09-10-26 thesis

58
Interactions between the Wnt Pathway and Inflammatory mediators in mesenchymal stem cells Nadene FangTing Tam 243454 Thesis submitted to the University of Melbourne for the Degree of BSc with Honours The University of Melbourne Department of Medicine Royal Melbourne Hospital 26 th October 2009

Upload: nadene-tam

Post on 27-Jul-2015

83 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: 09-10-26 Thesis

Interactions between the Wnt Pathway and Inflammatory

mediators in mesenchymal stem cells

Nadene FangTing Tam

243454

Thesis submitted to the

University of Melbourne for the

Degree of BSc with Honours

The University of Melbourne

Department of Medicine

Royal Melbourne Hospital

26th October 2009

Page 2: 09-10-26 Thesis

2

Acknowledgements

I would like to thank my supervisor Dr Derek Lacey for his tremendous support he provided

throughout the year and especially during the weeks prior to handing in my thesis. Dr Lacey

passed on many different laboratory techniques and his guidance was fundamental to this project.

Many thanks to Prof Gary Anderson, Prof John Hamilton and Dr Derek Lacey for providing me

with such a great opportunity by allowing me to undertake this honours project in such a

fantastic department.

I would also like to thank the Hamilton group especially Agnieszka Swierczak, Jarrad Pobjoy,

Dominic Stipanov and Thao Nguyen for being such great company and providing much needed

laughs throughout the year. Special thanks to Thao and Dr Glen Scholz for passing on their

expertise in laboratory techniques and advice on data presentation.

To my fellow honours students, Shalini Maran and Zohra Miazoi, thank you for being such

amazing friends, this year would not have been the same without your friendship and support.

Page 3: 09-10-26 Thesis

3

Declaration

DECLARATION BY SCHOLAR:

I, Nadene FangTing Tam

certify that

- the thesis comprises only my original work, except where indicated in the

accompanying Acknowledgement statement

- the thesis conforms to the specifications outlined in the Honours Handbook

Signature: (Nadene Tam)

Date:

DECLARATION BY SUPERVISOR:

I confirm that the declaration above of Nadene FangTing Tam thesis are a true and fair representation of

the student’s work.

Signature: (Dr Derek Lacey)

Date:

Page 4: 09-10-26 Thesis

4

Table of Contents

TITLE PAGE ............................................................................................................................................................. 1

ACKNOWLEDGEMENTS .......................................................................................................................................... 2

DECLARATION ........................................................................................................................................................ 3

TABLE OF CONTENTS .............................................................................................................................................. 4

ABSTRACT .............................................................................................................................................................. 6

ABBREVIATIONS .................................................................................................................................................... 8

CHAPTER 1: INTRODUCTION ................................................................................................................................ 10

1.1 STEM CELLS ........................................................................................................................................................... 10

1.1.1 Embryonic Stem Cells ................................................................................................................................ 10

1.1.2 Mesenchymal Stem cells (MSCs) ............................................................................................................... 11

1.1.4 Identification of MSCs ............................................................................................................................... 12

1.2 MSC TO OSTEOBLAST ............................................................................................................................................. 13

1.3 OSTEOGENIC GENES ................................................................................................................................................ 14

1.4 EXTRACELLULAR FACTORS ........................................................................................................................................ 15

1.5 MARKERS OF DIFFERENTIATION ................................................................................................................................. 15

1.6 WNT PATHWAY ..................................................................................................................................................... 16

1.6.1 Wnt proteins ............................................................................................................................................... 16

1.6.2 Wnt signalling pathways ............................................................................................................................ 17

1.6.3 Inhibitors of the Wnt pathway .................................................................................................................... 19

1.6.4 Role of the Wnt pathway in MSCs .............................................................................................................. 20

1.7 HYPOTHESIS AND AIMS ............................................................................................................................................ 22

CHAPTER 2: MATERIALS AND METHODS .............................................................................................................. 23

2.1 MATERIALS ........................................................................................................................................................... 23

2.2 CELL CULTURE ........................................................................................................................................................ 23

2.3 TRANSFECTION OF PLASMIDS .................................................................................................................................... 24

2.4 DUAL-LUCIFERASE REPORTER ASSAY .......................................................................................................................... 24

2.5 NUCLEAR EXTRACTION ............................................................................................................................................. 25

2.6 OSTEOBLAST DIFFERENTIATION .................................................................................................................................. 25

2.7 RNA EXTRACTION, REVERSE TRANSCRIPTION AND REAL-TIME POLYMERASE CHAIN REACTION ................................................. 26

2.8 WESTERNBLOT ANALYSIS .......................................................................................................................................... 27

CHAPTER 3: RESULTS ........................................................................................................................................... 28

3.1 DETERMINING THE OPTIMAL CONCENTRATION OF TOPFLASH AND FOPFLASH PLASMIDS ..................................................... 28

3.2 EFFECTS OF ∆45, IL-1Β AND TNF-Α ON NUCLEAR Β-CATENIN ACTIVITY ............................................................................. 28

3.3 LICL INCREASES Β-CATENIN ACTIVITY ........................................................................................................................... 29

3.4 THE EFFECTS OF WNT3A, IL-1Β AND TNF-Α ON Β-CATENIN ACTIVITY ................................................................................ 29

3.5 ∆45 DID NOT INCREASE Β-CATENIN LEVELS IN WHOLE CELL LYSATES .................................................................................. 30

3.6 DETERMINING THE OPTIMAL STIMULATION PERIOD PRIOR TO NUCLEAR EXTRACTION ............................................................ 31

3.7 NUCLEAR Β-CATENIN LEVELS IN CELLS TREATED WITH LICL WERE NOT INFLUENCED BY IL-1Β OR TNF-Α .................................... 31

Page 5: 09-10-26 Thesis

5

3.8 NUCLEAR Β-CATENIN LEVELS – TREATMENTS WITH IL-1Β OR TNF-Α AND WNT3A ............................................................... 31

3.9 DETERMINING THE INCUBATION TIME PERIOD FOR OPTIMAL ALP STAINING ........................................................................ 32

3.10 IL-1Β AND TNF-Α DECREASE ALP STAINING IN ∆45-TRANSFECTED CELLS ........................................................................ 32

3.12 WNT3A INHIBITS OSTEOBLAST DIFFERENTIATION ......................................................................................................... 33

3.13 CHANGE IN OSTEOBLASTIC GENE EXPRESSION WITH LICL OR WNT3A AND IL-1Β OR TNF-Α ................................................. 33

CHAPTER 4: DISCUSSION ...................................................................................................................................... 35

REFERENCES ......................................................................................................................................................... 42

FIGURES ............................................................................................................................................................... 46

Page 6: 09-10-26 Thesis

6

Abstract

Rheumatoid arthritis a chronic inflammatory disease, consisting of activated leukocytes,

inflamed synovium and the production inflammatory mediators such as IL-1β and TNF-α. These

cytokines have been implicated in the pathology seen in this autoimmune disease such as

cartilage destruction, the loss of bone mass, stiffness and swelling of the joints. Wnt molecules

are morphogens which are expressed at multiple sites, including sites of inflammation. There are

19 known mammalian Wnt family members which signal via binding to frizzled and LRP5/6

receptors. The canonical Wnt signalling pathway has been shown to promote osteogenesis and

has a role in the differentiation of MSCs into osteoblasts.

IL-1β and TNF-α have been shown to block MSC differentiation to osteoblasts. This project

aims to investigate the mechanism by which MSCs are blocked from differentiating in

osteoblasts. It is hypothesised that IL-1β and TNF-α are blocking Wnt/β-catenin signalling and

therefore blocking Wnt driven osteogenesis.

In this project, the effects of IL-1β and TNF-α have been investigated at multiple levels. Firstly,

dual luciferase reporter assays were conducted to determine if β-catenin activity is affected by

the cytokines. Inducing the overexpression of β-catenin with the plasmid ∆45 was used to

determine which part of the Wnt pathway IL-1β and TNF-α regulate. Initial results indicate that

IL-1β and TNF-α are able to inhibit Wnt/β-catenin signalling by reducing the activity of β-

catenin as measured by a reporter assay. However, later experiments using cells that had

undergone prolonged passaging contradict these results. Following reporter assays with cells

transfected with an axin-RFP plasmid failed to show a reduction firefly luciferase activity despite

Page 7: 09-10-26 Thesis

7

other studies showing that the plasmid induces the formation of the β-catenin destruction

complex.

To specifically identify β-catenin levels in cells that have been treated with IL-1β and TNF-α in

the presence or absence of LiCl or Wnt3a, western blots were conducted using whole cell lysates,

cytoplasmic and nuclear extracts. Results did not indicate any detectable change in the levels of

β-catenin regardless of the treatment.

Osteoblast differentiation assays measuring alkaline phosphatase (ALP) expression, which is a

osteoblast marker, seemed to show that IL-1β and TNF-α have been able to block the Wnt

pathway with reduced ALP staining.

In conjunction to the results investigating the effects of IL-1β and TNF-α on the osteoblast

differentiation, there seemed to be a correlation with the cell passage number to the effects seen

by the cytokines on osteoblast differentiation and Wnt signalling in MC3T3-E1 cells. Repetitions

of experiments using cells that had been passaged a number of times over a prolonged period

gave conflicting results to initial experiments using cells that had been freshly cultured after

removal from liquid nitrogen. These results suggest that the effects seen by IL-1β and TNF-α on

osteoblast differentiation are affected by the number of times MC3T3-E1 cells have been

passaged.

In conclusion, the effects of IL-1β and TNF-α seen in this project on the Wnt pathway and

osteoblast differentiation are conflicting. Though further investigation into how cell passage

number affects their Wnt/β-catenin signalling within cells could provide more a better

understanding of the relationship between the Wnt pathway and inflammation.

Page 8: 09-10-26 Thesis

8

Abbreviations

ALP Alkaline phosphatase

APC Adenmatous polyposis coli

BMPs Bone morphogenic protein

BSP Bone sialoprotein

CBP CREB binding protein

Cbfa1 Core binding factor a1

CFU-F Colony forming unit fibroblasts

CIA Collagen-induced arthritis

CKIα Casein kinase Iα

Colla1 Type I Collagen

CRD Cysteine-rich domain

CREB cAMP response element binding

DKK Dickkopfs

Dsh Dishevelled

ECM Extracellular matrix

ES Embryonic stem

FGFs Fibroblast growth factors

FZD Frizzled

GSK 3β Glycogen synthase kinase 3β

HDAC Histone deacetylases

hMSCs Human mesenchymal stem cells

IL-1β Interleukin-1β

Int Integration

Page 9: 09-10-26 Thesis

9

Lef lymphoid enhancer factor

LiCl Lithium chloride

LIF Leukaemia inhibitory factor

LDL Low-density lipoprotein

LRP5/6 Low-density lipoprotein receptor-related proteins 5/6

MAP Mitogen-activated protein

MSCs Mesenchymal stem cells

mMSCs Murine mesenchymal stem cells

NLK Nemo-like kinase

Osx Osterix

PCR Polymerase chain reaction

RA Rheumatoid arthritis

RT-PCR Real-time polymerase chain reaction

Runx2 Runt related transcriptional factor 2

sFRPs Secreted frizzled-related proteins

Tcf T-cell factor

TAZ Transcriptional co-activator with PDZ-binding motif

TNF-α Tumour necrosis factor-α

TrCP Transducin repeat-containing protein

TBP TATA binding protein

Wg Wingless

WISP Wnt-1 induced secreted proteins

Wnt Wingless/Int

Page 10: 09-10-26 Thesis

10

Chapter 1: Introduction

1.1 Stem Cells

Due to their unique characteristics, stem cells have been hailed as a potential cure to

multiple diseases and offer hope to thousands of patients. Early experiments initially described

stem cells as cells that are able to rapidly proliferate and form colonies of cells, which were later

determined to be clones generated from single cells (Till and Mc, 1961). Two important

characteristics distinguish stem cells from other cell types. Firstly, they are non-specialised cells

that have the ability to renew themselves through cell division. Secondly, they have the ability to

differentiate into any type of cell in the body (pluripotency) and thereby replace and repair

damaged tissue. Most crucially they are also able to undergo indefinite self-renewal, without

losing their differentiation potential. There are a number of different types of stem cells such as

embryonic stem cells, haematopoietic stem cells, mesenchymal stem cells, and tissue specific

stem cells, and they all have different degrees of pluripotency. Tissue-specific stem cells, for

example, have a limited range of cells they can differentiate into and are known to be multipotent.

