cdh11 acts as a tumor suppressor in a murine ... · ii cdh11 acts as a tumor suppressor in murine...

101
Cdh11 Acts as a Tumor Suppressor in a Murine Retinoblastoma Model by Facilitating Tumor Cell Death by Christine Laura Yurkowski A thesis submitted in conformity with the requirements for the degree of Master of Science Graduate Department of Molecular Genetics University of Toronto © Copyright by Christine Laura Yurkowski 2010

Upload: trinhkhue

Post on 19-Aug-2019

223 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

Cdh11 Acts as a Tumor Suppressor in a Murine Retinoblastoma Model by Facilitating Tumor Cell Death

by

Christine Laura Yurkowski

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

Graduate Department of Molecular Genetics University of Toronto

© Copyright by Christine Laura Yurkowski 2010

Page 2: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

ii

Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma

Model by Facilitating Tumor Cell Death

Christine Yurkowski

Master of Science

Molecular Genetics

University of Toronto

2010

Abstract

Retinoblastoma, a rare childhood cancer of the retina, is characterized by loss of both alleles of

the RB1 gene. However, additional mutational events are required for malignancy. CGH studies

described common chromosomal changes indicating potential oncogenes and tumor suppressor

genes. Previous work in the lab implicated Cadherin-11 (Cdh11) as a tumor suppressor after

analysis in human retinoblastomas and in the simian virus 40 large T-antigen induced murine

retinoblastoma model (TAg-RB) showed loss of Cdh11 expression. TAg-RB mice crossed with

Cdh11-/-

mice, revealed faster growing tumors in mice null for Cdh11. This thesis focused on

defining the tumor suppressor role of Cdh11 in retinoblastoma progression. The results showed

in vitro and in vivo evidence that Cdh11 was promoting apoptosis and not suppressing

proliferation. We also observed an increase in invasion markers upon the loss of Cdh11. We

conclude that Cdh11 acts as a tumor suppressor in retinoblastoma, through promotion of

apoptosis.

Page 3: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

iii

Acknowledgments

I would like to thank my supervisor, Dr. Brenda Gallie, for giving me the opportunity to

join her lab and be part of her amazing research team, which has vastly contributed to the study

of retinoblastoma. Her passion for science, medicine, life and helping others is truly

inspirational and she has served as such an incredible mentor. I admire her for her dedication

towards everything she does, her many accomplishments and her constant support and desire to

help everyone around her succeed. She is a big reason why I was able to take so much away

from my time in the lab and why I will always remember my experiences so fondly.

I would also like to thank my committee members for providing me with necessary

guidance and support along the way. It was greatly appreciated and will always be remembered.

Being around positive working environments is wondrous for your motivation and your

desire to want to come to work every day. I can honestly say the environment in the lab was one

of the most positive and supportive atmospheres I‟ve ever known. Everyone in the lab is kind,

generous, and knowledgeable. I would love to dearly thank all past and present members of the

lab (Ying Guo, Tim To, Helen Dimaras, Mellone Marchong, Ghada Kurban, Brigitte Theriault,

Stephanie Yee, Clarellen Spencer, Dr. Sanja Pajovic and Lucy Fuccillo). I wish you all the

absolute best in life. On a more personal note, I would like to thank Dr. Sanja Pajovic for her

constant guidance and support. You are an integral part of the lab, and it would absolutely not be

the same without you around, as you are always the greatest help to everyone and always have

something to say to put a smile on our faces. Clarellen Spencer also played an integral part in

my time at the lab, helping me with various protocols and especially in handling the mice. Your

patience is truly a virtue and your amazing stories of your travels and your generous treats are

always a delight. Brigitte Theriault and Ghada Kurban are such knowledgeable people and were

always of tremendous help if I ever needed anything. I will always treasure our morning

conversations in the office to get our days started on the right foot, your laughter and our many

many lunches to our favourite destinations. Lucy Fuccillo constantly manages to keep everyone

on task and organized on a daily basis. Not only does it feel you are a constant life saver, but in

addition to that, are one of the most positive people I know. Your pure zest for life and

involvement in so many wonderful activities is truly admirable. Lastly, to Stephanie Yee, who

was not only alongside for the ride as my peer, but became an amazing and dear friend. I feel

Page 4: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

iv

my experience was that much more special because we got to share everything from the process

of studying for a Master‟s Degree, to personal highs and lows, daily activities, interests, dreams,

concerns, laughs and life. I will never forget our endless conversations and will always deeply

value your friendship.

Finally I‟d like to thank my family and friends who are always there for me and are

constantly sources of love, support and joy. My friends are extended members of my family, and

since my parents are in Winnipeg, I treasure their loyalty, endless love and generosity that much

more. To my parents, Katherine and Malcolm Yurkowski, who have always supported me in

anything I did in life, I have learnt so much from you and continue to do so every day. Your

generosity is the reason I am in Toronto today and the reason I am able to continue following my

dreams. Although you are not in the same city as me, I am lucky enough to be able to be a short

plane ride away and visit you as much as I need to. I am blessed to have such caring parents who

always want the best for me, and you are and always will be the biggest part of my success.

Page 5: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

v

Table of Contents

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

Table of Contents .......................................................................................................................... v

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

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

List of Abbreviations .................................................................................................................... x

List of Appendices ...................................................................................................................... xiii

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

1.1 Retinoblastoma – The Disease ......................................................................................... 1

1.2 Current Treatment of Retinoblastoma ........................................................................... 2

1.3 Retinoblastoma Gene and Protein (pRB) ....................................................................... 3

1.4 Mouse Studies .................................................................................................................... 4

1.4.1 RB1-/- Mouse Models ............................................................................................ 4

1.4.2 TAg-RB Model Of Retinoblastoma ....................................................................... 5

1.5 Retinal Development ......................................................................................................... 6

1.6 Retinoma – The Benign Precursor .................................................................................. 7

1.7 Genomic Changes Driving Progression to Retinoblastoma .......................................... 8

1.7.1 Gain at 1q ............................................................................................................... 9

1.7.2 Gain at 6p ............................................................................................................. 10

1.7.3 Gain at 2p ............................................................................................................. 11

1.7.4 Loss at 16q ............................................................................................................ 11

1.8 Cadherin Biology ............................................................................................................ 13

1.8.1 Cadherin Structure .............................................................................................. 13

1.8.2 Cadherin Regulation and Expression ................................................................. 15

1.8.3 Cadherins in the Mammalian Retina .................................................................. 18

Page 6: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

vi

1.8.4 Cadherin-11 and its Isoforms .............................................................................. 20

1.8.5 Cadherin-11: Implicated Roles ............................................................................ 21

1.8.6 Cadherins in Cancer Progression ....................................................................... 23

1.9 Cadherin-11 as a Tumor Suppressor in Retinoblastoma Progression ....................... 26

1.10 Apoptosis .......................................................................................................................... 27

1.10.1 Extrinsic Pathway ................................................................................................ 28

1.10.2 Intrinsic Pathway ................................................................................................. 29

1.11 Project Aims and Hypothesis ......................................................................................... 30

1.11.1 To examine if Cadherin-11 is promoting apoptosis in TAg-RB tumors and

define the acting apoptotic pathway ..................................................................... 30

1.11.2 To determine if the loss of Cadherin-11 expression leads to invasive and

aggressive tumors through expression of invasion markers ................................. 30

Chapter 2 ..................................................................................................................................... 31

2.1 Mouse Models .................................................................................................................. 31

2.2 Genotyping ....................................................................................................................... 31

2.3 Histology and Slide Selection .......................................................................................... 32

2.4 Immunohistochemistry .................................................................................................... 33

2.5 BrdU Incorporation ......................................................................................................... 36

2.6 Image Analysis .................................................................................................................. 36

2.7 Statistical Analysis ........................................................................................................... 37

2.8 Tissue Culture and Cell Lines ......................................................................................... 37

2.9 RNA Isolation ................................................................................................................... 38

2.10 Protein Isolation ............................................................................................................. 39

2.11 Stealth RNAi ................................................................................................................... 40

Chapter 3 ..................................................................................................................................... 41

3.1 Cadherin-11 acts a tumor suppressor in TAg-RB tumors through promotion of

apoptosis ........................................................................................................................... 41

Page 7: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

vii

3.1.1 Active Apoptotic Pathway in TAg-RB Mice ........................................................ 41

3.1.2 Quantification of Apoptotic Protein Expression in TAg-RB Tumors................ 43

3.1.3 Analysis of Cadherin-11 in TAg-RB Derived Cell Lines.................................... 45

3.1.4 Apoptotic Protein Expression in a TAg-RB Derived Cell Line .......................... 46

3.1.5 Cdh11 and Proliferation in TAg-RB Tumors .................................................... 48

3.2 Loss of Cdh11 in TAg-RB Tumors Increases Invasion Potential ................................ 51

3.2.1 Knockdown of Cdh11 in a TAg-RB Cell Line Leads to an Increase of EMT

Markers and Metalloproteases ............................................................................ 51

3.2.2 β-catenin mRNA and Protein Levels Increase Upon Knockdown of Cdh11 .... 53

Chapter 4 ..................................................................................................................................... 55

4.1 Cdh11 as a Tumor Suppressor in Retinoblastoma Progression .................................. 55

4.2 Cdh11 Acts as a Tumor Suppressor by Promoting Apoptosis ..................................... 56

4.3 Loss of Cdh11 and Invasion ............................................................................................. 59

4.4 Cdh11 in Cancer Progression .......................................................................................... 61

4.5 Future Directions ............................................................................................................. 63

4.5.1 The Mechanism of Influence of Cdh11 on Apoptosis and Invasion Potential . 63

4.5.2 Mechanism of Cdh11 Downregulation in Retinoblastoma ................................ 65

4.6 Summary and Significance .............................................................................................. 66

References .................................................................................................................................... 68

Page 8: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

viii

List of Tables

Table 1 Antibodies. ....................................................................................................................... 35

Table 2 RT-PCR Primers. ............................................................................................................. 39

Page 9: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

ix

List of Figures

Figure 1.1 Retinoblastoma Genetic: Multistep Model. ................................................................... 8

Figure 1.2 Chromosome 16: Positioning of CDH11 and Cadherin Cluster. ................................ 13

Figure 1.3 Classical Cadherin Structure and Regulation. ............................................................. 18

Figure 1.4 Cadherin-11 and its Isoforms. ..................................................................................... 21

Figure 1.5 Cadherin-11 as a Tumor Suppressor in Retinoblastoma Progression. ........................ 27

Figure 1.6 Apoptotic Pathways. .................................................................................................... 30

Figure 3..1 Expression of Apoptotic Proteins within TAg-RB Tumors. ...................................... 42

Figure 3.2 Acting Apoptotic Pathway in Retinal Tumors of TAg-RB Mice. ............................... 44

Figure 3.3 Differences in Apoptotic Protein Expression in TAg-RB Tumors at PND84. ............ 45

Figure 3.4 Cdh11 Protein Expression in TAg-RB Derived Cell Lines. ........................................ 46

Figure 3.5 Cdh11 Knockdown Leads to Decreased Caspase-3 Expression. ................................ 47

Figure 3.6 Quantifying the Percentage of BrdU Proliferating Cells as a Function of Tumor Area.

