. (1995). Nestin mRNA expression correlates with the central nervous system progenitor cell state in many, but not all, regions of developing central nervous system. Dev. Brain Res. 84, 109-129. 153 Bystron, I., Blakemore, C. & Rakic, P. (2008). Development of the human cerebral cortex: Boulder committee revisited. Nat. Rev. Neurosci. 9, 111-122.

Page 94: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

84

154 Machon, O., Backman, M., Machonova, O., Kozmik, Z., Vacik., Anderson, L. & Krauss, S. (2007). A dynamic gradient of Wnt signaling controls initiation of neurogenesis in the mammalian cortex and cellular specification in the hippocampus. Dev. Biol. 311, 223-237. 155 Ono, K., Takebayashi, H., Ikeda, K., Furusho, M., Nishizawa, T., Watanabe, K. & Ikenaka, K. (2008). Regional- and temporal-dependent changes in the differentiation of Olig2 progenitors in the forebrain, and the impact on astrocyte development in the dorsal pallium. Dev. Biol. 320, 456-468. 156 Yang, J., Zhang, W., Sun, X., Han, Y., Ding, M., Shi, Y. & Deng, H. (2009). P21-overexpression in the mouse beta cells leads to improved recovery from streptozotocin-induced diabetes. PLoS One 4, e8344. 157 Levison, S.W., de Vellis, J. & Goldman, J.E. (2005). Developmental Neurobiology, 4th Edition, edited by Mahendra S. Rao and Marcus Jacobson. Kluwer Academic / Plenum Publishers, New York, NY. Pp. 197-222. 158 Rakic, P. (2009). Evolution of the neocortex: a perspective from developmental biology. Nat. Rev. Neurosci. 10, 724-735. 159 Rakic, P. (2002). Neurogenesis in adult primate neocortex: an evaluation of the evidence. Nat. Rev. Neurosci. 3, 65-71. 160 Davidson, K.C., Jamshidi, P., Daly, r., Hearn, M.T.W., Pera, M.F. & Dottori, M. (2007). Wnt3a regulates survival, expansion, and maintenance of neural progenitors derived from human embryonic stem cells. Mol. Cell. Neurosci. 36, 408-415. 161 Maccario, H., Perera, N.M., Davidson, L., Downes, C.P. & Leslie, N.R. (2007). PTEN is destabilized by phosphorylation on Thr366. Biochem. J. 405, 439-444. 162 Kockeritz, L., Doble, B., Patel, S. & Woodgett, J.R. (2006). Glycogen synthase kinase-3—an overview of an over-achieving protein kinase. Curr Drug Targets 11, 1377-1388. 163 Yaworsky, P.J. & Kappen, C. (1999). Heterogeneity of neural progenitor cells revealed by enhancers in the nestin gene. Dev. Biol. 205, 309-321. 164 Nieuwenhuys, R., Voogd, J. & van Huijzen (2008). “Development.” The human central neurvous system, 4th Edition. Springer, New York, NY, pp. 7-66. 165 Morshead, C.M., Reynolds, B.A., Craig, C.G., McBurney, M.W., Staines, W.A., Morassutti, D., Weiss, S. & van der Kooy, D. (1994). Neural stem cells in the adult mammalian forebrain: a relatively quiescent subpopulation of subependymal cells. Neuron 13, 1071-1082. 166 Castelo-Branco, G., Rawal, N. & Arenas, E. (2004). GSK3β inhibition/β-catenin stabilization in ventral midbrain precursors increases differentiation into dopamine neurons. J. Cell Sci. 117, 5731-5737. 167 Jope, R.S. & Roh, M. (2006). Glycogen synthase kinase-3 (GSK3) in psychiatric diseases and therapeutic interventions. Curr Drug Targets 7(11), 1421-1434.

Page 95: Wnt Signaling in Human Neural Stem Cells and Brain Tumour ... › bitstream › 1807 › ... · neural stem cells in EGF and FGF.....54 3.2 GSK3 inhibition induces neuronal differentiation

85

168 Phiel, C.J., Wilson, C.A., Lee, V.M. & Klein, P.S. (2003). GSK3α regulates production of Alzheimer’s disease amyloid-β peptides. Nature 423, 435-439. 169 Ryder, J. et al. (2003). Divergent roles of GSK3 and CDK5 in APP processing. Biochem. Biophys. Commun. 312, 922-929. 170 Verhaak, R.G.W. et al. (2009). Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 17, 98-110. 171 Phillips, H.S., et al. (2006). Molecular subclasses of high-grade glioma redict prognosis, delineate a pattern of disease progression, and resemble stages on neurogenesis. Cancer Cell 9, 157-173. 172 Sun, Y., Pollard, S., Conti, L., Toselli, M., Biella, G., Parkin, G., Willatt, L., Falk, A., Cattaneo, E. & Smith, A. (2008). Long-term tripotent differentiation capacity of human neural stem (NS) cells in adherent culture. Mol. Cell Neurosci. 38, 245-258. 173 Rakic, P. (2002). Adult neurogenesis in mammals: an identity crisis. J. Neurosci. 22, 614-618. 174 Kikuchi, A. (2003). Tumour formation by genetic mutations in the components of the Wnt signaling pathway. Cancer Sci. 94, 225-229. 175 Ellison, D.W., onilude, O.E., Lindsey, J.C., Lusher, M.E., Weston, C.L., Taylor, R.E., Pearson, A.D. & Clifford, S.C. (2005). Β-catenin status predicts a favourable outcome in childhood medulloblastomas: the united Kingdom children’s cancer study group brain tumour committee. J. Clin. Oncol. 23, 79517957. 176 Korur, S., Huber, R.M., Sivasankaran, B., Petrich, M., Morin, P., Hemmings, B.A., Merlo, A. & Lino, M.M. (2009). GSK3β regulates differentiation and growth arrest in glioblastoma. PLoS One 4, e7443. 177 Abdouh, M., Facchino, S., Chatoo, W., Balasignam, V., Ferreira, J. & Bernier, G. (2009). Bmi1 sustains human glioblastoma multiforme stem cell renewal. J. Neurosci. 29, 8884-8896. 178 Kotliarova, S., Pastorino, S., Kovell, L.C., Kotliarov, Y., Song, H., Zhang, W., Bailey, R., Maric, D., Zenklusen, J.C., Lee, J. & find, H.A. (2008). Glycogen synthase kinase-3 inhibtion induces glioma cell death through c-Myc, Factor-κB, and glucose regulation. Cancer Res. 68, 6643-6651. 179 Vescovi, A.L., Galli, R. & Reynolds, BA. (2006). Brain tumour stem cells. Nat. Rev. Cancer 6, 425-436. 180 Diamandis, P., Sacher, A.G., Tyers, M. & Dirks, P.B. (2009). New drugs for brain tumours? Insights from chemical probing of neural stem cells. Med Hypotheses 72, 683-687.