Stem cells adjust their responses according to the signals received from the surrounding

microenvironment. This microenvironment that promotes stem cells longevity and enables their

differentiation is known as the stem cell niche (Watt and Hogan, 2000). Under appropriate

extracellular signals, tissue-specific stem cells differentiate into committed progenitors which are

committed to differentiating into a specific cell type (Weissman, 2000).

1.1.1 Embryonic Stem Cells

Embryonic stem (ES) cells develop after fertilization of the oocyte (and before the

development of the foetus), upon which the blastocyst forms first, then there is the formation of

the three germ layers – endoderm, mesoderm and ectoderm. ES cells can be isolated from the

Page 11: 09-10-26 Thesis

11

inner cell mass of the blastocyst (Evans and Kaufman, 1981). However, there is much debate

about the ethical considerations with regards to the use of embryonic stem cells for research and

possible therapeutic use. Furthermore, the research conducted using animal models have shown

that there are many difficulties in maintaining ES cells in their undifferentiated state (Martin et

al., 2005).

The three germ layers mature and develop into cells specific to each post-natal tissue

present in the adult. These post-natal tissues also contain tissue-specific stem cells and are also

termed adult stem cells. Adult stem cells can be broadly classified as haematopoietic, epithelial,

mesenchymal and neural stem cells.

1.1.2 Mesenchymal Stem cells (MSCs)

Mesenchymal stem cells (MSCs) are fibroblast-looking cells that were first observed and isolated

from the bone marrow (Friedenstein et al., 1966). Recent studies however, have shown that

MSCs can be found in most organs and tissues throughout the body (da Silva Meirelles et al.,

2006). They are typically characterised as cells that are able to undergo continual self-renewal,

extensive proliferation and can differentiate into multiple cell types – including osteoblasts,

chondrocytes and adipocytes (Caplan, 1991; Dominici et al., 2006; Pittenger et al., 1999). MSCs

can be distinguished from haematopoietic cells by their adherence to culture dishes in vitro

(Dominici et al., 2006). Friendenstein et al. first coined the term colony forming unit fibroblasts

(CFU-F) to describe the phenomenon of a single cell forming a colony of morphologically

similar cells isolated from bone marrow (Friedenstein et al., 1970).

As mentioned above, stem cells are located within a niche that is conducive to their self

renewal and differentiation. Exposure and stimulation by extracellular signalling factors that are

Page 12: 09-10-26 Thesis

12

secreted into the microenvironment promotes differentiation of MSCs into various non-

haematopoietic cell types (Caplan, 1991). A network of signalling interactions between stem

cells and their daughter cells or surrounding cells are also part of this niche which eventually

regulates their differentiating and proliferation potentials. There are several cytokines and growth

factors such as leukaemia inhibitory factor (LIF) and fibroblast growth factors (FGFs) that have

been shown to be involved in the self-renewal of MSCs, maintaining a pool of undifferentiated

MSCs, but also have been shown to be important in MSC differentiation (Ito et al., 2008; Ng et

al., 2008; Pruijt et al., 1997; Szilvassy et al., 1996; Whitney et al., 2009). Another family of

proteins that have been implicated in the differentiation of MSCs into numerous cell types

including osteoblasts is the wingless/int (Wnt) family of proteins and will be elaborated on in the

following sections.

1.1.4 Identification of MSCs

Much of the early work on MSCs has concentrated on the identification and isolation of stem

cells using a combination of cell surface markers. However, as there is no single cell surface

antigen that exists to positively identify a MSC, a combination of markers is the only means of

phenotypic identification of MSCs. Human MSCs (hMSCs) can be isolated based on the

expression of Stro-1, CD105, CD73 and CD90 while lacking the expression of CD34+, CD-14 or

CD79α or CD19 (Dominici et al., 2006). MSCs isolated from mice are based on the expression

of Sca-1 and CD44 while failing to express CD45, CD 31 or CD11b. Both mouse and human

MSC consistently do not express CD45 and CD11b on their cell surface (Dominici et al., 2006;

Tropel et al., 2004). In comparison, hMSCs are better characterised compared to murine MSCs

(mMSCs) (Tropel et al., 2004).

Page 13: 09-10-26 Thesis

13

Essentially, MSCs are phenotypically identified due to the lack of expression of

haematopoietic markers. Isolating MSCs from different species such as rabbits and rats has

shown that the expression of these markers are different but there has been widespread

agreement that MSCs, regardless of their source, lack the expression of CD45. Due to the

variable expression of cell surface markers depending on the tissue sources, this has made for the

isolation of MSCs for research challenging.

MSC isolation has remained a challenge for researchers. The utilisation of stem cell lines

or precursor lines has helped overcome some of these issues, but this however has created their

own issues.

1.2 MSC to osteoblast

It was first noticed by Friedenstein et al. that by transplanting bone marrow cells to a

bone graft, these cells were able to differentiate into osteoblasts and eventually the formation of

new bone would occur (Friedenstein et al., 1966). It would later be realised that these bone

marrow cells conferred a population of MSCs. The ability of MSCs to differentiate into

osteoblasts is important for the maintenance of bone health, with damaged bone being replaced

by the formation of new bone, for example, when there is a bone fracture. Experiments using

hMSCs have shown that there is an increase in alkaline phosphatase activity when hMSCs are

cultured in osteogenic media which consists of ascorbate, β-glycerol and dexamethasone (Majors

et al., 1997; Pittenger et al., 1999; Tropel et al., 2004) and similarly with mMSCs (Tropel et al.,

2004).

Page 14: 09-10-26 Thesis

14

1.3 Osteogenic genes

There are several osteogenic genes that have been shown to be associated in the process

of MSC differentiation. The transcription of these genes has been shown to be upregulated in

osteoblasts and they include Runx2 (runt related transcriptional factor 2)/Cbfa-1(core binding

factor a1) and Osterix (Osx). These transcription factors are of great interest as they could be

crucial for furthering our understanding of chronic bone diseases such as rheumatoid arthritis and

osteoarthritis.

For MSCs to differentiate into pre-osteoblasts, the gene Runx2/Cbfa-1 is required and

experiments conducted by Komori et al. using mice have shown that the deletion of the

Runx2/Cbfa1 gene results in the inhibition of osteoblast differentiation. The Runx2/Cbfa1 gene

encodes for the Runx2/Cbfa1 transcription factor which is expressed in osteoblasts. In mutant

mice that did not express the Runx2/Cbfa1 gene, there was a lack of ossification with the mice

developing short legs and some showing signs of dwarfism. Furthermore, using x-ray

examinations, these mutant mice, compared to wild type mice of the same embryonic age,

showed weak calcification of various skeletal components such as the skull, ribs and mandibula

(Komori et al., 1997). Using Alazarin Red staining, this result has been replicated in the same

study as well as by other groups (Komori et al., 1997; Otto et al., 1997). The newborn Runx2-

null mice died shortly after birth due to the developmental failure of the ribs resulting in their

suffocation. These studies highlight the importance of the Runx2/Cbfa1 gene in bone formation.

Another gene that was found to be important for osteoblast development is Osterix (Osx)

which is specifically expressed in osteoblasts and is necessary to promote osteoblast

differentiation from preosteoblast prescursors (Ducy and Karsenty, 1995; Ducy et al., 1997;

Towler et al., 1994). As a consequence of the inactivation of Osx, there was little mineralisation

Page 15: 09-10-26 Thesis

15

of the facial and skull bones and deformation of other parts of the skeletal structure including the

ribs and limb bones (Nakashima et al., 2002). The absence of mineralisation and lack of

ossification in these animal models have shown that Runx2/Cbfa1 and Osx genes are essential

for the maturation of osteoblasts. In the same study by Nakashima et al., they were able to

determine that Osx and Runx2/Cbfa1 genes operate in the same osteoblast differentiation

pathway, with Osx gene downstream of Runx2/Cbfa1 (Nakashima et al., 2002). Furthermore,

through staining of the skeletons of Runx2/Cbfa1 null mice, they were also able to determine that

the expression of Osx was dependent on the presence of Runx2/Cbfa1 (Nakashima et al., 2002).

1.4 Extracellular Factors

It has also been shown that a group of growth factors known as bone morphogenetic

proteins (BMPs) promote osteogenesis of MSCs (Zhang et al., 2009). Binding to cell surface

receptors, BMPs are able to induce β-catenin signalling and promote bone formation (Chen et al.,

2007). This is an important and area of research, however it is outside the scope of this thesis.

1.5 Markers of Differentiation

There are several osteoblast markers that can be used in determining the progression of

the differentiation of MSCs into osteoblasts and these markers are commonly used in research

regarding osteoblast differentiation.

Proteins such as alkaline phosphatase (ALP), bone sialoprotein (BSP), osteonectin, and

type I collagen (Col1a1) can be considered early markers of osteoblast differentiation (van

Straalen et al., 1991).

During the proliferation of osteoblasts, there is an increased secretion of extracellular

matrix (ECM) and Col1a1 is one of the ECM proteins (Owen et al., 1990). This increased

Page 16: 09-10-26 Thesis

16

expression of Colla1 makes it a suitable marker for early osteoblast differentiation. Eventually

the proliferative stage in osteoblasts wanes and an increase in the expression of ALP proceeds,

with increased mineralization occurring in MSC cultures. Staining for ALP expression will be

used to determine the effects of Wnt ligands which will be discussed later in this thesis, on the

differentiating potential of pre-osteoblast cells into osteoblasts.

Other proteins such as osteocalcin, osteopontin and bone sialoprotein (BSP) can be found

in mature osteoblasts with osteocalcin being the most cell specific late marker for osteoblast

differentiation. It is expressed in osteoblasts, not in any other ECM producing cells, and can be

measured by real-time polymerase chain reaction (RT-PCR) (Ducy and Karsenty, 1995).

Several of these osteoblastic genes will be used in RT-PCR experiments to determine how

activation of the Wnt pathway affects MSCs differentiating into osteoblasts.

1.6 Wnt Pathway

1.6.1 Wnt proteins

Initial experiments in drosophila identified a gene that was required for wing formation

and was termed the wingless (Wg) gene as flies without this gene did not develop wings

(Nusslein-Volhard and Wieschaus, 1980). Around the same time, cancer researchers identified a

gene at the integration region of a tumour and normal tissue which was termed Int (integration)

(Wainwright et al., 1988). Both genes were found to be homologous and led to the coining of the

Wnt proteins through the combination of both gene names. These Wnt proteins are a family

cysteine-rich secreted glycoproteins, normally ranging from 350 – 400 amino acids in length,

and to date there are 19 known mammalian Wnt proteins.