....................................................................................................................................................... 49

Figure 3.7 Average Area of Proliferation Cells Per Tumor Area at PND84. ............................... 50

Figure 3.8 Knockdown of Cdh11 Leads to Increased mRNA Expression of EMT and Invasion

Markers. ........................................................................................................................................ 52

Figure 3.9 Cdh11 Knockdown Leads to Upregulation of ϐ-catenin mRNA and Protein

Expression Levels. ........................................................................................................................ 54

Page 10: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

x

List of Abbreviations

ADAM A Disintegrin and Metallopeptidase

AJ Adherens Junction

AP Alkaline Phosphatase

APAF1 Apoptotic Peptidase Activating Factor 1

APC Adenomatosis Polyposis Coli

BAX Bcl-2-associated X Protein

BH3 Bcl-2 Homology Domain 3

BrdU Bromodeoxyuridine

BSA Bovine Serum Albumin

Cadherin-11 (i) Cadherin-11 intact isoform

Cadherin-11 (s) Cadherin-11 secreted isoform

Cadherin-11 (v) Cadherin-11 variant isoform

CAM Cadherin Cell Adhesion Complex

CDH Cadherin

CDK Cyclin Dependent Kinase

CGH Comparative Genomic Hybridization

CNS Central Nervous System

CP Cytoplasmic Domain

DAB Diaminobenzidine

DAPI 4‟,6-diamidino-2-phenylindole

DEK DEK oncogene (DNA binding)

dNTP Deoxynucleoside Triphosphate

E2F E2F Transcription Factor

EBR External Beam Therapy

Page 11: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

xi

EC Extracellular Domain

ECL Enhanced Chemiluminescence

ED Embryonic Day

EMT Epithelial to Mesenchymal Transition

F-W Flexner Wintersteiner Rosette

FADD Fas-associated Death Domain

FGFR Fibroblast Growth Factor Receptor

FLIP Caspase-8 (FLICE)-like Inhibiting Protein

GCL Ganglion Cell Layer

GSK-3ϐ Glycogen Synthase Kinase-3ϐ

H-W Homer Wright Rosette

H&E Hematoxylin and Eosin

HAV Histidine Alanine Valine

HRP Horseradish Peroxidase

INL Inner Nuclear Layer

IP Intra-Peritoneal

KIF Kinesin Family Member

LEF Lymphocyte Enhancer Binding Factor

L-Hϐ Luteinizing Hormone Beta

LOH Loss of Heterozygosity

M1, 2, 3…, n Mutational Event 1, 2, 3…, n

MAPK Mitogen-activated Protein Kinases

MMP Matrix Metalloproteinase

MYCN v-myc Myelocytomatosis Viral Related Oncogene, Neuroblastoma

Derived

ONL Outer Nuclear Layer

Page 12: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

xii

PCNA Proliferating Cell Nuclear Antigen

PFA Paraformaldehyde

PI3K Phosphoinositide 3-kinases

PND Post Natal Day

QAV Glutamine Alanine Valine

QM-PCR Quantitative Multiplex PCR

RB Retinoblastoma

RTK Receptor Tyrosine Kinase

RT-PCR Reverse Transcriptase PCR

SCID Severe Combined Immunodeficiency

SDS-PAGE Sodium Dodecyl Sulfate Polyacrylamide Gel Electrophoresis

TAg SV40 Large T-antigen

TAg-RB SV40 Large T-antigen Induced Murine Retinoblastoma Model

TBP TATA Box Binding Protein

TBS Tris Buffered Saline pH 8.0

TBST Tris Buffered Saline pH 8.0/ Tween-20

TCF T-cell Factor

TGFϐ Transforming Growth Factor Beta

TM Transmembrane Domain

TNE Tris-Sodium-EDTA Buffer

TNF Tumor Necrosis Factor

TRAIL TNF-related Apoptosis-inducing Ligand Receptor

Wnt Wingless Type

Page 13: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

xiii

List of Appendices

Parts of this thesis were submitted for publication and are currently in press in PLOs Genetics as

follows: Cdh11 acts as a tumor suppressor in a murine retinoblastoma model by facilitating

tumor cell death. Mellone N. Marchong, Christine Yurkowski, Clement Ma, Clarellen Spencer,

and Brenda L. Gallie.

Page 14: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

1

Chapter 1

Introduction

1.1 Retinoblastoma – The Disease

Retinoblastoma is the most common childhood intraocular malignancy that occurs in

approximately 1 in 20,000 births (Balmer et al., 2006). The disease is initiated by two

mutational events in the developing retina, resulting in the loss of both alleles of the RB1 gene.

The disease exists in two forms: hereditary and non-hereditary. Hereditary cases are

characterized by an inherited mutation in the RB1 gene that is present in all cells in the body. A

second spontaneous mutation in the retina results in the loss of the remaining normal allele.

These patients often develop multiple tumors in both eyes (bilateral retinoblastoma), and

furthermore leave the patient more susceptible to secondary malignancies later in life

(Abramson, 1999; Marees et al., 2008). The non-heritable form arises by two sporadic

mutational events occurring within the same cell, and results in unilateral retinoblastoma,

affecting one eye. The observation that children with bilateral retinoblastoma had an earlier age

of diagnosis compared to children with unilateral retinoblastoma was the basis of Knudson‟s

“two-hit” hypothesis (Knudson, 1971). This concept was expanded to suggest the two „hits‟

consisted of inactivation of the predisposing gene that would normally suppress retinoblastoma

Page 15: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

2

(Comings, 1973), now a general idea of the genetic basis of cancer, in which at least two

mutations are necessary for cancer initiation.

1.2 Current Treatment of Retinoblastoma

Retinoblastoma is readily curable today in the developed world, including good prognosis of

visual function. Treatment strategies usually involve a multidisciplinary approach and depend

on constant re-evaluation with survival and salvage of visual function as the primary aim

(Balmer et al., 2006). Most patients presenting with unilateral retinoblastoma are advanced

cases and are usually cured (>96%) by enucleation (Chintagumpala et al., 2007). In children

with bilateral retinoblastoma, treatment often entails multimodality therapy (Chintagumpala et

al., 2007). Often, the eye and sometimes vision can be saved. Therapies include brachytherapy,

thermotherapy, laser photocoagulation, cryotherapy, chemotherapy and external beam therapy

(EBR) (Balmer et al., 2006). The choice between these therapies lies within the size and severity

of the tumor. Most often, chemotherapy will be used in order to shrink the tumor so local

therapies can be used. Primary chemotherapy can shrink the tumor volume by over 50% (Chan

et al., 2005; Shields et al., 1997). This then allows local therapies such as cryotherapy and laser

therapy to eradicate the remaining disease (Chan et al., 2005; Chintagumpala et al., 2007).

EBR is often used as a last resort. It can be effective in curing retinoblastoma, but is proven to

have major long-term side effects, with a reported increase in second cancers in patients

compared to those who do not receive EBR (Abramson and Frank, 1998). In cases of

extraocular disease, patients often have a very poor prognosis, however few patients may benefit

from a combination of EBR and chemotherapy (Chantada et al., 2005). Current therapies

contribute significantly to vision salvage and overall cure rates, however they are not without

significant morbidities. Therefore, emphasis has recently been placed on developing therapies

with fewer long term morbidities, which requires a better understanding of the pathogenesis of

Page 16: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

3

retinoblastoma. In an attempt to do so, the use of preclinical models has been important

(Chintagumpala et al., 2007; Hurwitz et al., 1999).

1.3 Retinoblastoma Gene and Protein (pRB)

RB1 was the first tumor suppressor gene to be discovered (Cavenee et al., 1983; Godbout et al.,

1983). It was mapped to chromosomal region 13q14 (Sparkes et al., 1980), and was first cloned

a few years after (Dryja et al., 1986; Friend et al., 1986; Fung et al., 1987; Lee et al., 1987).

RB1 encodes the retinoblastoma protein pRB, belonging to the „pocket protein‟ family. This

family of proteins, including p107 and p130, all share similar and redundant functions in

regulation of the cell cycle, modulating members of the E2F family of transcription factors,

which regulate the expression of genes required for cell proliferation (Cao et al., 1992;

Chellappan et al., 1991; Cobrinik, 2005). The pRB protein has three distinct domains; the N-

terminus, R motif and A/B „pocket‟. The „pocket‟ is necessary for biological function, including

transcriptional regulation and interaction with viral and cellular proteins (DiCiommo et al., 2000;

Kouzarides, 1995). Specifically, pRB interacts with E2F transcription factors to inhibit their

activity. This negatively regulates the cell cycle, halting it in G1 phase. This occurs with pRB in

a hypophosphorylated state. Upon phosphorylation by cyclin dependent kinases (CDK) and

cyclin complexes, pRB releases the E2Fs, allowing transcription of their targets to commence,

subsequently allowing the re-entry to the cell cycle and promoting cell division (Mittnacht,

1998).

Although all three family members show similar functions, only pRB has proven to have tumor

suppressor abilities in human tumors (Lipinski and Jacks, 1999). Mutations in both RB alleles

initiate disease in a variety of cancers, including retinoblastoma, an increase of incidence of

osteosarcoma developing in children and teenagers, small lung carcinomas, and in cancers of the

Page 17: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

4

breast, prostate and bladder (Bookstein and Lee, 1991; Deshpande and Hinds, 2006; Harbour et

al., 1988). Additionally, pRB is commonly inactivated in many other types of tumors, through

mutation, viral oncoprotein or by alteration of other proteins involved in the same pathway.

Certain viruses, such as simian virus 40 large T antigen (TAg), human papilloma virus , E7

(HPV), and adenovirus (E1a), consist of an LxCxE motif, required for viral transformation,

allowing them to bind the pRB and „pocket‟ family members at the A/B pocket domain,

specifically the repressor motif (Dyson et al., 1989; Felsani et al., 2006). Inactivation of pRB,

therefore, leads to abnormal division of cells and continual proliferation due to the inability to

exit the cell cycle.

1.4 Mouse Studies

1.4.1 RB1-/-

Mouse Models

Generating mouse models to study retinoblastoma development has been extremely difficult.

Firstly, spontaneous retinoblastoma seems to be almost exclusively a human disease, seen very

rarely in other species. Mice with a RB+/-

genotype show no retinal phenotype, however develop

both pituary tumors and thyroid carcinomas (Jacks et al., 1992). Secondly, genetic RB-/-

knockout proved to be lethal in early embryonic stages, around ED14.5, initially thought to be

due to defects in neural and erythroid systems and massive apoptosis (Clarke et al., 1992; Jacks

et al., 1992; Lee et al., 1992). Instead, this was later found to be due to hypoxia because of

placental defects, as when RB-/-

embryos were studied with wildtype placental, they survived till

birth, but later died due to respiratory problems (de Bruin et al., 2003; Wu et al., 2003). Studies

then focused on producing chimeric mice, but these mice only showed pituitary tumor

development, and no retinoblastoma phenotype (Maandag et al., 1994). A breakthrough came

when these chimeric mice were generated by knocking out both RB1 and one of its family

members, p107 or p130 ((Dannenberg et al., 2004; Robanus-Maandag et al., 1998). The need

Page 18: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

5

for RB and at least one of its family members to be lost for retinoblastoma development in mice

was confirmed by the use of conditional knockout models (CKO), generating heritable mouse

models of retinoblastoma ((Chen et al., 2004; MacPherson et al., 2004; Zhang et al., 2004).