Page 17: 09-10-26 Thesis

17

At the cellular level, the Wnt pathway is thought to have many roles including the

regulation of cell motility and polarity. In addition, the signalling pathway is also known to be

crucial in the causation of diseases such as cancer. This project focuses on the importance of the

Wnt signalling pathway in chronic bone related disease such as rheumatoid arthritis.

Our skeletal structure undergoes continual remodelling and Wnt proteins are thought to

play an important role in the regulation of bone mass. Through the Wnt signalling pathway, Wnt

proteins are thought to regulate bone formation, by controlling differentiation, proliferation and

cell death.

1.6.2 Wnt signalling pathways

1.6.2.1 Canonical and Non-canonical

There are at least 2 intra-cellular signalling pathways involved in Wnt signalling – the

canonical and non-canonical pathways (Figure 1). Most of the 19 Wnt proteins are known to

activate either the canonical Wnt pathway or the non-canonical Wnt pathway. The non-canonical

pathway does not utilise β-catenin, which is the central molecule in the canonical pathway, and is

not as well-studied. This project focuses the canonical pathway and β-catenin signalling in

osteoblast precursors and therefore the following is a description of the canonical pathway.

It is understood that in the steady state the Wnt/β-catenin pathway is not activated, and a

destructive complex forms in the cytoplasm. This complex results in stabilising of cytoplasmic

β-catenin in a complex, with adenmatous polyposis coli (APC) and Axin, allowing glycogen

synthase kinase 3β (GSK3β) and casein kinase Iα (CKIα) to phosphorylate β-catenin (Nakamura

et al., 1998). Phosphorylation of β-catenin results in its degradation via the β-TrCP (transducin

repeat-containing protein) mediated proteasome pathway (Hart et al., 1999; Liu et al., 1999).

Page 18: 09-10-26 Thesis

18

Biochemical experiments have shown that Wnt proteins interact with cell surface

receptors Frizzled (FZD) and low-density lipoprotein (LDL) receptor-related proteins 5 and 6

(LRP5/6) to initiate the canonical signalling pathway. FZD receptors are 7 transmembrane

molecules with a N-terminal cysteine-rich domain (CRD) while its co-receptor LRP5/6 is a

single-pass transmembrane protein. Wnt proteins bind directly to the CRD of the FZD receptor.

Upon the binding of Wnt proteins to FZD and LRP5/6 receptors, CKIα hyperphosphorylates

Dishevelled (Dsh) causing it to have an increased affinity for the C-terminal domain of the FZD

receptor and Frat-1 (Cong et al., 2004; Klein et al., 2006). Simultaneously, both GSK3β and

CKIα cause the phosphorylation of the LRP5/6 receptor, in turn resulting in the recruitment of

Axin to the cytoplasmic domain of the LRP5/6 receptor (Hino et al., 2003). Furthermore, at the

cell surface, Dsh, Frat-1 and Axin are thought form a complex which results in the disassembly

of the β-catenin destructive complex (Fagotto et al., 1999; Li et al., 1999). Therefore, an

accumulation of β-catenin within the cytoplasm occurs and is able to translocate to the nucleus

via directly interacting with the nuclear pore components (Yokoya et al., 1999).

Accumulation and translocation of β-catenin into the nucleus results in the physical

displacement of Groucho, transiently inducing the conversion of T-cell factor (Tcf)/Lymphoid

enhancer factor (Lef) into a transcriptional activator (Daniels and Weis, 2005). Thus, the

transcriptional complex of β-catenin and Tcf/Lef bind to the promoter and induce the

transcription of Wnt target genes. In the absence of Wnt signalling, Tcf/Lef normally acts as a

transcriptional repressor preventing the transcription of Wnt target genes due its association with

transcriptional inhibitor Groucho which together with histone deacetylases (HDAC) which

prevent the unwinding of DNA (Cavallo et al., 1998; Waltzer and Bienz, 1998). The shuttling of

β-catenin to and from the nucleus has been shown to be regulated by Axin and Tcf/Lef, either

Page 19: 09-10-26 Thesis

19

inducing cytoplasmic or nuclear retention by the 2 respective proteins (Cong and Varmus, 2004;

Tolwinski and Wieschaus, 2001).

There are two other important nuclear proteins, Bcl9/Legless and Pygopus which are

thought to be implicated in the nuclear retention of β-catenin. Pygopus associates with

Bcl9/Legless which in turn binds to the N-terminus of β-catenin (Kramps et al., 2002; Parker et

al., 2002). Simultaneously, there is recruitment of transcriptional co-activators such as histone

acetylase CBP/p300 and Brg-1, part of the SWI/SNF chromatin remodelling complex, to the C-

terminus of β-catenin (Barker et al., 2001; Hecht et al., 2000), facilitating the transcription of

osteoblastic genes.

1.6.3 Inhibitors of the Wnt pathway

There are a number of inhibitors of the canonical Wnt signalling pathway including

secreted frizzled-related proteins (sFRP) 1 – 4 which resembles the CRD of the FZD receptor.

Binding of sFRP to Wnt proteins physically prevents Wnt proteins from binding to FZD

receptors. Using β-catenin specific luciferase reporter gene assay, it has been shown in human

osteoblasts that there is complete suppression of Wnt signalling when Tcf and Wnt plasmids are

co-transfected with sFRP.

Wnt proteins are also able to bind to an unusual Wnt receptor known as the

Derailed/RYK class of tyrosine kinase transmembrane receptors. The inhibitor Wnt inhibitory

factor (WIF) has similarities with the extracellular domain of the Derailed/RYK receptors which

is involved in binding Wnt proteins – both of which are thought to be members in the non-

canonical pathway.

Page 20: 09-10-26 Thesis

20

The Wnt signal transduction pathway has also been shown to be inhibited by Dickkopfs

(Dkk) 1 – 5, secreted glycoproteins, binding to the LRP5/6 receptor (Bafico et al., 2001; Mao et

al., 2001; Semenov et al., 2001). Dkk-1 is able to bind to another transmembrane protein known

as Kremens and this association with Kremens and LRP5/6 receptors, possiblely induces the

internalisation of LRP5/6 receptors (Mao et al., 2002). Since the Wnt canonical signalling

cascade can only be initiated upon the binding of Wnt ligands to LRP5/6, Wnt signalling cannot

occur due to Dkk-1 and LRP5/6 binding.

Another inhibitor of the Wnt pathway is the mitogen-activated protein (MAP) kinase-

related protein Nemo-like kinase (NLK). Unlike the other inhibitors mentioned above, NLK

inhibits the Wnt pathway by phosphorylating Tcf/LEF thus causing a decrease in DNA-binding

affinity of the β-catenin-Tcf/LEF complex (Ishitani et al., 2003).

1.6.4 Role of the Wnt pathway in MSCs

1.6.4.1 Rheumatoid Arthritis

Rheumatoid arthritis (RA) is a chronic inflammatory, auto-immune disease of the joints

resulting in the activation of leukocytes and the excessive production of inflammatory cytokines.

Synovial membrane lining the joints plays an essential role in regulating inflammation and

changes to this thin tissue results in the pathology seen in patients with RA. The synovial

membrane is normally only a few cells thick, containing synoviocytes which produce

extracellular matrix and synovial fluid. In patients with RA, there is an increase in the number

and types of cells that are found in the synovial lining – including activated B and T cells,

macrophages, mast cells and plasma cells and extensive angiogenesis is also observed. These

activated leukocytes produce cytokines that act in an auto- and paracrine fashion to induce the

production of inflammatory mediators and tissue degrading enzymes. This chronic induction of

Page 21: 09-10-26 Thesis

21

cytokines by these pro-inflammatory cells results in the perpetual inflammation seen in

rheumatoid arthritis. This leads to the destruction of bone and cartilage, as well as swelling and

redness of the joints due to the increased numbers of synoviocytes at the joint.

One of the most commonly studied cytokines implicated in the pathogensis of RA is

tumour necrosis factor (TNF)-α. TNF-α was first discovered to have similar necrosing effects on

tumour cells as endotoxin. Later studies showed that TNF-α causes the inhibition of bone

formation and stimulates bone resorption (Bertolini et al., 1986). Since the identification of the

presence of TNF-α in the synovial membrane and synovial fluid of patients with RA, TNF-α has

been implicated in the pathogenesis seen in RA (Chu et al., 1992; Hopkins and Meager, 1988).

Currently, anti-TNF-α therapy is the most successful biological therapeutic used in the treatment

of RA.

Another pro-inflammatory cytokine involved in the pathogenesis of RA is interleukin

(IL)–1 with early experiments detecting its activity in the synovial fluid of patients with RA

(Fontana et al., 1982) and its ability to induce bone resorption (Gowen et al., 1983). Initial

studies using mice with collagen-induced arthritis (CIA), which is a widely used animal model

for arthritis, have shown that early treatment with anti-IL-1 antibodies can delay the onset of RA

(van den Berg et al., 1994) and others indicating that bone damage is abolished with these

antibodies (Joosten et al., 1999).

The mechanism of action of IL-1β and TNF-α on stem cells is unknown but there seems

to be an inter-relationship between TNF-α and IL-1β. Previous studies have shown that anti-

TNF-α antibodies reduce the level of IL-1β production in cell cultures from patients with RA

(Brennan et al., 1989), while others show that TNF-α is able to induce the production of IL-1β

Page 22: 09-10-26 Thesis

22

(Dinarello et al., 1986). Both IL-1βand TNF-α have also been shown to have additional

inhibitory effects on MSC differentiation into osteoblasts (Lacey et al., 2009).

The implications of these two pro-inflammatory cytokines in the pathogenesis of RA,

leads to the hypothesis that IL-1β and TNF-α blocks the Wnt pathway preventing MSCs from

differentiating into osteoblasts, thus causing the reduction in bone mass seen in patients with RA.

1.7 Hypothesis and Aims

The hypothesis to be tested is that IL-1β and TNF-α inhibit the differentiation of

mesenchymal stem cells into osteoblasts by antagonising the Wnt signalling pathway. To

determine at which level these cytokines interact with the Wnt pathway, the specific aims were

1. To determine, at the cytoplasmic level, how accumulation of β-catenin is affected by

IL-1β and TNF-α

2. To determine how nuclear levels of β-catenin are affected by IL-1β and TNF-α after

stimulation of the Wnt pathway

3. To test the functional importance of IL-1β and TNF-α in osteoblast differentiation

Page 23: 09-10-26 Thesis

23

Chapter 2: Materials and Methods

2.1 Materials

Alpha-modified minimum essential medium (MEM), Sodium pyruvate, Penicillin Streptomycin,

(Invitrogen – Gibco); 10% Tris Glycine Gel, 1.5mm × 10/15 well (Invitrogen, USA); Purified

Mouse Anti-β-Catenin (BD Biosciences Pharmingen, USA); Monoclonal Anti-β-tubulin clone

TUB 2.1 (Sigma-Aldrich, Inc., USA); Histone H3 antibody (Abcam, Sapphire Bioscience Pty

Ltd, Australia); Polyclonal Swine Anti-rabbit immunoglobulin HRP, polyclonal rabbit Anti-

mouse immunoglobulin HRP ( Dako, Denmark); Detection Reagent (GE Healthcare Limited,

UK); Immobilon Western HRP Substrate Luminol Reagent (Millipore Corporation, USA);

Complete, EDTA-free Protease Inhibitor cocktail tablets, FuGENE HD Transfection Reagent

(Roche Diagnostics GmbH, Germany); Bio-Rad Protein Assay Dye Reagent concentrate (Bio-

Rad Laboratories Inc., USA); Dual-Luciferase Reporter Assay System (Promega Corporation,

USA); 0.05% Trypsin-EDTA 1× (Gibco, Invitogen Australia); Recombinant mouse Wnt-3A,

Recombinant mouse IL-1β, Recombinant mouse TNFα (R&D Systems, Inc., USA); TaqMan

Universal PCR Master Mix (Applied Biosystems, USA); SYBR GREEN PCR Master Mix

(Applied Biosystems, UK); Axin-RFP, TOPFlash/FOPFlash (gift from Dr Maree Faux, Ludwig

Institute for Cancer Research, Victoria, Australia); Renilla (gift from Dr Glen Scholz, University

of Melbourne, Victoria, Australia).

2.2 Cell culture

Mouse pre-osteoblast MC3T3-E1 cells were obtained from Riken – Osaka, Japan. Cells were

maintained in α-modified minimum essential medium (α-MEM) with L-glutamine,

ribonucleosides, deoxyribonucleosides, and supplemented with 5% inactivated fetal calf serum

Page 24: 09-10-26 Thesis

24

(FCS), penicillin (10,000units/ml), streptomycin (10,000μg/ml) and sodium pyruvate (100mM)

in a humidified atmosphere of 5% CO2 in air at 37°C. After reaching confluence, MC3T3-E1

cells were detached and subcultured by treating the cells with trypsin.

2.3 Transfection of plasmids

MC3T3-E1 cells were seeded the day prior to transfection and incubated in the α-MEM culture

media overnight. A master mix of diluted FuGENE HD Reagent was made up by diluting it at a

ratio of 1:15.5 in serum-free α-MEM and allowed to incubate for 5 minutes at room temperature.

Plasmid DNA was mixed with a suitable amount of diluted FuGENE HD reagent, dependent on

the culture plate used, and incubated at room temperature for 15 minutes. After placing fresh

culture medium on the cells, the FuGENE HD Reagent/plasmid DNA transfection cocktail was

added drop wise to the cells. The cells were incubated overnight and the media changed the

following morning. Cells were lysed 48 hours post-transfection.

2.4 Dual-Luciferase Reporter Assay

MC3T3-E1 cells seeded in 12-well plates at a cell density of 2 × 104 cells per well and

transfected with the TOPFlash and FOPFlash plasmids, a gift from Dr Maree Faux from the

Ludwig Institute, the following day. As previously described, a cocktail of transfection reagent

Fugene HD transfection reagent/plasmid and 25μl per well was added drop-wise to 1ml of

culture media. The concentration of the plasmids used is as follows: Fopflash – 0.5μg/well,

Topflash – 0.5μg/well, Renella – 0.01μg/well, ∆45 – 0.5μg/well. 48 hours post transfection, the

cells were first washed twice with PBS then lysed with 200μl/well of Passive Lysis Buffer which

was diluted 1:4 with distilled water and placed in the ˗20°C freezer overnight. Using the rubber

policeman from a syringe, the cells were scraped from the bottom of the well and the lysates

collected. 20μl of each sample was transferred to luminometer tube and the luciferase activity

Page 25: 09-10-26 Thesis

25

measured using Luciferase Assay Substrate (LAR) and Stop & Glo Reagent following

manufacturer’s instructions.

2.5 Nuclear extraction

Cells were subcultured onto 10cm Falcon culture plates at a cell density of 1 × 106 cells per plate

and allowed to grow overnight. At various time points, the cells were stimulated with LiCl (1mM)

or Wnt3a (20ng/ml), or were not treated with either stimulus. Cells were washed twice with ice

cold phosphate buffer solution (PBS) prior extraction of nuclear and cytoplasmic proteins. To

obtain the cytoplasmic proteins, the cells were lysed on ice with a hypotonic solution – 5mM

HEPES pH 7.9, 0.25% (v/v) IGEPAL, 25% (v/v) glycerol, 500mM NaCl, 1.5mM MgCl2, and

0.2mM EDTA. To this ice cold solution 10% (v/v) IGEPAL and 1× CompleteTM

Protease

Inhibitor were added. To harvest the nuclei, the solution was centrifuged at 10,000g, 4°C for 30

seconds. The cytoplasmic supernatant is removed and the nuclear pellet washed with the

hypotonic solution. The nuclear pellet is resuspended in a hypertonic lysis buffer made up of

5mM HEPES pH 7.9, 10mM KCl, and 1.5mM MgCl2, and the nuclear proteins are extracted by

rotating the sample on a rotary mixer for 2 hours at 4°C.

2.6 Osteoblast differentiation

Cells were seeded into 12-well plates at a cell density of 2 × 104 cells per well and incubated

overnight. To induce osteoblastic differentiation, the culture media was removed and replace

with osteogenic media (OM) consisting of α-MEM supplemented with 10% FCS,

dexamethasone (0.1mM), β-glycerophosphate (0.1μM) and ascorbate acid (10μM). The

osteogenic media was changed every 3 days. Cells were also treated with murine IL-1β (1ng/ml)

or murine TNF-α (10ng/ml). The cells were washed 3 times with PBS followed by fixing the

cells with 10% ice cold buffered formalin for 40 minutes. Cells were washed twice with distilled

Page 26: 09-10-26 Thesis

26

water and covered with a filtered alkaline phosphatase stain made up of 0.1M Tris-HCL, napthol

ASMX-PO4 dissolved in N,Ndimethylformamide and Red Violt LB Salt. The plates were placed

in the dark for 30 minutes prior to washing the cells with distilled water and subsequently air

dried and pictures taken by scanning the plates.

2.7 RNA extraction, reverse transcription and real-time polymerase chain reaction

Cells were seeded at a concentration of 0.5 × 106 cells/well in 6 well plates and incubated

overnight before the culture media was replaced with OM. The cells were also treated with LiCl

(1mM), IL-1 (1ng/ml), TNF-α (10ng/ml) and Wnt3a (20ng/ml). After the 3 or 7 day stimulation,

the cells were washed twice with PBS and 350μl of Buffer RLT added to each well to lyse the

cells. The wells were scraped, the cellular extract collected and passed through a 20-gauge

needle using an RNase-free syringe 5 times. The rest of the RNA extraction process was carried

out as per RNeasy Mini Handbook 04/2006 with the removal of any potential DNA

contamination by RNase-free DNase treatment.

Concentrations of RNA extracts were measured and a maximum of 2μg of template RNA was

added to the master mix. The components of the master mix were as set out in the Omiscript

Reverse Transcription Handbook 05/2004 with the following changes – 1μl/reaction Oligo-dT

primer (10μM) and 1μl/reaction Random Primer (3μg/μl). cDNA was synthesised by placing the

RNA/master mix solutions in an incubator at 37°C for 2 hours before being stored at -20°C.

RT-PCR was run for the transcripts of the following genes: ALP, Runx2, Osx, and control gene

TBP. On a 384 well plate, each well was loaded with 5μl of SYBR Green Master mix and 1μl of

the primers. 4μl of diluted cDNA, 1/20 dilution, was transferred into each well. PCR reactions

were conducted on an ABI 7900HT real time PCR machine (Applied Biosystems).

Page 27: 09-10-26 Thesis

27

2.8 Westernblot analysis

To measure the protein concentration of each sample, 5 × Protein Assay Dye Reagent

concentrate was diluted with Milli-Q water to make up 1 × Protein Assay Dye Reagent. 10μl of

each sample and 7.5μl of bovine serum albumin (BSA) standard was added to 1.5ml of 1 ×

Protein Assay Dye Reagent. The final concentration of BSA in the Protein Assay Dye Reagent is

5μg/μl. The absorbance of each sample was determined using the Bio-Rad SmartSpec 3000 and

the BSA standard was used to determine the concentration for each sample.

10%, 1.5mm Tris Glycine gels (Invitrogen) were loaded with a total of 40μg of protein/well and

15μl of each sample into each well. The gels were run at 150V for 1.5 hours, till the dye front

was close to the bottom of the gel and the gels were transferred to PDF membranes at 100V for

40 minutes. After blocking the membrane for 1 hour with 3% BSA in TBST, the membrane was

incubated overnight at 4°C with the primary antibody which was diluted in 5ml of 1% BSA in

TBST. The dilutions for each primary antibody are as follows: anti β-catenin – 1/5000, anti β-

tubulin – 1/106, anti histone H3 – 1/1000, anti actin – 1/5000. The membrane was washed 3

times, for 15 minutes each, with 1 × TBST then incubated for 1 hour with the appropriate

secondary antibody in 10ml of 1% BSA in TBST. After the membrane was washed another 3

times in 1 × TBST, the membrane was covered with a mixture of equal amounts of Detection

Reagents 1 and 2 (GE Healthcare Limited, UK) or Immobilon Western HRP Substrate Luminol

Reagents ( Millipore Corporation, USA) – depending on the strength of the primary antibody.

Page 28: 09-10-26 Thesis

28

Chapter 3: Results

3.1 Determining the optimal concentration of TOPFlash and FOPFlash plasmids

Initial experiments were performed to determine the optimal ratio of TOPFlash plasmid to the

Renilla control plasmid. TOPFlash is a plasmid that has 7 Tcf/Lef binding sites upstream to a

thymidine promoter that drives the transcription of the firefly luciferase gene. Hence, the firefly

luciferase gene will only be transcribed in the presence of β-catenin, which binds to Tcf/Lef. The

FOPflash plasmid is similar to the TOPFlash plamid, but with mutated Tcf/Lef binding sites and

thus acts as a negative control. Renilla is also another control plasmid which induces the

constitutive transcription of the Renilla luciferase gene, acting as a transfection efficiency control.

The Dual Luciferase Reporter Assay was used to measure firefly and Renilla luciferase activity,

with the firefly activity standardised against the Renilla control luciferase activity. All treatments

were either transfected with TOPFlash or FOPFlash, together with Renilla, and the data for

FOPFlash is not shown as luciferase/Renilla ratios were consistently low (as expected). The

following concentrations of TOP and FOPFlash plasmids were tested – 0.5μg, 0.25 μg and 0.1μg

per well. The optimal amount of 0.01μg of the Renilla plasmid that was used to transfect cells in

each well had been determined by previous experiments conducted by other members of the

laboratory. A mutant β-catenin overexpression plasmid called ∆45, which encodes a mutant form

of β-catenin that cannot be degraded by the Axin-Gsk3β destructive complex, was used to

specifically activate the TOPFlash reporter plasmid. Figure 2 shows that the optimal amount of

TOP and FOPFlash per well was 0.5μg.

3.2 Effects of ∆45, IL-1β and TNF-α on nuclear β-catenin activity

Preliminary reporter assays conducted, using a range of ∆45 plasmid concentrations in MC3T3-

E1 cells, indicate that the greatest fold change in firefly luciferase activity was obtained by

Page 29: 09-10-26 Thesis

29

transfecting cells with 0.25μg of the ∆45 plasmid (Figure 3). All subsequent reporter assays used

this concentration for ∆45. IL-1β and TNF-α treatment caused a reduction in the firefly luciferase

activity induced by ∆45 (Figure 4).

3.3 LiCl increases β-catenin activity

Lithium chloride (LiCl) is a non-specific inhibitor of GSK3β and therefore non-specifically

activates the Wnt/β-catenin pathway. LiCl treatment resulted in an increased fold change in the

luciferase activity indicating that there was an overall increase in β-catenin binding to the

Tcf/Lef binding sites (Figure 5). Initial experiments with cells treated with either IL-1β or TNF-α,

in the presence of LiCl, showed that there was a decrease in TOPFlash reporter activity when

compared to cells treated with LiCl alone (Figure 5A). However, later experiments, using cells

that had undergone serial passaging, seem to contradict these results. The cells treated with LiCl

and IL-1β showed an increased fold change in the luciferase activity (Figure 5B), while the

effects of TNF-α remained the same.