1.4.2 TAg-RB Model Of Retinoblastoma

Transgenic mouse models provided the earliest heritable models of retinoblastoma, involving the

expression of viral oncoproteins. Extensively used for study of retinoblastoma is the simian

virus 40 (SV40) large T-antigen induced model of retinoblastoma (TAg-RB). This model arose

fortuitously, as it was originally intended for the study of pituitary adenoma. One founder

presented with bilateral retinoblastoma tumors, from high expression levels of TAg within the

retina (Windle et al., 1990). The transgene is under control of the luteinizing hormone-beta (LH-

beta) promoter, and integration into an unknown genomic control element at chromosomal

location 4p directing transgene expression to cells in the retina, where LH-beta is not normally

expressed. This model gave rise to hereditary, multifocal retinoblastoma presenting from 1

month of age. These tumors resemble human retinoblastomas (Windle et al., 1990), and

originate from the inner nuclear layer (Pajovic et al, unpublished data). Ultrastructurally, these

tumor cells present as microtubules in the characteristic “9+0” arrangement, also seen in normal

photoreceptor cells, a distinguishing characteristic of human retinoblastomas. Histologically,

they contain two multicellular structures common to human retinoblastomas. Flexner-

Wintersteiner rosettes (F-W) consist of a single layer of cuboidal cells surrounding a central

lumen. They resemble primitive photoreceptors and are unique to retinoblastoma. Homer-

Wright rosettes (H-W) are single-layer rows of tumor cells that surround a central area filled

with neurofibrils (Mills et al., 1999; Ts'o et al., 1969; Windle et al., 1990; Wippold and Perry,

2006). These are also highly characteristic of neuroblastoma and medulloblastoma. The TAg-

RB model not only presents histologically similar tumors to human retinoblastoma, but also

Page 19: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

6

proves to have similar expression changes associated with tumor development, presented in

human retinoblastoma as gain and overexpression of KIF14, E2F3 and DEK (Corson et al.,

2005; Orlic et al., 2006), as well as loss of p75NTR

expression (Dimaras et al., 2006) and genomic

and expression loss of CDH11 (Marchong et al., 2004). The TAg-RB model is the most used

for pre-clinical and therapeutic studies relevant to human retinoblastoma. Transgenic models are

important as they allow therapeutic manipulation not otherwise possible. Studies have included

use of a variety of chemotherapeutic agents, radiation and use of attenuated viruses to treat

tumors (Kang et al., 2009; Mills et al., 1999; Sobrin et al., 2004; Suarez et al., 2007; Tsui et al.,

2008). Although it is similar to human disease, the TAg-RB model presents some limitations in

the study of human retinoblastomas. As the virus binds to pRB family member, p53 and other

proteins, it is possible other pathways are affected in this model not affected in the human form

of the disease. However, interspecies differences in the mechanisms of tumorigenesis, provides

general limits to uses of all mouse models (Dyer and Bremner, 2005; Mills et al., 1999).

1.5 Retinal Development

The retina is a sheet of neural tissue that takes in input signals from light and sends this

information to the brain to determine what we are seeing. It is first formed by an invagination of

the neural tube, marked by an appearance of an optic pit. This leads to the formation of the optic

vesicles. Next, a lens placode is formed, a thickening of the surface ectoderm, which is in

contact with the optic vesicle. Invagination of these two structures together leads to the lens and

a two layered optic cup, the inner layer forming the neural retina [reviewed in(Chow and Lang,

2001; Young, 1985a; Young, 1985b). This close contact is necessary for neural retinal

development and studies have shown fibroblast growth factor (FGF) plays a role in signaling

neural retinal development during this time period (Guillemot and Cepko, 1992).

Page 20: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

7

The retina contains seven main different types of cells. The earliest step in development is

production of retinal cells by the „progenitors‟ of the neurepithelial layer of the optic cup. These

cells migrate to form one of three layers after they have undergone the last mitosis. These cells

start to form synapses with eachother, upon which proper processing is crucial. Patterning and

proper ratios of each cell type are dependent on these early processes and is necessary for proper

retinal and visual development (Cepko et al., 1996; Young, 1985a). The three major layers of

the developed retina are the outer nuclear layer (ONL), made up of rod and cone photoreceptors,

the inner nuclear layer (INL), consisting of horizontal, amacrine, bipolar and Muller glia cells

and the ganglion cell layer (GCL), including ganglion cells and amacrine cells. The ONL uses

light as its stimulus and translates the information through synapses of the INL to the GCL,

which transmits the results to various targets of the brain. Just like in other structures of the

brain, glial cells provide retinal architecture and different external cues for retinal development

and apoptosis.

1.6 Retinoma – The Benign Precursor

Studies of retinoblastoma indicated early on that additional genomic aberrations after the loss of

RB1 exist in all retinoblastomas (Squire et al., 1985) suggesting further mutational events were

needed for full development of the disease. In earlier years, inactive tumors resembling

retinoblastoma seen on rare occasions were referred to as „spontaneous regression of

retinoblastoma‟. Without much evidence to support this definition, the term „retinoma‟ was

proposed to describe non-progressive retinal lesions that lacked malignant characteristics (Gallie

et al., 1982). Following, many case studies have supported the idea that retinomas are benign

precursors to malignant retinoblastoma (Balmer et al., 1991; Eagle et al., 1989; Singh et al.,

2000). This idea is supported by studies in mouse models and tumor progression models which

explore the idea that cancer is multistep process (Redston, 2003; Vogelstein and Kinzler, 1993).

Page 21: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

8

Much evidence for a multistep process in retinoblastoma has accumulated. Alone, the loss of

RB1 (M1 and M2) is not sufficient for malignant transformation. Further genomic changes are

needed to transform the benign retinoma into malignant retinoblastoma (M3-Mn) (Dimaras et al.,

2008) (Figure 1.1). This study showed that after the loss of RB1, lesions remained

nonproliferative and also expressed senescence markers, not seen in retinoblastomas. This and

other studies have focused on defining genomic changes that drive progression of the disease to

malignant retinoblastoma.

Figure 1.1 Retinoblastoma Genetic: Multistep Model.

Retinoblastoma is initiated by the biallelic loss of the RB1 gene in a pre-mature retinal cell, mutational events M1

and M2. The loss of RB1 rendering cells RB1-/- is sufficient for retinoma, the benign form of retinoblastoma.

However, additional mutations, termed M3-Mn events are necessary to drive progression to malignancy,

retinoblastoma.

1.7 Genomic Changes Driving Progression to Retinoblastoma

Early studies to determine genomic changes in retinoblastoma focused on revealing recurrent

chromosomal abnormalities. Karyotyping studies defined consistent patterns of genomic change

in retinoblastoma samples. These studies reported most frequent gains at 6p, 1q and loss of

chromosome 16 (Benedict et al., 1983; Chaum et al., 1984; Kusnetsova et al., 1982; Pogosianz

and Kuznetsova, 1986; Squire et al., 1985; Workman and Soukup, 1984). Comparative genomic

hybridisation (CGH) technology followed 15 years later, allowing for higher resolution of

Page 22: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

9

chromosomal aberrations. Five major CGH studies and one matrix study studied a total of 179

tumor samples to find common regions of gains and losses [reviewed in (Corson and Gallie,

2007)]. The results confirmed earlier observations and defined additional changes occurring at

lower frequency. The most common changes included gains at 1q (53%), 6p (54%), 2p (34%),

13q (16%) as well as losses at 13q (12%) and 16q (32%) (Chen et al., 2001; Herzog et al., 2001;

Lillington et al., 2003; Mairal et al., 2000; van der Wal et al., 2003; Zielinski et al., 2005).

These chromosomal regions of gain and loss suggested loci of tumor suppressor and oncogenes

and were presumed to contain target genes that contribute to the overall progression of

retinoblastoma. Several candidate genes have arisen from work in recent years, and defining

their role in retinoblastoma is essential in understanding the pathway of tumorigenesis. This is

necessary to develop therapeutics that can ultimately be used to halt retinoblastoma progression

at an early stage.

1.7.1 Gain at 1q

Genomic gain of chromosome 1q was first seen in retinoblastoma as rearrangements leading to

trisomy of 1q25-1q32 (Gardner et al., 1982). The chromosomal region of 1q31-1q32 is gained in

a variety of cancers (Baudis and Cleary, 2001; Mertens et al., 1994) and in 53% of

retinoblastoma tumors (Corson and Gallie, 2007). This strongly suggested a candidate oncogene

at this locus and QM-PCR revealed candidate genes. Only KIF14 was overexpressed in various

cancers and identified as the target gene with 20/22 retinoblastomas showing mRNA expression

levels of 100-1000 fold increase from normal retina (Corson et al., 2005). KIF14, a member of

the kinesin family, is required for efficient cytokinesis and RNAi mediated knockdown results in

multinucleated cells and acute apoptosis (Carleton et al., 2006; Gruneberg et al., 2006). KIF14

is also overexpressed in breast cancer primary tumors and cell lines, primary lung tumors and

medullablastoma cell lines (Corson et al., 2005). Further studies on KIF14 revealed that mRNA

Page 23: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

10

expression was prognostic for patient outcome in breast and lung cancer (Corson and Gallie,

2006; Corson and Gallie, 2007). Additionally, when KIF14 was knocked down with siRNA in a

lung carcinoma cell line H1299, decreased proliferation and colony formation on soft agar was

observed (Corson et al., 2007). Recent experiments in both retinoblastoma and ovarian cancer

cell lines show similar results (Theriault, unpublished data). This gives strong evidence for

KIF14 as a prominent oncogene in multiple cancers and a potential therapeutic target.

1.7.2 Gain at 6p

Gains in regions 6p were consistently shown in retinoblastomas (Corson and Gallie, 2007;

Potluri et al., 1986; Squire et al., 1985). Recent studies looked to define the minimal region of

gain and two genes were implicated as targets, DEK and E2F3 (Grasemann et al., 2005; Orlic et

al., 2006). E2F3 is a family member of the E2F transcription factors, and has a crucial role as a

cell cycle promoter in proliferating cells (Leone et al., 1998). It was first implicated in Wilms‟

tumor (Baudry et al., 2002) and its overexpression in bladder cancer correlates to histological

grade (Hurst et al., 2008; Oeggerli et al., 2006). DEK is an abundant nuclear phosphoprotein that

shares structural similarity to E2F family members and has multiple roles in chromatin

remodeling, mRNA splicing and transcriptional regulation (Kappes et al., 2001; Waldmann et

al., 2004). It is overexpressed in numerous cancers including bladder, colon, cervical

carcinomas, melanomas and highly defined in myeloid leukemia, where it is translocated to

NUP214 (CAN) (6;9 translocation), forming a DEK-CAN fusion protein ((Carro et al., 2006;

von Lindern et al., 1992; Wu et al., 2008). In retinoblastoma, DEK and E2F3 have been

implicated to have separate roles, E2F3 in growth, and DEK in both growth and survival (Orlic

et al, submitted).

Page 24: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

11

1.7.3 Gain at 2p

Karyotype studies consistently identified chromosomal abnormalities of gain and amplification

at chromosome 2p in retinoblastoma (Squire et al., 1985). Later CGH studies confirmed a

minimal region of gain at 2p24 in 34% of retinoblastoma (Corson and Gallie, 2007) and QM-

PCR revealed MYCN amplification in 3% of tumors and 29% of retinoblastoma cell lines

(Bowles et al., 2007). Earlier studies also reported low-level and high-level MYCN

amplification in retinoblastoma (Lee et al., 1984; Squire et al., 1986). Interestingly, a recent

transgenic mouse study revealed that murine metastatic tumors exhibit high levels of MYCN

amplification (MacPherson et al., 2007). MYCN is a nuclear protein that regulates genes

involved in proliferation and apoptosis (Cavalieri and Goldfarb, 1988; Ramsay et al., 1986).