3.4 The effects of Wnt3a, IL-1β and TNF-α on β-catenin activity

Wnt3a constantly induced the accumulation of β-catenin in the nucleus resulting in the increased

firefly luciferase activity when compared to the control (Figure 6). This shows that the Wnt3a

ligand is able to activate the canonical Wnt pathway, which requires β-catenin for signalling, in

these cells.

Similar to earlier LiCl results, early reporter assays showed IL-1β and TNF-α were able to

decrease firefly luciferase activity when cells were co-treated with Wnt3a and either of the

cytokines. TNF-α reduced the Wnt3a-induced firefly luciferase activity to the greatest extent –

resulting in similar firefly luciferase activity seen in the control (Figure 6A). These results

Page 30: 09-10-26 Thesis

30

seemed to indicate that these pro-inflammatory cytokines are able to inhibit the activation of the

β-catenin specific reporter, accumulation of β-catenin within the nucleus and therefore blocking

the Wnt pathway. However, subsequent reporter assays carried out to confirm these results

contradicted the initial findings – TNF-α caused no change in firefly luciferase activity while

treatment with IL-1β seemed to cause a 5 fold increase the firefly luciferase activity compared to

cells treated with Wnt3a (Figure 6C).

However, in the later experiments the axin-RFP plasmid was used as a positive control for the

inhibition of the Wnt/ β-catenin pathway, and over expression of axin-RFP initially caused a

decrease in β-catenin activity (Figure 6B). Axin is crucial for the formation of the β-catenin

destructive complex. By treating the cells with Wnt3a after transfection with the axin-RFP

plasmid, it was determined that the optimal transfection amount of axin-RFP plasmid for the

reporter assay was 0.05μg per transfection. These results indicated that overexpression axin-RFP

was able to inhibit Wnt-induced firefly luciferase activity (Figure 6B). Despite these preliminary

experiments indicating the inhibition of the Wnt/β-catenin pathway using axin-RFP, later

experiments conducted showed an increase in firefly luciferase activity in axin-transfected cells,

suggesting possible errors had occurred in one or more aspects of the experiment (Figure 6C).

3.5 ∆45 did not increase β-catenin levels in whole cell lysates

To confirm the results from luciferase assays showing an increase in β-catenin when MC3T3-E1

cells are transfected with ∆45 (mutant β-catenin plasmid) (Figures 3 and 4), western blots were

conducted using whole cell lysates from MC3T3-E1 cells that had been transfected with

increasing amounts of ∆45 (Figure 7). Results indicate no detectable change in β-catenin levels

as band intensity remained constant.

Page 31: 09-10-26 Thesis

31

3.6 Determining the optimal stimulation period prior to nuclear extraction

Determining changes in the levels of β-catenin in whole cell lysates is very difficult therefore β-

catenin levels were measured in nuclear extracts from treated cells to observe changes in the

levels of nuclear β-catenin. To determine the best time to obtain maximal change in nuclear β-

catenin levels, cells were stimulated over a range of time points – 1,2,4,6,and 24 hours, with

either LiCl or Wnt3a (Figure 8). The band intensity was greatest for the 2 hour stimulation when

compared to untreated cells and was thus used as one of the suitable time points to obtain

maximal β-catenin stimulation. The 6 hour time point was also chosen as another time point.

3.7 Nuclear β-catenin levels in cells treated with LiCl were not influenced by IL-1β or TNF-α

Initial western blots using nuclear lysates from cells treated with LiCl showed an increase in

nuclear β-catenin levels with stronger band intensity when cells were stimulated for 2hrs (Figure

8A). However, subsequent western blots using nuclear extracts from cells treated with either IL-

1β or TNF-α, in the presence of LiCl, showed no detectable change in nuclear β-catenin levels

(Figure 9) when compared to extracts from cells treated with LiCl alone. Furthermore, LiCl

alone failed to increase β-catenin levels in the nucleus.

3.8 Nuclear β-catenin levels – treatments with IL-1β or TNF-α and Wnt3a

Western blots show that the band intensity of nuclear extracts from cells treated with Wnt3a

alone, for 2 hours, was similar to bands using nuclear extracts from cells treated with either IL-

1β or TNF-α, in the presence of Wnt3a, indicating there was no detectable change in nuclear β-

catenin levels despite treatment with the pro-inflammatory cytokines (Figure 9). In addition,

Wnt3a alone failed to increase β-catenin levels in the nucleus. Similar results were obtained

using cytoplasmic and nuclear extracts from cells treated for 6 hours (Figure 9).

Page 32: 09-10-26 Thesis

32

3.9 Determining the incubation time period for optimal ALP staining

Alkaline phosphatase (ALP) is a marker of osteoblast differentiation. To determine whether IL-

1β and TNF-α were inhibiting the biological effects of Wnt pathway activation, ALP expression

was measured as a marker of MC3T3-E1 differentitation from a pre-osteoblast state to a

differentiated osteoblast state by staining for ALP.

In the process of determining the effects of IL-1β and TNF-α, each osteoblast differentiation

assay was conducted in duplicate. Initially, 2 sets of the same assay would be differentiated after

being incubated in osteogenic media for 1 week or 2 weeks. Upon the realisation that ALP

staining had reached maximal intensity after 1 week, subsequent assays were differentiated at

either 5 days or 1 week. The 5 day incubation in ostegenic media gave the best results –

maximum difference in staining intensity between osteogenic media and normal media.

3.10 IL-1β and TNF-α decrease ALP staining in ∆45-transfected cells

Earlier reporter assays using cells transfected with ∆45 and treated with either IL-1β or TNF-α

showed a decreased in firefly luciferase activity, indicating a reduction in the accumulation of β-

catenin activity (Figure 4). Osteoblast differentiation assays also support this finding with the

transfected cells treated with IL-1β or TNF-α showing a decrease in ALP staining when

compared to wells treated ∆45 alone. However, wells treated with ∆45 alone failed to show an

additive effect on ALP staining, showing similar staining intensity to the control wells (Figure

10A). 3.11 IL-1β and TNF-α decreased ALP staining in LiCl-treated cells

Both IL-1β and TNF-α were able to reduce the intensity of ALP staining seen in wells treated

with LiCl (Figure 10B). However, LiCl treated cells had no additional effect on ALP staining

compared to osteogenic media alone.

Page 33: 09-10-26 Thesis

33

3.12 Wnt3a inhibits osteoblast differentiation

Cells were treated with osteogenic media for 7 days in the presence or absence of Wnt3a, axin-

RFP plasmid, IL-1β and TNF-α. Wnt3a appeared to inhibit ALP staining while the axin-RFP

plasmid had no effect on ALP staining (Figure 11). Both IL-1β and TNF-α inhibited ALP

staining alone and in the presence of Wnt3a (Figure 11B).

3.13 Change in osteoblastic gene expression with LiCl or Wnt3a and IL-1β or TNF-α

The expression of several known osteoblastic marker genes can be quantitatively measured via

real-time-PCR (RT-PCR). RNA was extracted from cells treated for 3 or 7 days in osteogenic

media treated with a combination treatments with LiCl, Wnt3a, IL-1β or TNF-α. The expression

of different osteoblast genes – Runx2, Osterix (Osx) and ALP was measured. The TATA binding

protein (TBP) was used as a control gene and was used in analysis of the results to determine

differential expression of the genes.

Treatment of the cells for 3 days in LiCl did not show a significant change in gene expression for

Runx2, Osx or ALP when compared to negative control. However, a general pattern showing a

moderate decrease when co-treated with IL-1β and a greater reduction with TNF-α was observed

in the expression of all 3 genes (Figure 12).

As with the treatment with LiCl, Wnt3a treatment for 3 days did not induce a significant change

in any of the osteoblastic genes. Unlike the results observed in cells treated with LiCl, a

reduction in the expression was only observed in ALP when cells were treated with either IL-1β

or TNF-α in the presence of Wnt3a. Expression of Runx2 and Osx after treatment with Wnt3a

for 3 days did not seem to be affected, though a minor increase in their expression was observed

when co-treated with IL-1β. Furthermore, there was no difference seen in the expression in either

Page 34: 09-10-26 Thesis

34

of the 2 genes when comparing treatments with IL-1β or TNF-α. However, Osx expression was

lowered to the same extent by both IL-1β and TNF-α. Interestingly, treatment with TNF-α on its

own induced a greater reduction in ALP expression compared to treating cells only with IL-1β.

As seen with the expression of the osteoblastic genes using cells that have been treated for 3 days,

there did not seem to be a significant change in any of the genes using cells that had been treated

for 7 days with either LiCl or Wnt3a. Treatment of the cells for 7 days in osteogenic media with

either IL-1β or TNF-α, in the presence of LiCl, resulted in similar fold changes in the expression

of Runx2, Osx and ALP (Figure 13). This was also observed for Runx2 and Osx in cells treated

with Wnt3a. However, in cells treated with Wnt3a, the expression of ALP seemed to be reduced

to a greater extent when treated with IL-1β as compared to co-treatment with TNF-α.

Page 35: 09-10-26 Thesis

35

Chapter 4: Discussion

The Wnt pathway is thought to play a role in the differentiation of MSCs into osteoblasts

based on studies showing Wnt pathway activity in MSCs and osteoblasts. However, many

studies do not use specific Wnt ligands to investigate Wnt signalling. In this project, a specific

Wnt ligand – Wnt3a was used to stimulate murine calvaria pre-osteoblast MC3T3-E1 cells to

study the effects that pro-inflammatory cytokines, IL-1β and TNF-α have on the Wnt pathway.

Given that IL-1β and TNF-α can inhibit stem cells differentiation into osteoblasts (Lacey et al.,

2009), it was hypothesised that IL-1β and TNF-α may inhibit the Wnt/β-catenin pathway and

thereby block stem cell differentiating into osteoblasts.

To test this hypothesis, the Wnt/β-catenin pathway was activated using either a β-catenin

overexpression plasmid (∆45), LiCl or Wnt3a, while the pathway was specifically inhibited

using the axin-RFP overexpressing plasmid. The Wnt/β-catenin pathway was measured under the

above the conditions in the presence or absence of IL-1β or TNF-α.

The results from the dual luciferase reporter assay which measures β-catenin activity are

inconclusive as early experiments showed increases in β-catenin activity in the presence of LiCl,

Wnt3a and the β-catenin overexpressing plasmid ∆45. While the axin-RFP plasmid, which is

crucial to the formation of the β-catenin destructive complex, showed the expected decrease in β-

catenin activity, IL-1β and TNF-α also showed similar decreases. However, later experiments

using cells transfected with the axin-RFP plasmid and treated with both IL-1β and TNF-α did not

show the expected decrease in the firefly luciferase activity. This was surprising as the axin-RFP

plasmid, which was a gift from Dr Maree Faux, was also used by Faux et al. and they

successfully showed the destruction of β-catenin by the expression of axin-RFP (Faux et al.,

Page 36: 09-10-26 Thesis

36

2008). However, our early experimental testing using different axin-RFP plasmid concentrations

also showed a decrease in Wnt activity (Figure 6B). As axin-RFP is a control for reducing Wnt

activity, this suggests possible experimental errors occurred as the axin-RFP plasmid should

inhibit Wnt activity.