High amplification in retinoblastoma indicates MYCN expression may be favourable for cell

growth and proliferation. MYCN is commonly amplified in tumors of neuroectodermal origin

including small lung carcinoma and most commonly in neuroblastoma (25-30%), where

amplification has prognostic significance (Brodeur, 2003; Chan et al., 1997). Recent studies

have identified a small subset of tumors in which no RB1 mutation can be found (nmf) (Richter

et al., 2003). Recent work in our lab showed that 56% of these tumors have abnormally high

frequency of MYCN amplification (Yee, unpublished data). Furthermore, these tumors do not

show typical genomic changes of retinoblastoma, including gain at 1q and 6p, or loss of 16q.

This leads to an interesting question of whether MYCN could cause retinoblastoma in patients

with no RB1 mutations.

1.7.4 Loss at 16q

Loss of chromosome 16 was seen in early karyotype studies of retinoblastoma (Potluri et al.,

1986; Squire et al., 1985). Six CGH studies later revealed that 32% (58/179) of tumors showed

loss of either the entire chromosome 16 or 16q, with a minimal region of loss at 16q22 (all but 7

Page 25: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

12

tumors) (Corson and Gallie, 2007). Analysis by loss of heterozygosity (LOH) and QM-PCR

further minimized the region of loss to an area of 2.62Mb, including two sequence tagged sites

(STS) at 16q22.1 (lost in 54% of 71 tumors) and 16q23.3 (lost in 39% of 28) tumors, within

CDH11 and CDH13, respectively (Marchong et al., 2004). CDH13 was ruled out as a candidate

gene as it showed expression levels comparable to healthy human retina in all retinoblastoma

primary tumors and cell lines. Cadherin-11 (Cdh11) expression, however, was lost or decreased

in 91% of tumors and cell lines, compared to adult healthy retina. In addition, RT-PCR revealed

that Cdh11 mRNA expression was lost in 38% of TAg-RB mouse model primary tumors

(Marchong et al., 2004). Supporting these observations, further studies showed decreased Cdh11

expression in retinoblastoma samples, but levels of expression in adjacent retinoma were

equivalent to normal retina (Dimaras et al., 2008).

Cdh11 is a classical cadherin family member that is involved in cell-cell adhesion. It is also

known as OB-cadherin, for its original isolation from mouse osteoblasts (Okazaki et al., 1994).

It is known to be involved in differentiation and migration of different cell types (Kii et al.,

2004; McCusker et al., 2009; Monahan et al., 2007; Zhou and Snead, 2008).

16q21-22 loss has been implicated in a variety of cancers, strongly suggestive of tumor

suppressor genes located at this region. This might be due to the cluster of cadherin molecules

located at this locus (Figure 1.2). Cadherins are important for a wide variety of roles including

adhesion, signaling pathways, tissue morphology and maintenance of cell integrity, and so it is

not unexpected that the loss of these molecules would lead to progressive cancer phenotypes.

Cadherin genes located at this locus include CDH8, CDH11, CDH1, CDH3, CDH5, and CDH16.

16q loss has been seen in chromosomal aberration studies of cancers of the breast (Loo et al.,

2004), prostate (Saramaki and Visakorpi, 2007), lung (Sato et al., 1994), ovary (Kawakami et al.,

Page 26: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

13

1999), fallopian tubes (Snijders et al., 2003), pediatric medullablastomas (Lo et al., 2007),

neuroendocrine tumors (Kim do et al., 2008), prognosis of Wilms‟ tumors (Davidoff, 2009),

rhabdomyosarcoma (Visser et al., 1997), acute myeloid leukemia (Mrozek, 2008), astrocytomas

(Zhou and Skalli, 2000), and hepatocellular carcinomas (Herath et al., 2006).

Figure 1.2 Chromosome 16: Positioning of CDH11 and Cadherin Cluster.

Ideogram of chromosome 16 is from the website: http://www.genecards.org. Diagram is not drawn to scale and

represents the cadherin cluster at 16q21-22.1.

1.8 Cadherin Biology

1.8.1 Cadherin Structure

Cadherin molecules make up a superfamily of transmembrane glycoproteins that have been

implicated in many roles such as embryonic development, tissue morphogenesis, cellular

maintenance, differentiation, migration, and cancer progression (Behrens, 1999; Jeanes et al.,

2008; Tepass et al., 2000). This is due to their abilities to adhere to neighboring cells through

interactions via their extracellular domains and their cell signaling capabilities through their

interactions with catenin molecules via their cytoplasmic domains (Takeichi, 1995) (Figure

1.3A). Cell to cell interactions are usually homophilic, although heterophilic contacts have been

Page 27: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

14

observed (Volk et al., 1987). The cadherin superfamily is very large of >100 members, and

contains very diverse protein structures. However, they all share the characteristic extracellular

cadherin repeats (ECs) (Nollet et al., 2000).

The cadherin superfamily is split into many different subfamilies, which include the classical

cadherins. This subfamily is the most extensively studied, and includes approximately 20

members, that are split into two types, Type I and Type II. These types differ solely on the

presence of an HAV (Histidine-Alanine-Valine) motif in the Type I, and a QAV (Glutamine-

Alanine-Valine) motif in Type II (Blaschuk et al., 1990). Their protein structures are very

similar, with five extracellular domain (EC) repeats, a transmembrane domain and a cytoplasmic

domain that is highly conserved among cadherin subtypes (Halbleib and Nelson, 2006). In

classical cadherins, the cytoplasmic domain associates with α-catenin, β-catenin, γ-catenin, and

p120 (Kemler, 1993; Reynolds et al., 1996).

Homophilic interactions occur in the presence of calcium, which binds to the EC domains, and

regulates stability of the extracellular domain (Harrison et al., 2005; Hirano et al., 1987). Upon

cell-cell contact, cadherin molecules bind directly to β-catenin, γ-catenin, or p120. β-catenin

recruits α-catenin to the complex (Aberle et al., 1994), an actin filament binding protein and

links the complex to the cytoskeleton (Kobielak and Fuchs, 2004). This complex, known as the

cadherin cell adhesion complex (CAM), or the adherens junction (AJ), is necessary to maintain

cell-cell adhesion and cellular architecture (Aberle et al., 1996). These junctions are dynamic

and the structure and signaling provided by the complex ultimately determines the cellular

phenotype and behaviour (Wheelock and Johnson, 2003).

Page 28: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

15

1.8.2 Cadherin Regulation and Expression

Regulation of cadherin molecules is involved in normal tissue development and it is now clear

that classical cadherin dysfunction is a major contributor to cancer progression (Birchmeier and

Behrens, 1994; Thiery, 2002). Regulation happens at many levels, including gene expression,

transport to, and protein turnover at the cell surface (Figure 1.3B). Cadherin transcription is

directly regulated by DNA methylation and repression of promoter activity. Methylation of the

E-cadherin promoter is associated with reduced E-cadherin expression, disease progression and

metastasis in diffuse-type gastric cancer and ductal breast carcinoma (Strathdee, 2002).

Transcriptional repression of E-cadherin has been observed by zinc finger proteins of the

Slug/Snail family, resulting in decreased cell-cell adhesion and an increase in cell migration

(Halbleib and Nelson, 2006).

At the cell surface, cadherins are regulated by phosphorylation, ubiquitination and proteolysis.

Phosphorylation and dephosphorylation of various proteins in the AJ can positively or negatively

influence the structural integrity of the complex. Phosphorylation of the cadherin itself or β-

catenin, can affect and alter the binding affinity of the complex, or even target the cadherin for

endocytosis, resulting in degradation by the proteosome (Fujita et al., 2002; Roura et al., 1999;

Stappert and Kemler, 1994). In developmental processes, cells have to rapidly adapt to changes

in morphogenesis and tissue structure. This is often reflected in the dynamics of cadherin cell

adhesion, which can be regulated by protein turnover. One study showed that N-terminal

cleavage of Cdh11 by ADAM9 and 13 was essential for the migration of cells during the

development of the cranial neural crest, in vivo (McCusker et al., 2009). Furthermore, C-

terminal cleavage of N-cadherin by ADAM10, allows the cytoplasmic fragment to activate

CREB-mediated transcription, thought to have significance in neuronal growth and survival

(Marambaud et al., 2003).

Page 29: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

16

Receptor tyrosine kinases (RTKs) play important roles in development and cancer, and interact

with cadherins to influence cell-cell adhesion and signaling pathways (Pece and Gutkind, 2000).

For example, fibroblast growth factor receptors (FGFRs) are implicated in promoting epithelial

to mesenchymal transition (EMT) events in cancer progression (Thiery, 2002). In a mouse

pancreatic tumor model, N-cadherin forms a multi-protein complex with FGFR, leading to loss

of adhesion, dissemination of tumor cells and increased invasive properties (Cavallaro et al.,

2001). Mechanistically, binding inhibits internalisation of the cadherin and activates MAPK

signaling, leading to proliferation and migration (Suyama et al., 2002).

Cadherins are also regulated through the cytoplasmic proteins they bind to (Figure 1.3B). p120

has dual roles in the cell as it acts as a member of the AJ, and interacts with the Rho family of

GTPases in the cytoplasm. It has been seen to regulate and stabilize cadherin strength and

turnover, its significance shown by experiments where ablation of p120 in mice led to down-

regulation of E-cadherin levels (Davis et al., 2003). Upon loss of cadherins, p120 accumulates

in the cytoplasm, repressing RhoA and activating Cdc42 and Rac1, which modulate the

cytoskeleton and increase migration and invasiveness (Christofori, 2006; Stemmler, 2008). β-

catenin also has a dual role in cell adhesion and as the intracellular transducer in the canonical

Wnt signaling pathway, acting as a transcriptional coactivator in the nucleus (Gumbiner, 1995).

In the presence of cadherins, newly synthesized β-catenin saturates the pool of the AJs, and is

never available for signaling. Changes in cell-cell adhesion lead to excess β-catenin, where the

cytoplasmic pool is regulated by Wnt signaling. If the pathway is inactive, the

APC/AXIN/GSK-3β complex phosphorylates β-catenin. It is then ubiquitinated, and targeted for

destruction by the proteosome (Aberle et al., 1997). Upon activation of Wnt ligands, the

complex is inhibited and β-catenin accumulates and transports to the nucleus where it binds to

transcription factors T cell factor/ lymphocyte enhancer binding factor (LEF/TCF) affecting

Page 30: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

17

transcription of target genes (Behrens et al., 1996; Ikeda et al., 2000). Wnt signaling ultimately

activates transcriptional programs and there is no limit to the type of biological event that may be

controlled (Clevers, 2006). Wnt signaling has been implicated in many cancers. For instance,

some direct target genes include proto-oncogenes c-myc and cyclinD1, and have been the focus

of many studies. Furthermore, in some cancers, one or more of the Wnt signaling proteins are

mutated, leading to aberrant signaling, and malignant phenotypes (Kinzler and Vogelstein, 1996;

Morin et al., 1997). Wnt signaling has even been implicated as having tumor suppressor

functions in retinoblastoma (Tell et al., 2006).

Cadherin switching is a prominent feature in morphogenetic function and cell sorting during

development (Wheelock and Johnson, 2003). The same phenomenon highly occurs in tumor

progression (Christofori, 2003). In a number of cancer types, E-cadherin expression is lost, and

occasionally, de novo expression of another cadherin, N-cadherin or Cdh11, has been observed

(Li and Herlyn, 2000; Tomita et al., 2000). This recapitulates what is seen during EMT in

development; for example when cells switch from E-cadherin to N-cadherin in primordial germ

cells when migrating to populate the genital ridge (Bendel-Stenzel et al., 2000). Based on

numerous studies, cadherin switching seems to be a hallmark of the transition from a benign to

malignant form of cancer, and correlates with invasiveness and poor prognosis (Cavallaro and

Christofori, 2004; Christofori, 2003; Halbleib and Nelson, 2006).

Page 31: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

18

Figure 1.3 Classical Cadherin Structure and Regulation.

A. This depicts a classical cadherin involved in the Adherins Junction, mediating cell-cell adhesion. In the presence

of Calcium, the extracellular domains bind to cadherin molecules on neighbouring cells in homophilic interactions.

The cytoplasmic domains interact with various catenin molecules, ϐ-catenin, α-catenin and p120, which link the

complex to the actin cytoskeleton, mediating cell-cell adhesion and intracellular signalling pathways. B. Signalling

Pathways may be affected with the down-regulation of cadherin molecules. p120 has a dual role in cell adhesion

and interacting with Rho-GTPases. Upon loss of cell-cell adhesion, p120 interacts with Cdc42 and Rac1, involved

in promoting cell motility and invasiveness. ϐ-catenin also has dual roles in cell adhesion and as a main player in

Wnt signalling. Upon loss of cell-cell adhesion, ϐ-catenin accumulates in the cytoplasm, and when Wnt signalling is

active, translocates to the nucleus interacting with transcription factors to regulate expression of many genes,

including proto-oncogenes, cyclin-D1 and c-myc. When Wnt is not activated, the free ϐ-catenin in the cytoplasm is

phosphorylated, targeting it for degradation.

1.8.3 Cadherins in the Mammalian Retina

Cadherin molecules have major roles in development, suggested by their tightly regulated

expression patterns and numerous functional studies (Gumbiner, 1996; Huber et al., 1996). They

have a role in the CNS, in establishing and maintaining synaptic connections and in

Page 32: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

19

differentiation of the cranial neural crest (Colman, 1997; Zhou and Snead, 2008). The retina, in

common with the CNS, develops from the neuroepithelium, and the processes that govern this

process are similar to that of the brain (Honjo et al., 2000). Earlier studies examining the

cadherin expression patterns in the developing mouse retina suggest that cadherin expression

coupled with temporal regulation suggested roles for these molecules in cell migration, synaptic

formation, and selective cell associations (Faulkner-Jones et al., 1999b). Cumulatively, studies

showed eleven different cadherin molecules to be expressed in the postnatal mouse and chick

retinas. These include N-cadherin (CDH1), E-cadherin (CDH2), P-cadherin (CDH3), R-cadherin

(CDH4), VE-cadherin (CDH5), CDH6, CDH7, CDH8, CDH10, OB-cadherin (CDH11), and

CDH12 (Faulkner-Jones et al., 1999a; Faulkner-Jones et al., 1999b; Honjo et al., 2000; Wohrn et

al., 1998). Out of these, N-cadherin has been the most extensively studied in the mammalian

retina. Studies show that early in development all undifferentiated cells express N-cadherin,

leading to axonal outgrowth (Kljavin et al., 1994), and downregulated later in development,

where it is localized to the INL (Honjo et al., 2000). Deregulation leads to deficits in visual

behaviour of Drosophila correlated to disruptions in photoreceptor connectivity (Lee et al.,

2001). Further studies in zebrafish reported crucial roles for N-cadherin in retinal lamination and

retinal organization (Erdmann et al., 2003; Masai et al., 2003). Cadherin-6 has also been

implicated to have a role in formation of the retina as knockdown zebrafish embryos showed

severely disrupted differentiation of retinal cell layers (Liu et al., 2008). In Xenopus embryos,

loss-of-function studies reported a decrease in cell proliferation, retinal lamination defects and

disruptions in structural organization (Ruan et al., 2006). Other cadherins have not been as

extensively studied, but are suggested to have various roles during retinogenesis, based on

restrictions in expression patterns throughout development. For instance, both R-cadherin and

cadherin-6 are concentrated in the GCL and amacrine layers, suggestive of a role for these

Page 33: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

20

cadherins in interneural connections between restricted pairs of neurons in retinogenesis (Honjo

et al., 2000). The role of Cdh11 in retinal development is still unclear, as studies in knockout

mice show no obvious phenotype (Marchong, Yurkowski, et al, PLOs Genetics (in press)). It is

postulated that this could be due to compensation by expression of other cadherins.

1.8.4 Cadherin-11 and its Isoforms

Cadherin-11 is a member of the Type II classical cadherins. Type I cadherins usually have broad

distributions segregated by embryonic germ layer or tissue type (Nishimura et al., 1999), while

Type II cadherins are often found in overlapping and tightly regulated patterns of expression, in

loosely associated tissues and in weaker intercellular adhesion (Takeichi, 1995). Cdh11 is

primarily expressed in tissues derived from the mesoderm (MacCalman et al., 1996; Shibata et

al., 1996). Cdh11 is a 796 amino acid protein that exists in three different isoforms: intact,

variant and secreted. It was originally cloned from mouse osteoblasts, but was first found in

nervous tissue (Okazaki et al., 1994; Tanihara et al., 1994). It resides in the human genome on

Chr. 16, and is a 120-kD protein, with a 97% homology to its mouse counterpart, on Chr. 8 in the

mouse genome (Hoffman and Balling, 1995; Okazaki et al., 1994). The variant form shows a

179bp insertion in the transmembrane domain, leading to a frameshift and alternative splicing,

resulting in an altered cytoplasmic region and an 85-kd protein (Kawaguchi et al., 1999). It also

lacks cell-cell adhesion properties, but assists the intact form in adhesion. The secreted form is

an 80-kd protein and thought to be a result of proteolysis of the intact form as seen in

osteosarcoma samples, resulting in downregulation of Cdh11 (Kashima et al., 1999) (Figure 1.4).

Page 34: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

21

Figure 1.4 Cadherin-11 and its Isoforms.

A. Human cadherin-11 exists in three isoforms: an intact form, an alternatively spliced variant form and a secreted

form, thought to be derived from the intact form via proteolysis. B. Mouse cadherin-11 is located on chromosome 8

and is 97% homologous to human cadherin-11. Also shown is the inactive form of cadherin-11 found in cadherin-

11 knockout mice generated by using a targeting vector. A PGKNeopA gene cassette replaced the last 56 AA of the

extracellular domain and some of the transmembrane domain (Horikawa et al, 1999).

1.8.5 Cadherin-11: Implicated Roles

Cadherin-11 knockout mice were previously used to analyze its role in different tissues. In

Cdh11 null mice, a significant decrease in bone density was observed at specific parts of the

skeleton, suggesting a role in osteoblast differentiation and mineralization of the osteoid matrix

(Kawaguchi et al., 2001). Another study observed that mice null for Cdh11 showed reduced fear

Page 35: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

22

and anxiety responses in the brain, as well as increased long-term potentiation, suggesting Cdh11

junctions are necessary for normal development of synaptic organization in the brain (Manabe et

al., 2000). Additionally, developing somites of mice with disrupted Cdh11 genes were observed,

but showed no structural anomalies. However, mice null for both Cdh11 and N-cadherin

resulted in greatly fragmented somites, a more dramatic phenotype than in N-cadherin null mice

alone, suggesting the two cadherins worked together in somite morphogenesis (Horikawa et al.,

1999).

These and further roles for Cdh11 have since been studied. Cdh11 is expressed in motor and

sensory neurons in the developing mouse embryo, implicating a role in growing motor axons,

controlling growth and interactions of the growth cone (Marthiens et al., 2005). Additionally,

roles have been observed for Cdh11 in differentiation of osteoblasts and chondroblasts from

mesenchymal cells (Kii et al., 2004), as well as in human trophoblast cells (Getsios and

MacCalman, 2003). Recent studies have focused on identifying the role for Cdh11 in formation

and organization of the synovium (Kiener et al., 2006; Valencia et al., 2004). Mice null for

Cdh11 showed a resistance to inflammatory arthritis (Lee et al., 2007), and L cells transfected

with Cdh11 showed an increase in invasive capabilities in chronic synovitis and rheumatoid

arthritis (Kiener et al., 2009). Lastly, Cdh11 has been observed to have roles in cranial neural

crest differentiation and migration. One study showed human embryonic stem (ES) cells

positive for Cdh11 expression, were multi-progenitor cells capable of differentiating into fates

associated with the cranial neural crest (Zhou and Snead, 2008). Cdh11 extracellular cleavage

by a disintegrin and metalloprotease (ADAM) promoted migration of CNC cells through

modifying cell-cell adhesion (McCusker et al., 2009). These studies have proven the wide

variety of roles cadherin molecules can have. Cdh11 itself has multiple roles ranging from

development to differentiation and migration in different tissue types.

Page 36: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

23

1.8.6 Cadherins in Cancer Progression

1.8.6.1 Cadherin-11

Allelic loss of 16q21-22 has been seen in a variety of cancers. In retinoblastoma, we observed

loss of Cdh11 expression in 91% of cancer cell lines and retinoblastoma primary tumors

(Marchong et al., 2004). Cdh11 has also been implicated in the progression of a variety of

tumors over the past decade. Like retinoblastoma, similar patterns of loss have been observed in

osteosarcoma. Original studies showed altered expression of Cdh11 in human osteosarcomas

(Kashima et al., 1999). High expression of the alternatively spliced variant form of Cdh11

suppressed the intact form (Kawaguchi et al., 1999). Additionally, the secreted form was most

prevalent and was suggested to disrupt cell-cell adhesion. It was hypothesized that down-

regulation of Cdh11 was related to morphology and metastatic potential (Kashima et al., 1999).

The same group then showed that while there was strong expression of Cdh11 in normal

osteoblasts, there was faint expression in osteosarcoma. Their study showed overexpression of

Cdh11 in osteosarcoma cell line LM8 led to a marked reduction pulmonary metastasis in vivo

(Kashima et al., 2003). More recently, two cell lines developed from the primary tumor and

metastasis of an osteosarcoma patient, showed decreased expression of Cdh11 in the metastatic

cell line, suggesting a role in the metastatic process (Zou et al, 2008). These results are

paralleled by a recent study where expression of Cdh11 in normal osteoblast cell lines was

followed by marked decreases in expression in primary tumors cell lines and again in metastatic

cell lines (Nakajima et al., 2008). This same study revealed Cdh11 expression was significantly

correlated to the patient‟s survival and was suggested as a prognostic marker for osteosarcoma.

Besides osteosarcoma, decreased Cdh11 expression has been observed in a subset of colon

cancers (5 of 23) (Braungart et al., 1999), and as a frequent molecular event with the transition of

normal astrocytes to astrocytomas (Zhou and Skalli, 2000). However, Cdh11 has also been

Page 37: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

24

observed to be increased in a variety of cancers. This is evident in numerous studies that

implicated Cdh11 in the progression of breast cancer, suggestive of Cdh11 as a marker of

invasive and aggressive forms (Bellahcene et al., 2007; Pishvaian et al., 1999; Sarrio et al.,

2008). It also has been seen to be up-regulated in rhabdomyosarcomas (Markus et al., 1999),

signet ring carcinomas (Shibata et al., 1996), nephroblastomas (Ramburan et al., 2006), and in

prostate cancer cell lines (Bussemakers et al., 2000), tumor samples (Tomita et al., 2000), and

metastasis (Chu et al., 2008). The observation that Cdh11 is lost in some cancers and gained in

others suggests a role based on tissue type and microenvironmental signals, although the

mechanisms are still unclear. Differences in adhesion strength, extracellular binding partners or

alterations in cell signaling via the cytoplasmic domains are all possibilities.