A possible explanation for these differing results could be due to differentiation of the

pre-osteoblast cells that has attributed to the number of passages the cells had undergone before

they were used in these experiments. While the passage number for these cells was not noted, the

morphology of the cells was observed before the plating of the cells. In experiments where IL-1β

was able to reduce the nuclear β-catenin levels in the presence of LiCl or Wnt3a (Figure 5A, 6A),

the cells were observed to be round. However, cells used in experiments where there was an

increase in the fold change by both IL-1β and TNF-α in the presence of LiCl or Wnt3a (Figure

5B, 6C), the cells appeared to be differentiated, adopting an elongated, more fibroblast-like

shape. These observations seem to indicate that the cells were at different stages of

differentiation and are similar to the cell morphology seen in other studies using early and late

passage MC3T3-E1 cells (Chung et al., 1999). Furthermore, the study by Chun et al. suggests

that serial passaging of MC3T3-E1 cells reduced osteoblastic function with lowered alkaline

phosphatase (ALP) activity and osteocalcin secretion. Other studies using human osteoblasts

indicate ALP mRNA levels from samples obtained from older donors were lower than those

from younger donors (Sutherland et al., 1995).

The lack of inhibition by the axin-RFP plasmid could be due to the plasmid not being

expressed in the cells, though this is unlikely as both the TOPFlash and Renilla plasmids were

expressed. However, as our positive control for the inhibition of the Wnt pathway did not work,

Page 37: 09-10-26 Thesis

37

it suggests that all the results from these experiments are not valid. Additional measures such as

using green fluorescent protein (GFP) to transfect the cells could be taken to avoid transfection

inefficiency speculation.

TNF-α consistently blocks the Wnt/β-catenin pathway in all experiments – in LiCl treated

cells this is likely due to cell death, as there was an increase in dead cells seen in wells co-treated

with LiCl and TNF-α. In other studies using TNF-sensitive cells, LiCl has been shown to cause

an increase in TNF-cytotoxicity in a dose-dependent manner, while the concentrations of LiCl

used did not affect cell survival when the cells were only treated with LiCl (Beyaert et al., 1989).

This effect of LiCl on the MC3T3-E1 cells line used for this project was similar to that seen by

Beyaert et al. In observations of cells co-treated with Wnt3a and TNF-α, there did not appear to

be an increase in cell death, however this was not specifically measured. Therefore although

TNF-α blocked Wnt3a activation of the TOPFlash reporter, cell survival would need to be

measured to ensure that this inhibition is not due to possible cytotoxic effects of TNF-α on this

cell line.

Given that both IL-1β and TNF-α inhibited ∆45-induced increase of firefly luciferase

activity in the reporter assay and if this result is real, then these cytokines are possibly up

regulating a transcriptional regulator of the β-catenin/Tcf transcription. Any inhibition of the

Wnt/β-catenin pathway above the level of β-catenin should have no effect on the ∆45 expressing

cells as ∆45-β-catenin cannot be degraded by the destructive complex. Therefore, in order for IL-

1β and TNF-α to inhibit the β-catenin signal in these cells, they would need to up-regulate a

Tcf/Lef antagonist such as NLK or groucho. Therefore further experiments need to be done to

confirm these results and to determine the possible mechanism of inhibition. However, it is still

Page 38: 09-10-26 Thesis

38

possible that IL-1β and TNF-α could regulate this pathway at multiple levels, and thus upregulate

Wnt antagonists such as Dkk-1.

To further investigate the effects of Wnt3a, IL-1β and TNF-α on the Wnt pathway,

western blots using whole cell lysates, cytoplasmic and nuclear extracts were conducted. Initial

western blots using whole cells lysates from cells that had been transfected with increasing

amounts of ∆45 (a plasmid that causes the overexpression of β-catenin) did not show a detectable

increase in β-catenin. This observation could be due to the fact that there are large pools of

different β-catenin isoforms found within cells, and that most commercial anti- β-catenin

antibodies detect all isoforms of β-catenin. Furthermore, both phosphorylated β-catenin and

active β-catenin are known to interact with cadherins that are found at cell adhesions (Maher et

al., 2009), as such any small changes in β-catenin levels that signal to the nucleus would be very

difficult to detect using whole cell lysates. Hence, to detect any changes in β-catenin levels, it

was decided that nuclear extracts could provide better results with the majority of β-catenin

sequestered within the cytoplasmic fraction.

After establishing that stimulation with LiCl or Wnt3a for a period of 2 hours results in a

greater increase in the amount of β-catenin detected via western blot (Figure 8), subsequent

western blots conducted used cytoplasmic and nuclear extracts from cells that had been

stimulated for 2 and 6 hours. However, detection using anti-β-catenin antibodies failed to show

any significant increase in band intensity in nuclear lysates from cells that were treated with

either LiCl or Wnt3a (Figure 9). While there could be speculation that there could be residual

cytoplasmic proteins in the nuclear extracts obtained, the anti-β-tubulin control antibody has

clearly shown no detection of any β-tubulin. β-tubulin is required for the assembly of

Page 39: 09-10-26 Thesis

39

microtubules which is involved in cell motility, and as such is used a loading control for

cyotplasmic proteins. Likewise, a control for the nuclear extracts, anti-histone H3 antibodies,

was used to ensure that there was no cross-contamination of proteins of the 2 separate fractions

during the extraction process. The observed changes in band intensity in the preliminary

westernblots using the nuclear extracts ascertain the best time point for nuclear extraction could

have been arbitrary as the anti-histone H3 loading control had not been used. Without the loading

control, there is no means to determine if the amount of sample loaded in each well was

consistent and that the observed changes in band intensity with the anti-β-catenin were real.

However, based on the subsequent western blots, which indicate no change in β-catenin levels

regardless of the treatment, this could indicate that the initial change in band intensity seen was

due to inconsistent loading of samples. Repeating western blots, with loading controls, using

nuclear extracts from cells stimulated over various time points would provide an indication as to

whether further investigation in this direction would be worthwhile.

Wnt3a has been implicated in promoting skeletal development and playing a role in the

differentiation of MSCs into osteoblasts. ALP expression is used as an osteoblastic marker and

many studies use osteoblast differentiation assays to determine the extent of ALP expression by

cells that have been subjected to different stimulus. The results from the osteoblast

differentiation assays conducted as part of this project appear to show that Wnt3a is inhibiting

the differentiation of osteoblasts (Figure 11). This is seen by the reduced ALP staining intensity

when the MC3T3-E1 cells are transfected with the axin-FRP plasmid and treated with Wnt3a

when compared to the wells with cells only transfected with axin-RFP. This osteoblast inhibition

by Wnt3a has also been seen in other studies staining for ALP expression (De Boer et al., 2004).

However, since the cells are cultured in osteogenic media, there should be a decrease in the

Page 40: 09-10-26 Thesis

40

intensity of ALP staining in the cells transfected with the axin-FRP plasmid alone when

compared to the control wells, which was not observed in these experiments. This could indicate

that the effect of β-catenin destructive complex is minimal in cells that have already begun to

differentiate as the cells used in these experiments had been passaged a number of times. This is

in concordance with results seen in experiments previously conducted on hMSCs treated with

conditioned media from Wnt3a-secreting cells (Boland et al., 2004).

The osteogenic genes Runx2/Cbfa1, osterix (Osx) and ALP were measured in cells

treated for 3 days with LiCl, Wnt3a, IL-1β and TNF-α. Real-time PCR (RT-PCR) results suggest

that there was no significant time-dependent effect on the change in osteoblastic gene expression,

while there seemed to be a descending fold change in the expression of all genes when treated

with LiCl for 3 days. A small increase in fold change was observed when the cells were treated

with Wnt3a and IL-1β reflecting the reduction in firefly luciferase activity seen with the same

treatment in the reporter assays as mentioned above. However, the fold changes seen in gene

expression were minimal and not significantly higher or lower than the expression of these genes

in the control.

There is a possibility that the rate of differentiation the MC3T3-E1 cells had influenced

the outcomes of the experiment conducted in this project. Experiments conducted by Khatiwala

et al. to determine how the rigidity of the extracellular matrix affects osteoblast differentiation

have shown that MC3T3-E1 cells grown on polystyrene had a greater extent of differentiation

compared to cells grown on less rigid collagen substrates (Khatiwala et al., 2006). In the same

study, they also managed to show that, when cells were grown on polystyrene, cell density was

almost double the density of cells grown on the hydrogels with the least collagen density, over

Page 41: 09-10-26 Thesis

41

the same period of time. The greater ALP staining observed in this project, using MC3T3-E1

cells that had been passaged a greater amount of times, could partly be due to the increased

differentiation and rapid proliferation of the cells on plastic wells.

Results obtained from the experiments conducted so far are inconclusive with regards to

the effects of IL-1β and TNF-α on the Wnt pathway. However, it has been observed that the cell

passage number of the MC3T3-E1 cells and hence their differentiated state, seems to play an

important role in the osteoblast phenotype observed, and the effects seen by Wnt3a, IL-1β and

TNF- α on these cells. These experiments suggest that using MC3T3-E1 cells that have been

serially passaged, over an extended period of time, result in their spontaneous differentiation

towards osteoblasts. Experiments conducted with MC3T3-E1 cells that have been freshly

cultured from liquid nitrogen, and hence closer to a stem cell line state yield different results,

suggesting that cytokines have different effects on the stages of differentiation. In addition, using

a member of different stem cell lines would also be advantageous.

Further investigation into the effects of IL-1β and TNF-α on the Wnt pathway using cells

with different passage numbers could provide an insight to the effectiveness of current anti-

cytokine therapy at different stages of severity in rheumatoid arthritis and other autoimmune

diseases.

Page 42: 09-10-26 Thesis

42

References

Bafico, A., Liu, G., Yaniv, A., Gazit, A., and Aaronson, S.A. (2001). Novel mechanism of Wnt signalling

inhibition mediated by Dickkopf-1 interaction with LRP6/Arrow. Nat Cell Biol 3, 683-686.

Barker, N., Hurlstone, A., Musisi, H., Miles, A., Bienz, M., and Clevers, H. (2001). The chromatin

remodelling factor Brg-1 interacts with beta-catenin to promote target gene activation. EMBO J 20, 4935-

4943.

Bertolini, D.R., Nedwin, G.E., Bringman, T.S., Smith, D.D., and Mundy, G.R. (1986). Stimulation of

bone resorption and inhibition of bone formation in vitro by human tumour necrosis factors. Nature 319,

516-518.

Beyaert, R., Vanhaesebroeck, B., Suffys, P., Van Roy, F., and Fiers, W. (1989). Lithium chloride

potentiates tumor necrosis factor-mediated cytotoxicity in vitro and in vivo. Proc Natl Acad Sci U S A 86,

9494-9498.

Boland, G.M., Perkins, G., Hall, D.J., and Tuan, R.S. (2004). Wnt 3a promotes proliferation and

suppresses osteogenic differentiation of adult human mesenchymal stem cells. J Cell Biochem 93, 1210-

1230.

Brennan, F.M., Chantry, D., Jackson, A., Maini, R., and Feldmann, M. (1989). Inhibitory effect of TNF

alpha antibodies on synovial cell interleukin-1 production in rheumatoid arthritis. Lancet 2, 244-247.

Caplan, A.I. (1991). Mesenchymal stem cells. J Orthop Res 9, 641-650.

Cavallo, R.A., Cox, R.T., Moline, M.M., Roose, J., Polevoy, G.A., Clevers, H., Peifer, M., and Bejsovec,

A. (1998). Drosophila Tcf and Groucho interact to repress Wingless signalling activity. Nature 395, 604-

608.

Chen, Y., Whetstone, H.C., Youn, A., Nadesan, P., Chow, E.C., Lin, A.C., and Alman, B.A. (2007). Beta-

catenin signaling pathway is crucial for bone morphogenetic protein 2 to induce new bone formation. J

Biol Chem 282, 526-533.