1.8.6.2 E-Cadherin

E-cadherin and its role in cancer progression is the most intensely studied of any of the

cadherins. It is down-regulated in a variety of cancers such as gastric carcinomas, breast, colon,

esophagus, liver, and pancreatic cancers (Beavon, 2000; Munro et al., 1995; Oka et al., 1993;

Tamura et al., 1996a; Tamura et al., 1996b). Its function in the progression of cancer was best

shown by abrogation of invasive phenotype when different cells of different cancer types were

transfected with E-cadherin cDNA (Chen and Obrink, 1991; Luo et al., 1999; Vleminckx et al.,

1991). Its tumor suppressive function was highlighted by revealing its prominent role in driving

numerous cancers to invasive and metastatic forms (Beavon, 2000; Gottardi et al., 2001;

Nawrocki-Raby et al., 2003; Perl et al., 1998). The mechanism behind its suppression is not yet

fully known, but many studies have supported its tumor suppressor abilities through the finding

of different mutations, and biallelic inactivation in breast and diffuse gastric tumors (Becker et

al., 1996; Becker et al., 1994). Germline mutations have been identified in gastric cancer

samples (Gayther et al., 1998; Guilford et al., 1998), as well as somatic mutations in gastric,

Page 38: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

25

breast, endometrium and ovary cancers (Becker et al., 1996; Hajra and Fearon, 2002). Missense

mutations lead to increased cell motility and decreased cell adhesion (Handschuh et al., 1999;

Handschuh et al., 2001). Promoter hypermethylation has been seen in acute leukemia and

numerous carcinomas including breast, gastric, hepatocellular, esophageal and renal cell (Corn et

al., 2001; Corn et al., 2000; Hu et al., 2002; Kanai et al., 1997; Nojima et al., 2001; Tamura et

al., 2001; Zhao et al., 2007). Transcriptional repression via proteins such as Slug and Snail, have

been suggested by binding to the CDH1 promoter (Halbleib and Nelson, 2006). Other

mechanisms recently reported include post-transcriptional mechanisms such as phosphorylation

by tyrosine kinases, which induce endocytosis and subsequently target E-cadherin for

ubiquitination and degradation (Christofori, 2006; Fujita et al., 2002). Phosphorylation of

catenins such as β-catenin has can lead to invasion through disassembly of the complex and

induction of migration (Hu et al., 2001). Posttranslational studies looking at truncation of the E-

cadherin protein present another argument in down-regulation in cancers. Studies show both

extracellular cleavage through metalloproteinases and cytoplasmic cleavage yields truncated

proteins that are inactivated, inhibit cell aggregation, and enhance tumorigenesis and metastasis

(Noe et al., 2001; Rashid et al., 2001; Rios-Doria et al., 2003).

1.8.6.3 N-Cadherin

N-cadherin (CDH2) is usually expressed in tissues of neuronal origin. N-cadherin is a hallmark

of EMT and is usually up-regulated in cancers, especially when E-cadherin is down-regulated,

leading to an increased affinity for mesenchymal cells (Yilmaz and Christofori, 2009). The gain

of N-cadherin provokes increased cell migration, invasion and metastasis (Hulit et al., 2007;

Nieman et al., 1999). An up-regulation of N-cadherin is typically seen in cancers such as breast,

prostate and gastric carcinomas (Gravdal et al., 2007; Hazan et al., 2000; Hazan et al., 2004;

Nagi et al., 2005). An increase in N-cadherin expression and presence of an N-cadherin/catenin

Page 39: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

26

complex is observed in invasive retinoblastoma samples (Mohan et al., 2007; Van Aken et al.,

2002). Mechanisms leading to up-regulation of N-cadherin are undefined, but several proteins

have been linked to its increased expression. Twist, a transcriptional repressor, was seen to be

up-regulated in certain cancers, and led to the induction of N-cadherin expression (Alexander et

al., 2006; Yang et al., 2007). N-cadherin interacts with different signal transduction pathways.

For example, it interacts with the fibroblast growth factor receptor (FGFR), activating the MAPK

pathway. As a result, increased proliferation and matrix metalloproteinase (MMP) secretion

promote invasiveness in N-cadherin expressing cells (Suyama et al., 2002).

1.9 Cadherin-11 as a Tumor Suppressor in Retinoblastoma Progression

As a candidate gene in retinoblastoma progression, preliminary assessment of the tumor

suppressor role of Cdh11 was done in TAg-RB mice (Figure 1.5). Immunohistochemical

staining revealed Cdh11 was lost in 38% of primary tumors of TAg-RB mice, over a time period

of five months, paralleling human retinoblastoma genomic changes. When TAg-RB+/-

mice were

crossed with Cdh11+/+

and Cdh11-/-

mice to examine tumorigenesis, tumor volumes were

calculated per single TAg positive cell at post-natal day 8 (PND8), to a large tumor at PND84.

Volumes were larger overall in Cdh11-/-

mice compared to mice with normal Cdh11 alleles and

tumor growth was faster when quantified between PND28 and PND84 (Marchong, Yurkowski et

al, PLOs Gentics (in press)). This suggested Cdh11 was acting as a tumor suppressor. Tumor

growth can be defined as a fine balance between cell proliferation and cell death and therefore,

both possibilities were considered. Preliminary experiments showed no statistical differences in

the expression of proliferation marker PCNA, but showed a statistical difference in activated

caspase-3 staining as a marker of apoptosis when extrapolated to the entire tumor. This

preliminary data suggested that Cdh11 has a role in apoptosis and led to two hypotheses. These

hypotheses are addressed in the specific aims for this study and are discussed in section 1.11.

Page 40: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

27

Figure 1.5 Cadherin-11 as a Tumor Suppressor in Retinoblastoma Progression.

A. Cadherin-11 expression in normal murine retina resides in the INL, where human and murine retinoblastomas

initiate from. B. Cadherin-11 expression was lost in 38% of TAg-RB tumors analyzed by RT-PCR (Marchong et al,

2004). C. When quantifying tumor growth in TAg-RB tumors from post natal day (PND) 28 to PND84, growth was

faster in tumors of mice null for Cdh11. D. No statistical difference was seen between TAg-RB tumors null and

wildtype for Cdh11 in staining for proliferation marker, PCNA. Statistical difference was seen between genotypes

in staining for caspase-3, a pro-apoptotic marker, with mice null for Cdh11 showing decreased caspase-3 staining.

1.10 Apoptosis

Apoptosis is an evolutionary conserved programmed cell death mechanism that is crucial in

development and homeostasis (Hengartner, 2000). It is characterized by typical morphological

Page 41: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

28

and biochemical hallmarks such as cell shrinkage, DNA fragmentation and membrane blebbing

(Hengartner, 2000; Wyllie et al., 1980). The genetic basis of apoptosis implies that cell numbers

can be regulated by cell death, in addition to proliferation and differentiation, and like any other

metabolic or developmental program, can be affected by mutation (Thompson, 1995). Studies

first showed large percentages of cell loss seen in tumors was due to apoptosis (Kerr et al.,

1972). Subsequent studies revealed a higher frequency of apoptosis in spontaneously regressed

tumors and in tumors treated with cytotoxic anticancer agents (Kerr et al., 1994). In recent

years, numerous studies have reported deregulation of apoptosis in the progression of multiple

cancers (Kerbauy and Deeg, 2007). Research has also focused on what factors trigger this

deregulation, such as imbalances of growth and survival factors, loss of cell-cell adhesion,

hypoxia and radiation. Imbalances in internal factors such as DNA damage, telomere

malfunction, or inappropriate proliferation signals can also lead to apoptosis (Hartland et al.,

2009; King and Cidlowski, 1995; Lowe and Lin, 2000).

1.10.1 Extrinsic Pathway

It is now well accepted that there are two distinct pathways of cell death, the extrinsic and

intrinsic pathways (Figure 1.6). The extrinsic pathway is initiated by „death receptors‟, members

of the tumor necrosis factors (TNF) receptor gene superfamily. This consists of more than 20

proteins which have a wide range of biological functions such as cell death, survival,

differentiation and immune regulation (Walczak and Krammer, 2000). The best characterized

death receptors include CD95 (Apo-1/Fas), TNF receptor 1 (TNFR1), and TNF-related

apoptosis-inducing ligand-receptor 1 (TRAIL-1) and TRAIL-2 (Walczak and Krammer, 2000).

Upon binding of their ligands the receptors are trimerized and signal transducing molecules are

recruited (Walczak and Krammer, 2000). For example, upon TRAIL binding to its receptor, the

Fas-associated death domain (FADD) is recruited along with the initiator caspase-8 (Sprick et

Page 42: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

29

al., 2000). Caspase-8 cleaves itself to its activated form, which initiates a protease cascade,

cleaving cellular targets and resulting in cell death (Cohen, 1997). This signaling can be

negatively regulated by proteins recruited to receptor cytoplasmic domains, like cellular caspase-

8 (FLICE)-like inhibitory protein (cFLIP). Recruitment of FLIP instead of procaspase-8 can

block caspase activity (Krueger et al., 2001).

1.10.2 Intrinsic Pathway

The intrinsic pathway of apoptosis is initiated by activating permeabilization of the outer

mitochondrial membrane. This is achieved by numerous cytotoxic and pro-apoptotic signals

converging on the outer membrane, including proteins of the Bcl-2 family (Green and Kroemer,

2004). BH3-containing molecules activate mitochondrial permeability transition by inactivation

of anti-apoptotic Bcl-2 family members, like Bcl-2 or Bcl-Xl, or through activation of pro-

apoptotic members, such as BAX (Ghiotto et al., 2009). This results in the release of proteins

into the cytosol that activate the central apoptotic pathway. Cytosolic cytochrome c interacts

with apoptotic-protease-activating factor 1 (APAF1) and caspase-9, which forms a heptameric

complex, the apoptosome (Acehan et al., 2002). The formation of this complex initiates a

protease cascade, similar to described above, leading to widespread cleavage of apoptotic

substrates (Li et al., 1997).

Apoptosis cannot be simply described by two parallel pathways. Firstly, studies in mice showed

that activation by death receptors was highly variable depending on cell type and specific to

microenvironmental stimuli (Hakem et al., 1998; Yoshida et al., 1998). Secondly, links between

the mitochondrial and death receptor pathway exist leading to „cross-talk‟ on different levels.

For example, caspase-8 activation through death receptors can lead to cleavage and activation of

Page 43: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

30

BID (tBID), a member of the Bcl-2 family thought to facilitate release of cytochrome c from the

mitochondria (Billen et al., 2008).

Figure 1.6 Apoptotic Pathways.

A. The extrinsic pathway is initiated by death receptors of the tumor necrosis (TNF) death receptor gene

superfamily activating caspase-8 and initiating the protease cascade leading to cell death. B. The intrinsic pathway

is initiated by permeability of the mitochondrial membrane, leading to release of proteins and cytochrome c,

interacting with caspase-9 and APAF1 to form the apoptosome, initiating a protease cascade. Cross-talk also occurs

through caspase-8 activation of BID, a Bcl-2 family member that releases cytochrome c from the mitochondria.

1.11 Project Aims and Hypothesis

1.11.1 To examine if Cadherin-11 is promoting apoptosis in TAg-RB tumors and define the

acting apoptotic pathway

1.11.2 To determine if the loss of Cadherin-11 expression leads to invasive and aggressive

tumors through expression of invasion markers

Page 44: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

31

Chapter 2

Methods and Materials

2.1 Mouse Models

Cdh11 knockout mice (Cdh11-/-

), background strain 129, were originally provided by Dr. M

Takeichi (Horikawa et al., 1999; Kawaguchi et al., 2001). One generation crosses were made

between Cdh11-/-

129 and Cdh11+/+

C57Bl-6 to get a mixed background of 129/C57Bl-6.

Cdh11-/-

littermates were used to make one generational crosses with SV40 TAg-RB (TAg+/-

),

background strain C57Bl-6, which were available in the lab and first provided by Joan O‟Brien

(Windle et al., 1990). These littermate, double heterozygotes Cdh11+/-

TAg+/-

, on a 129/C57Bl-6

background, were further crossed with Cdh11-/-

, Cdh11+/-

or Cdh11+/+

of a 129/C57Bl-6 mixed

background. These crosses yielded the three genotypes used for this study: Cdh11-/-

;TAg+/-

,

Cdh11+/-

;TAg+/-

and Cdh11+/+

;TAg+/-

. These mice were sacrificed at post natal day 84 (PND84),

a timepoint of three months.

2.2 Genotyping

Genotyping of Cdh11-/-

mice and their littermates were carried out using genomic DNA isolated

from mouse tail. PCR conditions were previously described by Dr. M Takeichi (Horikawa et al.,

1999). PCR reactions were performed in a RoboCycler Gradient 96 thermal cycler (Stratagene).

This included 940C, 2 min, 1 cycle [94

0C, 30s, 50

0C, 30s, 72

0C, 30s] 35 cycles, 72

0C 10 min, and

400C, cool block. Primers used were: forward, 5‟ to 3‟ (21bp): TTC AGT CGG CAG AAG CAG

Page 45: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

32

GAC and backward, 5‟ to 3‟ (19bp): GTG TAT TGG TTG CAC CAT G, and neo 5‟ to 3‟ (23bp):

TCT ATC GCC TTC TTG ACG AGT TC. The sizes of expected PCR products were: Cdh11+/+

:

240bp, Cdh11+/-

: 480bp and 240bp, and Cdh11-/-

: 480bp. The reaction mixture included 2.5 µl

of 2 mM dNTPs, 1.5 µl 25 mM MgSO4, 2.5 µl KOD 10X PCR buffer (Novagen), 0.5 µl forward

5‟ primer, 1.0 µl reverse 3‟ primer and 0.5 µl neo primer, 0.25 µl KOD hot start DNA

polymerase (Novagen), 1.0 µl template, up to 25 µl with ddH2O. Genotyping of TAg mice and

littermates were carried out using PCR conditions: 940C, 2 min, 1 cycle, [94

0C, 1 min, 58

0C, 1

min, 720C, 1min] 30 cycles, 72

0C, 10 min, 1 cycle and 4

0C, cool block. Primers used were:

forward, 5‟ to 3‟ (30bp): GAC TTT GGA GGC TTC TGG GAT GCA ACT GAG and backward: 5‟

to3‟ (30bp): GGC ATT CCA CCA CTG CTC CCA TTC ATC AGT. The size of the expected PCR

product was 420bp. The reaction mixture included 2.5 µl 10XPCR buffer (100 mMTris-HCl pH

8.3; 500 mM KCl), 0.5 µl 2 mM dNTPs, 2 µl 25 mM MgCl2, 0.5 µl each of forward and reverse

primers, 0.5 µl Taq polymerase isolated from E.coli that has been transformed with pTaq

plasmid, 1.0 µl template up to 25 µl total volume. All animals were treated in accordance with

protocols approved by the Animal Care Committee of the Ontario Cancer Institute.

2.3 Histology and Slide Selection

Mouse eyes were dissected and fixed in freshly prepared 4% PFA for 24hrs and then stored in

70% EtOH at 40C. Eyes were then paraffin embedded and sectioned at 0.5 µm (Pathology,

Hospital for Sick Children, ON, Canada). For littermates Cdh11+/+

;TAg+/-

, Cdh11+/-

;TAg+/-

and

Cdh11-/-

;TAg+/-

, horizontal serial sections were specifically made through the entire eye and

optic nerve averaging approximately 270-420 sections per eye with 5-7 sections per slide and

approximately 60 slides per eye. To estimate apoptosis and proliferation throughout the entire

tumor, every 10th

slide was chosen for analysis, averaging to 6 slides per eye. Every section on

each slide was used in the analysis. One eye was analyzed per mouse and 4-6 mice were

Page 46: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

33

analyzed in each experiment. To examine the histology, slides were stained with hematoxylin

and eosin and then scanned using the Aperio ScanScope XT.

2.4 Immunohistochemistry

Slides that were chosen for analysis were processed through 100% xylene (2 times 10 min

incubations), 100% ethanol washes (2 X 5 min incubations), and 2 min each in 95%, 70% and

50% ethanol. This was followed by a rinse in 1X TBS for 1 x 5 min incubation. Slides were

placed into PBS citrate solution and heat treated for antigen retrieval. This was performed in a

pressure cooker in the microwave at power level 10 for 15 min and then power level 7 for an

additional 5 min. Slides were removed after reaching room temperature, rinsed in 1X TBS for 2

min and treated with Triton-X-100 for 10 min. Afterwards, the slides were rinsed in 1X TBS for

5 min, and then protein blocked using 10% DakoCytomation Protein Block (Ref#X0909,

Lot#1001228) for 30 min. Slides were incubated in primary antibodies made in 10%

DakoCytomation Antibody Diluent (Ref# S3022, Lot# 036205) and 1% BSA/TBST overnight at

40C. These were used at various dilutions, depending on the specific antibody. The next day the

slides were washed in 0.1% BSA/TBST for 3 X 10 min incubations. To visualize pro and anti-

apoptotic stained cells, a Biotin/Streptavidin protocol was used. Slides were incubated with

biotinylated secondary antibodies, either anti-mouse, anti-rabbit, or anti-goat, used at a dilution

of 1:200 in 1% BSA/TBST and 10% DakoCytomation Antibody Diluent, for 1 h at room

temperature. Slides were then again washed with 0.1% BSA/TBST for 3 X 10 min incubations.

Slides were then incubated with either Streptavidin-Alexa488 or Streptavidin-Alexa594, used at

1:200 dilution made in 1X TBS, for 15 min at room temperature. Slides were briefly washed in

1X TBS and incubated for 15 min in 4‟,6-diamidino-2-phenylindole (DAPI), used at 1:50

dilution in 1X TBS, at room temperature. After another brief wash in 1X TBS, slides were

mounted using DakoCytomation Fluorescent Mounting Medium (Ref#S3023, Lot#10027230).

Page 47: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

34

Slides were analyzed under a fluorescent microscope and quantified for apoptotic expression.

Slides stained for BrdU were first treated in 2M HCl for 20 min at room temperature, followed

by incubation with 2M sodium borate for 2 min. This was followed by a 5 min wash in PBS and

then treated as previously described until detection of the antibody. To analyze T-antigen and

BrdU stained cells, an Immunopure metal-enhanced diaminobezidine (DAB) Substrate Kit

(Thermoscientific) was used. Slides were incubated for 1h in ABC solution (Vectastain ABC

Elite, Vector Laboraties). The solution was prepared one half hour before incubation by adding

1 drop of each reagent A and reagent B, in 2.5 ml 1X TBS. After incubation slides were washed

for 5 min in 1X TBS, and stained cells visualized after treatment for 3-10 min in DAB Substrate

solution (Thermoscientific) prepared fresh with 10% DAB/Metal Concentrate, 10x (Lot#

KA129317, Prod#1856090) made in 1X Stable Peroxide Buffer (Lot#JA119477, Prod#

1855910). Slides were scanned and analyzed on computer (Section 2.6). Every 15th

slide

sectioned was also stained for Hematoxylin and Eosin (H&E) and analyzed using light

microscopy. Table 2.1 provides a complete list of all antibodies used in experimental analysis.

Page 48: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

35

Table 1 Antibodies used in Experimental Analysis.

Antibody Name Company Dilution Used

OB-Cadherin (anti-Cadherin-11)

rabbit polyclonal

ZYMED Laboratories Cat#71-7600,

Lot#389746A

1:100

BrdU (purified anti-bromodeoxy-

uridine) mouse monoclonal

BD Biosciences Pharmigen

Cat#555627, Lot#52817

1:200

Caspase-3 rabbit polyclonal Promega

Cat#G7481, Lot#242477

1:100

Caspase-8 rabbit polyclonal Abnova Antibody Innovation

Cat#PAB0246, Lot#30476849

1:1000

Caspase-9 rabbit polyclonal Cell Signaling Technology

Cat#9509, Lot#83

1:100

BAX (B-9) mouse monoclonal Santa Cruz Biotechnology, Inc

Cat#sc-7480

1:100

TRAIL (K-18) goat polyclonal Santa Cruz Biotechnology, Inc

Cat#sc-6079, Lot#E3008

1:100

Anti-Bid mouse monoclonal Novus Biologicals Cat#NB100-

94174, Lot#B1249056

1:50

FLIPs/l (G-11) mouse monoclonal Santa Cruz Biotechnology, Inc

Cat#sc-5276, Lot#F0208

1:100

FAS (X-20)-G goat polyclonal Santa Cruz Biotechnology, Inc

Cat#sc-1024-G, Lot#K037

1:100

TNF alpha rat monoclonal [MP6-

XT22]

Abcam Cat#ab39542 1:100

Bcl-xL (H-5) mouse monoclonal Santa Cruz Biotechnology, Inc

Cat#sc-8392, Lot#A079

1:100

Bcl-2 rat monoclonal Santa Cruz Biotechnology, Inc

Cat#sc-578, Lot#C187

1:100

Page 49: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

36

2.5 BrdU Incorporation

To analyze proliferating cells at PND84, mice were given an intraperitoneal (IP) injection with

bromodeoxyuridine (BrdU) reagent (5-bromo-2‟-deoxyuridine and 5-fluoro-2‟-deoxyuridine,

10:1, used at 1 ml reagent per 100g body weight, Cat#555627, Lot#33076 , BD Pharmigen).

After 4 h, the mice were sacrificed and eyes fixed as previously mentioned.

2.6 Image Analysis

To analyze proliferation within the tumors at PND84, the amount of BrdU staining was

quantified by scanning representative slides at the Advance Optical Microscopy Facility

(AOMF) at the Ontario Cancer Institute using an Aperio ScanScope CS. Images were retrieved

using ImageScope software and all analyzed as TIFF images using ImageJ: Image Processing

and Analysis in Java software, downloadable from http://rsb.info.nih.gov.ij/. The amount of

proliferating cells determined by BrdU positive staining was determined by previous methods

used in the lab (Dimaras et al., 2009). The BrdU positive cells were traced and the area was

measured in pixels. The traced areas were converted into an 8-bit scale. By a manually selected

threshold tool, the area of proliferating cells (DAB stained) was measured by the program in

pixels. The area of BrdU positive, proliferating cells in pixels, was compared to the total tumor

area, also measured in pixels (area of BrdU cells in pixels/tumor area in pixels)*100 (Figure 3.6).

24 sections were analyzed per eye to account for differences throughout the tumor. To quantify

the amount of apoptotic proteins, images were analyzed using a 40x inverted microscope (Leica

DMLB). Selected slides were analyzed under 40x to 100x to precisely identify positively stained

apoptotic cells. Double and triple staining was performed with additional antibodies and with

DAPI to confirm cellular staining. To quantify the amount of binding of each antibody, slides

were manually scanned through with the microscope and individual positively stained cells were

Page 50: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

37

counted. This was done for each section on the selected slides to total 36 sections per eye.

Representative slides were selected throughout the entire eye to account for any differences that

may be occurring throughout the tumor. From a total number of apoptotic cells counted from

each section, an average was taken of positively stained cells. This led to a total number of

apoptotic cells that could be found on any section within the tumors (Figure 3.3).