Chu, C.Q., Field, M., Allard, S., Abney, E., Feldmann, M., and Maini, R.N. (1992). Detection of

cytokines at the cartilage/pannus junction in patients with rheumatoid arthritis: implications for the role of

cytokines in cartilage destruction and repair. Br J Rheumatol 31, 653-661.

Chung, C.Y., Iida-Klein, A., Wyatt, L.E., Rudkin, G.H., Ishida, K., Yamaguchi, D.T., and Miller, T.A.

(1999). Serial passage of MC3T3-E1 cells alters osteoblastic function and responsiveness to transforming

growth factor-beta1 and bone morphogenetic protein-2. Biochem Biophys Res Commun 265, 246-251.

Cong, F., Schweizer, L., and Varmus, H. (2004). Casein kinase Iepsilon modulates the signaling

specificities of dishevelled. Mol Cell Biol 24, 2000-2011.

Cong, F., and Varmus, H. (2004). Nuclear-cytoplasmic shuttling of Axin regulates subcellular localization

of beta-catenin. Proc Natl Acad Sci U S A 101, 2882-2887.

da Silva Meirelles, L., Chagastelles, P.C., and Nardi, N.B. (2006). Mesenchymal stem cells reside in

virtually all post-natal organs and tissues. J Cell Sci 119, 2204-2213.

Daniels, D.L., and Weis, W.I. (2005). Beta-catenin directly displaces Groucho/TLE repressors from

Tcf/Lef in Wnt-mediated transcription activation. Nat Struct Mol Biol 12, 364-371.

De Boer, J., Wang, H.J., and Van Blitterswijk, C. (2004). Effects of Wnt signaling on proliferation and

differentiation of human mesenchymal stem cells. Tissue Eng 10, 393-401.

Dinarello, C.A., Cannon, J.G., Wolff, S.M., Bernheim, H.A., Beutler, B., Cerami, A., Figari, I.S.,

Palladino, M.A., Jr., and O'Connor, J.V. (1986). Tumor necrosis factor (cachectin) is an endogenous

pyrogen and induces production of interleukin 1. J Exp Med 163, 1433-1450.

Dominici, M., Le Blanc, K., Mueller, I., Slaper-Cortenbach, I., Marini, F., Krause, D., Deans, R., Keating,

A., Prockop, D., and Horwitz, E. (2006). Minimal criteria for defining multipotent mesenchymal stromal

cells. The International Society for Cellular Therapy position statement. Cytotherapy 8, 315-317.

Ducy, P., and Karsenty, G. (1995). Two distinct osteoblast-specific cis-acting elements control expression

of a mouse osteocalcin gene. Mol Cell Biol 15, 1858-1869.

Page 43: 09-10-26 Thesis

43

Ducy, P., Zhang, R., Geoffroy, V., Ridall, A.L., and Karsenty, G. (1997). Osf2/Cbfa1: a transcriptional

activator of osteoblast differentiation. Cell 89, 747-754.

Evans, M.J., and Kaufman, M.H. (1981). Establishment in culture of pluripotential cells from mouse

embryos. Nature 292, 154-156.

Fagotto, F., Jho, E., Zeng, L., Kurth, T., Joos, T., Kaufmann, C., and Costantini, F. (1999). Domains of

axin involved in protein-protein interactions, Wnt pathway inhibition, and intracellular localization. J Cell

Biol 145, 741-756.

Faux, M.C., Coates, J.L., Catimel, B., Cody, S., Clayton, A.H., Layton, M.J., and Burgess, A.W. (2008).

Recruitment of adenomatous polyposis coli and beta-catenin to axin-puncta. Oncogene 27, 5808-5820.

Fontana, A., Hengartner, H., Weber, E., Fehr, K., Grob, P.J., and Cohen, G. (1982). Interleukin 1 activity

in the synovial fluid of patients with rheumatoid arthritis. Rheumatol Int 2, 49-53.

Friedenstein, A.J., Chailakhjan, R.K., and Lalykina, K.S. (1970). The development of fibroblast colonies

in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kinet 3, 393-403.

Friedenstein, A.J., Piatetzky, S., II, and Petrakova, K.V. (1966). Osteogenesis in transplants of bone

marrow cells. J Embryol Exp Morphol 16, 381-390.

Gowen, M., Wood, D.D., Ihrie, E.J., McGuire, M.K., and Russell, R.G. (1983). An interleukin 1 like

factor stimulates bone resorption in vitro. Nature 306, 378-380.

Hart, M., Concordet, J.P., Lassot, I., Albert, I., del los Santos, R., Durand, H., Perret, C., Rubinfeld, B.,

Margottin, F., Benarous, R., et al. (1999). The F-box protein beta-TrCP associates with phosphorylated

beta-catenin and regulates its activity in the cell. Curr Biol 9, 207-210.

Hecht, A., Vleminckx, K., Stemmler, M.P., van Roy, F., and Kemler, R. (2000). The p300/CBP

acetyltransferases function as transcriptional coactivators of beta-catenin in vertebrates. EMBO J 19,

1839-1850.

Hino, S., Michiue, T., Asashima, M., and Kikuchi, A. (2003). Casein kinase I epsilon enhances the

binding of Dvl-1 to Frat-1 and is essential for Wnt-3a-induced accumulation of beta-catenin. J Biol Chem

278, 14066-14073.

Hopkins, S.J., and Meager, A. (1988). Cytokines in synovial fluid: II. The presence of tumour necrosis

factor and interferon. Clin Exp Immunol 73, 88-92.

Ishitani, T., Ninomiya-Tsuji, J., and Matsumoto, K. (2003). Regulation of lymphoid enhancer factor 1/T-

cell factor by mitogen-activated protein kinase-related Nemo-like kinase-dependent phosphorylation in

Wnt/beta-catenin signaling. Mol Cell Biol 23, 1379-1389.

Ito, T., Sawada, R., Fujiwara, Y., and Tsuchiya, T. (2008). FGF-2 increases osteogenic and chondrogenic

differentiation potentials of human mesenchymal stem cells by inactivation of TGF-beta signaling.

Cytotechnology 56, 1-7.

Joosten, L.A., Helsen, M.M., Saxne, T., van De Loo, F.A., Heinegard, D., and van Den Berg, W.B.

(1999). IL-1 alpha beta blockade prevents cartilage and bone destruction in murine type II collagen-

induced arthritis, whereas TNF-alpha blockade only ameliorates joint inflammation. J Immunol 163,

5049-5055.

Khatiwala, C.B., Peyton, S.R., and Putnam, A.J. (2006). Intrinsic mechanical properties of the

extracellular matrix affect the behavior of pre-osteoblastic MC3T3-E1 cells. Am J Physiol Cell Physiol

290, C1640-1650.

Klein, T.J., Jenny, A., Djiane, A., and Mlodzik, M. (2006). CKIepsilon/discs overgrown promotes both

Wnt-Fz/beta-catenin and Fz/PCP signaling in Drosophila. Curr Biol 16, 1337-1343.

Komori, T., Yagi, H., Nomura, S., Yamaguchi, A., Sasaki, K., Deguchi, K., Shimizu, Y., Bronson, R.T.,

Gao, Y.H., Inada, M., et al. (1997). Targeted disruption of Cbfa1 results in a complete lack of bone

formation owing to maturational arrest of osteoblasts. Cell 89, 755-764.

Kramps, T., Peter, O., Brunner, E., Nellen, D., Froesch, B., Chatterjee, S., Murone, M., Zullig, S., and

Basler, K. (2002). Wnt/wingless signaling requires BCL9/legless-mediated recruitment of pygopus to the

nuclear beta-catenin-TCF complex. Cell 109, 47-60.

Page 44: 09-10-26 Thesis

44

Lacey, D.C., Simmons, P.J., Graves, S.E., and Hamilton, J.A. (2009). Proinflammatory cytokines inhibit

osteogenic differentiation from stem cells: implications for bone repair during inflammation.

Osteoarthritis Cartilage 17, 735-742.

Li, L., Yuan, H., Weaver, C.D., Mao, J., Farr, G.H., 3rd, Sussman, D.J., Jonkers, J., Kimelman, D., and

Wu, D. (1999). Axin and Frat1 interact with dvl and GSK, bridging Dvl to GSK in Wnt-mediated

regulation of LEF-1. EMBO J 18, 4233-4240.

Liu, C., Kato, Y., Zhang, Z., Do, V.M., Yankner, B.A., and He, X. (1999). beta-Trcp couples beta-catenin

phosphorylation-degradation and regulates Xenopus axis formation. Proc Natl Acad Sci U S A 96, 6273-

6278.

Maher, M.T., Flozak, A.S., Stocker, A.M., Chenn, A., and Gottardi, C.J. (2009). Activity of the beta-

catenin phosphodestruction complex at cell-cell contacts is enhanced by cadherin-based adhesion. J Cell

Biol 186, 219-228.

Majors, A.K., Boehm, C.A., Nitto, H., Midura, R.J., and Muschler, G.F. (1997). Characterization of

human bone marrow stromal cells with respect to osteoblastic differentiation. J Orthop Res 15, 546-557.

Mao, B., Wu, W., Davidson, G., Marhold, J., Li, M., Mechler, B.M., Delius, H., Hoppe, D., Stannek, P.,

Walter, C., et al. (2002). Kremen proteins are Dickkopf receptors that regulate Wnt/beta-catenin

signalling. Nature 417, 664-667.

Mao, B., Wu, W., Li, Y., Hoppe, D., Stannek, P., Glinka, A., and Niehrs, C. (2001). LDL-receptor-related

protein 6 is a receptor for Dickkopf proteins. Nature 411, 321-325.

Martin, M.J., Muotri, A., Gage, F., and Varki, A. (2005). Human embryonic stem cells express an

immunogenic nonhuman sialic acid. Nat Med 11, 228-232.

Nakamura, T., Hamada, F., Ishidate, T., Anai, K., Kawahara, K., Toyoshima, K., and Akiyama, T. (1998).

Axin, an inhibitor of the Wnt signalling pathway, interacts with beta-catenin, GSK-3beta and APC and

reduces the beta-catenin level. Genes Cells 3, 395-403.

Nakashima, K., Zhou, X., Kunkel, G., Zhang, Z., Deng, J.M., Behringer, R.R., and de Crombrugghe, B.

(2002). The novel zinc finger-containing transcription factor osterix is required for osteoblast

differentiation and bone formation. Cell 108, 17-29.

Ng, F., Boucher, S., Koh, S., Sastry, K.S., Chase, L., Lakshmipathy, U., Choong, C., Yang, Z., Vemuri,

M.C., Rao, M.S., et al. (2008). PDGF, TGF-beta, and FGF signaling is important for differentiation and

growth of mesenchymal stem cells (MSCs): transcriptional profiling can identify markers and signaling

pathways important in differentiation of MSCs into adipogenic, chondrogenic, and osteogenic lineages.

Blood 112, 295-307.

Nusslein-Volhard, C., and Wieschaus, E. (1980). Mutations affecting segment number and polarity in

Drosophila. Nature 287, 795-801.

Otto, F., Thornell, A.P., Crompton, T., Denzel, A., Gilmour, K.C., Rosewell, I.R., Stamp, G.W.,

Beddington, R.S., Mundlos, S., Olsen, B.R., et al. (1997). Cbfa1, a candidate gene for cleidocranial

dysplasia syndrome, is essential for osteoblast differentiation and bone development. Cell 89, 765-771.

Owen, T.A., Holthuis, J., Markose, E., van Wijnen, A.J., Wolfe, S.A., Grimes, S.R., Lian, J.B., and Stein,

G.S. (1990). Modifications of protein-DNA interactions in the proximal promoter of a cell-growth-

regulated histone gene during onset and progression of osteoblast differentiation. Proc Natl Acad Sci U S

A 87, 5129-5133.

Parker, D.S., Jemison, J., and Cadigan, K.M. (2002). Pygopus, a nuclear PHD-finger protein required for

Wingless signaling in Drosophila. Development 129, 2565-2576.

Pittenger, M.F., Mackay, A.M., Beck, S.C., Jaiswal, R.K., Douglas, R., Mosca, J.D., Moorman, M.A.,

Simonetti, D.W., Craig, S., and Marshak, D.R. (1999). Multilineage potential of adult human

mesenchymal stem cells. Science 284, 143-147.

Pruijt, J.F., Lindley, I.J., Heemskerk, D.P., Willemze, R., and Fibbe, W.E. (1997). Leukemia inhibitory

factor induces in vivo expansion of bone marrow progenitor cells that accelerate hematopoietic

reconstitution but do not enhance radioprotection in lethally irradiated mice. Stem Cells 15, 50-55.

Page 45: 09-10-26 Thesis

45

Semenov, M.V., Tamai, K., Brott, B.K., Kuhl, M., Sokol, S., and He, X. (2001). Head inducer Dickkopf-

1 is a ligand for Wnt coreceptor LRP6. Curr Biol 11, 951-961.

Sutherland, M.S., Rao, L.G., Muzaffar, S.A., Wylie, J.N., Wong, M.M., McBroom, R.J., and Murray,

T.M. (1995). Age-dependent expression of osteoblastic phenotypic markers in normal human osteoblasts

cultured long-term in the presence of dexamethasone. Osteoporos Int 5, 335-343.

Szilvassy, S.J., Weller, K.P., Lin, W., Sharma, A.K., Ho, A.S., Tsukamoto, A., Hoffman, R., Leiby, K.R.,

and Gearing, D.P. (1996). Leukemia inhibitory factor upregulates cytokine expression by a murine

stromal cell line enabling the maintenance of highly enriched competitive repopulating stem cells. Blood

87, 4618-4628.

Till, J.E., and Mc, C.E. (1961). A direct measurement of the radiation sensitivity of normal mouse bone

marrow cells. Radiat Res 14, 213-222.

Tolwinski, N.S., and Wieschaus, E. (2001). Armadillo nuclear import is regulated by cytoplasmic anchor

Axin and nuclear anchor dTCF/Pan. Development 128, 2107-2117.

Towler, D.A., Bennett, C.D., and Rodan, G.A. (1994). Activity of the rat osteocalcin basal promoter in

osteoblastic cells is dependent upon homeodomain and CP1 binding motifs. Mol Endocrinol 8, 614-624.

Tropel, P., Noel, D., Platet, N., Legrand, P., Benabid, A.L., and Berger, F. (2004). Isolation and

characterisation of mesenchymal stem cells from adult mouse bone marrow. Exp Cell Res 295, 395-406.

van den Berg, W.B., Joosten, L.A., Helsen, M., and van de Loo, F.A. (1994). Amelioration of established

murine collagen-induced arthritis with anti-IL-1 treatment. Clin Exp Immunol 95, 237-243.

van Straalen, J.P., Sanders, E., Prummel, M.F., and Sanders, G.T. (1991). Bone-alkaline phosphatase as

indicator of bone formation. Clin Chim Acta 201, 27-33.

Wainwright, B.J., Scambler, P.J., Stanier, P., Watson, E.K., Bell, G., Wicking, C., Estivill, X., Courtney,

M., Boue, A., Pedersen, P.S., et al. (1988). Isolation of a human gene with protein sequence similarity to

human and murine int-1 and the Drosophila segment polarity mutant wingless. EMBO J 7, 1743-1748.

Waltzer, L., and Bienz, M. (1998). Drosophila CBP represses the transcription factor TCF to antagonize

Wingless signalling. Nature 395, 521-525.

Watt, F.M., and Hogan, B.L. (2000). Out of Eden: stem cells and their niches. Science 287, 1427-1430.

Weissman, I.L. (2000). Stem cells: units of development, units of regeneration, and units in evolution.

Cell 100, 157-168.

Whitney, M.J., Lee, A., Ylostalo, J., Zeitouni, S., Tucker, A., and Gregory, C.A. (2009). Leukemia

inhibitory factor secretion is a predictor and indicator of early progenitor status in adult bone marrow

stromal cells. Tissue Eng Part A 15, 33-44.

Yokoya, F., Imamoto, N., Tachibana, T., and Yoneda, Y. (1999). beta-catenin can be transported into the

nucleus in a Ran-unassisted manner. Mol Biol Cell 10, 1119-1131.

Zhang, M., Yan, Y., Lim, Y.B., Tang, D., Xie, R., Chen, A., Tai, P., Harris, S.E., Xing, L., Qin, Y.X., et

al. (2009). BMP-2 modulates beta-catenin signaling through stimulation of Lrp5 expression and

inhibition of beta-TrCP expression in osteoblasts. J Cell Biochem 108, 896-905.

Page 46: 09-10-26 Thesis

46

Figures

Figure 1: The canonical and non-canonical Wnt signalling pathway. In the absence of Wnt

ligands, a β-catenin destructive complex consisting of Axin, APC and GSK 3β forms and

phosphorylation of β-catenin results in its ubiquination. Upon the binding of Wnt ligands to both

the frizzled (FZD) and LRP5/6 receptors, signalling though the canonical pathway is transduced

by β-catenin. The key difference between the canonical and non-canonical pathway is the

requirement of β-catenin for signal transduction. Adapted from Ling, L., Nurcombe, V., and

Cool, S.M. (2009). Wnt signaling controls the fate of mesenchymal stem cells. Gene 433, 1-7.

Page 47: 09-10-26 Thesis

47

Figure 2: Dual-Luciferase Reporter Assay – The optimal TOPFlash firefly luciferase plasmid

to Renilla luciferase plasmid ratio, was measured using the mutant β-catenin overexpressing ∆45

plasmid (0.25μg) to specifically activate the TOPFlash promoter. Cells were treated with 0.1,

0.25 or 0.5μg/well of either TOPFlash or FOPFlash control (not shown). Experiments were

performed in triplicate and the firefly luciferase activity standardised against the Renilla

luciferase activity. The firefly luciferase/Renilla ratio is expressed as fold change, normalised

against samples that had not been transfected with ∆45.

Page 48: 09-10-26 Thesis

48

Figure 3: Dual-Luciferase Reporter Assay – The optimal ∆45 plasmid concentration was

determined by co-transfecting cells with TOPFlash plasmid and 0.05, 0.1 or 0.25μg/well of ∆45

plasmid. Each treatment was performed in triplicate and the firefly luciferase activity

standardised against the Renilla luciferase activity. The firefly luciferase/Renilla ratio is

expressed as fold change with the ratio normalised to control samples.

Page 49: 09-10-26 Thesis

49

Figure 4: Dual Luciferase Reporter Assay – Effect of IL-1β and TNF-α on β-catenin

activity. MC3T3-E1 cells transfected with ∆45, a mutant β-catenin overexpressing plasmid and

treated with 1ng/ml IL-1β or 1ng/ml TNF-α for 48hrs before luciferase activity was measured.

Each treatment was performed in triplicate and the firefly luciferase activity standardised against

the Renilla luciferase activity. The firefly luciferase/Renilla ratio is expressed as fold change with

the ratio normalised to control samples. The cells were treated with the cytokines for 48hrs.

Page 50: 09-10-26 Thesis

50

Figure 5: Effects of LiCl,IL-1β and TNF-α on β-catenin activity. MC3T3-E1 cells were

treated in the presence or absence of LiCl (10mM) and treated with IL-1β or TNF-α for 48hrs

before luciferase activity was measured. A: Experiments performed with less differentiated cells.

B: Experiments performed with more differentiated cells. Each treatment was performed in

triplicate and the firefly luciferase activity standardised against the Renilla luciferase activity.

The firefly luciferase/Renilla ratio is expressed as fold change with the ratio normalised to

control samples. Cells were treated with LiCl, IL-1 and TNF-α for 48 hrs.

Page 51: 09-10-26 Thesis

51

Figure 6: Effects of Wnt3a, IL-1β and TNF-α on β-catenin activity. A: MC3T3-E1 cells were

treated with Wnt3a in the presence or absence of IL-1β and TNF-α for 48hrs before firefly

luciferase activity was measured. B: The optimal concentration of the axin-RFP plasmid (which

inhibits Wnt/β-catenin activity) was determined by transfecting cells with either 0.05, 0.1,

0.25μg of plasmid and treating the cells for 48hrs with Wnt3a before firefly luciferase activity

was measured. C: Cells were treated with Wnt3a, IL-1β, TNF-α and Axin-RFP and firefly

luciferase activity was measured 48hrs later. Wnt3a results in a 60 fold increase in firefly

luciferase activity. Each treatment was performed in triplicate and the firefly luciferase activity

standardised against the Renilla luciferase activity. The firefly luciferase/Renilla ratio is

expressed as fold change with the ratio normalised to control samples.

Page 52: 09-10-26 Thesis

52

Figure 7: β-catenin western blot. Western blot using whole cell lysates from cells that had been

transfected with ∆45 at 0.1, 0.5 and 1.0μg/well. Increasing amounts of the plasmid failed to

induced an increase in the band intensities using anti-β-catenin 1° antibodies. Actin was used as

a loading control.

Page 53: 09-10-26 Thesis

53

Figure 8: β-catenin nuclear extract western blot. Western blot of nuclear extracts from cells

that had been stimulated with LiCl (10mM) (A) and Wnt3a (2ng/ml) (B) at various time points –

1,2,4,6, and 24hrs, to determine the best time point to obtain maximal change in nuclear β-

catenin levels.

Page 54: 09-10-26 Thesis

54

Figure 9: Effect of cytokines on β-catenin. Western blot of cytoplasmic and nuclear extracts

from cells stimulated for 2 or 6 hours with either LiCl or Wnt3a and IL-1β or TNF-α. β-tubulin

and histone H3 were used as loading controls for the cytoplasmic and nuclear extracts

respectively.

Page 55: 09-10-26 Thesis

55

Figure 10: Osteoblast differentiation assay staining for ALP. Cells were transfected with ∆45

(0.02μg/well) prior to treatment with LiCl and IL-1β or TNF-α. Cells were also treated with LiCl

and either IL-1β or TNF-α. Cells were treated for 5 days in osteogenic media.

Page 56: 09-10-26 Thesis

56

Figure 11: Osteoblast differentiation assay staining for ALP expression. Wnt3a seemed to

inhibit osteoblast inhibition. Determining the effect of IL-1β and TNF-α in the absence and

presence of Wnt3a on osteoblast differentiation. Cells were treated for 5 days osteogenic media

with IL-1β and TNF-α in the presence of Wnt3a.

Page 57: 09-10-26 Thesis

57

Figure 12: Real time-PCR using cDNA from cells that had been treated in osteogenic media for

3 days with LiCl, Wnt3a, IL-1β and TNF-α. Results are expressed as fold change and normalised

to TBP expression. The results are the average of 3 separate experiments performed in triplicate.

Page 58: 09-10-26 Thesis

58

Figure 13: RT-PCR using cDNA from cells that been treated in osteogenic media for 7 days

with LiCl, Wnt3a, IL-1β and TNF-α. Results are expressed as fold change and normalised to

TBP expression. The results are the average of 3 separate experiments performed in triplicate.