2.7 Statistical Analysis

A total of 5 Cdh11-/-

;Tag+/-

and 5 Cdh11+/+

;Tag+/-

mice were sacrificed at PND84 for the analysis

of apoptotic protein expression. A total of 6 mice per genotype were sacrificed at PND84 for

BrdU injection and analysis of proliferation within the tumors. The student‟s t-test was used to

assess results as significant, using a 95% confidence level. This was accessed from

http://www.graphpad.com/quickcalcs/ttest1.cfm.

2.8 Tissue Culture and Cell Lines

All cell lines used were grown in a humidified 370C incubator with 10% CO2 concentration.

They were grown in their respective tissue culture media (Tissue Culture Media Facility,

OCI/PMH, ON, CA). Cell lines used for analysis included T+532 (22wks), T+539b (20wks),

T+572a (22wks), T+794a (22wks), T+818a (19wks), and T+827b (20wks). These lines were all

derived from primary TAg-RB tumors, at their respective time points, as shown in brackets.

Each of these lines were grown in cell culture media DMEM-H21 (Gibco) with the addition of

100 U/ml penicillin and 0.1 mg/ml streptomycin (Wisent Bioproducts, QC, Canada), 15% Horse

Serum (Gibco, Cat#200-6050), 5% FBS (Wisent Bioproducts, 080-350) and 10 mM HEPES.

To study the role of Cdh11 in the progress of these tumors, cell lines were harvested for protein

and RNA and analyzed for Cdh11 expression. Additionally, Cdh11 in these cell lines was

Page 51: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

38

knocked down using Invitrogen stealth siRNA and analyzed for differences in apoptotic protein

expression and expression of different invasion markers.

2.9 RNA Isolation

Total RNA was isolated from cell lines after knockdown of Cdh11 with Invitrogen stealth

siRNA. RNA was extracted from cell lines through the use of TRIzol Reagent (Invitrogen,

Cat#15596-026), through the manufacturer‟s instructions. RNA was redissolved in 20 µl ddH20

and the concentration found using a Nanodrop-1000 spectrophotometer (Thermo Scientific). For

cDNA synthesis, 1 µg total of RNA was reverse transcribed using random primers (Fermentas),

and Superscript II Reverse Transcriptase (Invitrogen). Gene expression analysis was done by

PCR using a RoboCycler Gradient 96 thermal cycler (Stratagene). Conditions for PCR was the

same for all the primers used, and is as follows: 940C, 2 min, 1 cycle, [94

0C, 30s, 60

0C, 30s,

680C, 30s] 35 cycles, 68

0C, 10 min, 1 cycle, and 4

0C, cool block. The reaction mixture included

2.5 µl KOD PCR 10X buffer (Novagen), 2.5 µl 2 mM dNTPs, 1 ul 25 mM MgSO4, 0.5 µl each

of forward and reverse primers, 1.0 µl template, 0.5 µl KOD hot start DNA polymerase

(Novagen), up to 25 µl total volume. Primers used were all designed according to the mouse

genome sequence. They are listed below with expected product sizes in Table 2.2.

Page 52: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

39

Table 2 Primers used with Expected Product Sizes.

Gene Primer Sequence Expected Size

(bp)

Cdh11 5‟-AGGAGTATATGCCCGACGTG-3‟

5‟TCGTCCACATCCACACTGTT-3‟ 504

Twist 5‟-ACGACAGCCTGAGCAACAG-3‟ 5‟-

CATCTTGGAGTCCAGCTCGT-3‟ 488

SNai2 (Slug) 5‟-AACATTTCAACGCCTCCAAG-3‟ 5‟-

CAGTGAGGGCAAGAGAAAGG-3‟ 631

Ctnnb1 (B-catenin) 5‟-CAAGATGATGGTGTGCCAAG-3‟ 5‟-

CTGCACAAACAATGGAATGG-3‟ 502

Lef1 5‟-CTCATCACCTACAGCGACGA-3‟ 5‟-

CGTGCACTCAGCTACGACAT-3‟ 498

RhoA 5‟-AAGGACCAGTTCCCAGAGGT-3‟ 5‟-

ACAAGATGAGGCACCCAGAC-3‟ 582

MMP2 5‟-ATCTACTTGCTGGACATCAGGG-3‟

5‟- TGGCTCGAAATTCACAAGGTCC-3‟ 493

MMP9 5‟-GAAGGCAAACCCTGTGTGGT-3‟ 5‟-

GGCTTAGAGCCACGACCATA-3‟ 497

2.10 Protein Isolation

TAg-RB cell lines were collected and resuspended in cold TNE buffer (2% NP-40, 20 mM Tris

pH8.0, 150 mM NaCl, 5 mM EDTA, 2 mM NaN3, 0.1 mM phenylmethylsulfonyl fluoride, 2

µg/ml leupeptin, and 20 µg/ml aprotinin). Samples were incubated for 1 h at 40C on a rotor

mixer. After centrifugation at 12,600 g for 20 min, supernatants were collected and protein

concentration was determined using the BioRad Protein Assay (BioRad, Cat#500-0006), in a

Beckman Coulter DU640B spectrophotometer. Proteins (50 µg) were separated using SDS-

Page 53: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

40

PAGE, via a 4-20% Tris-Glycine gradient gel (Lonza, Cat#58511) at 125V. Following

separation, proteins were transferred to a PVDF membrane (BioRad, Cat#162-0177) at 100V for

1 h. Membranes were blocked using 5% Blotto (BioRad) in TBS overnight at 40C. The

following day, membranes were incubated with the proper primary antibody in TBS, 0.05%

Tween-20 for 0.5 h. The respective dilutions for all primary antibodies used are as follows:

Cdh11 (1:750, ZYMED), caspase-3 (1:200, Promega), caspase-8 (1:1000, Abnova), caspase-9

(1:1000, Cell Signalling Technology), β-catenin (1:1000, BD Biosciences) and β-tubulin

(1:1000, Sigma-Aldrich, TO198). Primary antibodies were followed by 3X 10 min washes in

TBS, 0.05% Tween-20, and incubated for 1 h in the respective secondary antibody. The

dilutions for the antibodies are as follows: anti-mouse-HRP (1:10000, Invitrogen, Cat#G21040)

and anti-rabbit -HRP (1:10000, Santa Cruz, sc-2004). This incubation was followed by 3X 10

min washes in TBS, 0.05% Tween-20. HyGLO chemiluminescence Detection Reagent

(Denville) and HyBlot autoradiography film (Denville) was used to detect the proteins.

2.11 Stealth RNAi

To analyze apoptotic protein expression and changes in invasion markers, Cdh11 was knocked

down in TAg-RB cell line of choice, T+539b (20wks). Stealth RNAi (siRNA) was provided by

Invitrogen (Cat# 1320003). Control siRNA was the GL-2 vector (Qiagen). T+539b cells were

transfected with the siRNA targeting mouse Cdh11, at time of plating in triplicate, in media

without the addition of penicillin and streptomycin. The procedure included transfection of 125

pmol of each siRNA oligo in Lipofectamine 2000 (Invitrogen), in a total of 2 ml plating medium.

Cells were left for time periods of 24 h, 48 h, 72 h, 5 d, 7 d or 10 d. Knockdown was confirmed

by Western blot analysis of Cdh11 expression.

Page 54: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

41

Chapter 3

Results

3.1 Cadherin-11 acts a tumor suppressor in TAg-RB tumors through

promotion of apoptosis

3.1.1 Active Apoptotic Pathway in TAg-RB Mice

Immunohistochemical analysis was performed on 5 Cdh11-/-

;TAg+/-

and 5 Cdh11+/+

;TAg+/-

mice

sacrificed at PND84, to analyze the apoptotic activity within TAg-RB tumors. Mice were

sacrificed at this timepoint as tumors at this time are well defined and have a distinct phenotype

and differences in tumor volume are observed (Marchong et al, submitted). Slides were stained

with antibodies to both pro-apoptotic and anti-apoptotic proteins and within both the extrinsic

and intrinsic pathways of apoptosis. The methods and materials section contains a full list of all

antibodies used in this experiment (Table 2.1). Positive staining occurred with pro-apoptotic

proteins, BAX, TRAIL, caspase-8, caspase-9, caspase-3, and anti-apoptotic proteins, Bcl-xL,

FLIP (Figure 3.1). This list includes proteins involved in both the intrinsic and extrinsic

pathways. Activation of the pro-apoptotic protein BID (tBID) was observed, and was the

common meeting point for the two pathways in our model.

Page 55: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

42

Figure 3.1 Expression of Apoptotic Proteins within TAg-RB Tumors.

Immunohistochemistry was performed on TAg-RB tumors wildtype for Cdh11 at PND84. Analysis of a variety of

pro and anti-apoptotic proteins was done to discover the acting apoptotic pathway (x100 and x400 magnification).

Positive staining was shown with Caspase-3, 8, 9, TRAIL, BAX, Bcl-xl, FLIP and BID. Positive staining was

confirmed with double staining with both DAPI and additional cell death markers.

Page 56: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

43

3.1.2 Quantification of Apoptotic Protein Expression in TAg-RB Tumors

The expression of the apoptotic proteins previously mentioned was quantified to assess the

difference in apoptotic activity between Cdh11+/+

;TAg+/-

and Cdh11-/-

;TAg+/-

tumors at PND84.

Initial data led to the idea that apoptosis may be affected by loss of Cdh11 in TAg-RB mice

(Section 1.9) (Marchong, Yurkowski, et al, PLOs Genetics (in press)). Therefore, detailed

analysis and quantification of apoptotic activity within these tumors was pursued in order to find

a mechanism of suppressive function for Cdh11. For this experiment, mice were sacrificed at

PND84 and the eyes paraffin fixed and horizontally sectioned onto slides. One eye from each of

the 5 Cdh11-/-

;TAg+/-

and 5 Cdh11+/+

;TAg+/-

mice was used in analysis. Upon sectioning, an

average of ~60 slides is obtained per mouse eye, and for this experiment, every 10th

slide was

chosen to get a good representative sample of the entire tumor. This equaled to using 6 slides

per eye (mouse) in analysis. Five proteins that had previously been described to be involved in

the TAg-RB apoptotic pathway (Figure 3.2) were used in the quantification and included BAX,

TRAIL, caspase-3, caspase-8, and caspase-9. Slides are produced with 6-7 0.5µm sections on

each slide and a total of 36 sections were used in the analysis. For each section, positively

stained cells were manually counted under the microscope. A total number of positively stained

cells were obtained for the entire tumor and then an average per section was derived from

dividing by the 36 sections used in analysis. This analysis yielded a true number of apoptotic

cells from anywhere within the tumor. Upon quantification, a very significant difference in

apoptotic activity was observed. In comparison, Cdh11-/-

;TAg+/-

mice were revealed to have

anywhere from 5 to 10 times less apoptotic activity happening within the tumors than

Cdh11+/+

;TAg+/-

mice. This was true for every cell death marker assessed (caspase-3, p=0.014;

caspase-8, p=0.029; caspase-9, p=0.014; TRAIL, p=0.008; and BAX, p=0.029) (Figure 3.3).

This gave very strong evidence that Cdh11 is promoting apoptosis within TAg-RB tumors.

Page 57: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

44

Figure 3.2 Acting Apoptotic Pathway in Retinal Tumors of TAg-RB Mice.

Immunohistochemical staining implicated extrinsic proteins (A) and intrinsic proteins (B) in the apoptotic pathway

in TAg-RB mice. Cross talk occurs through activation of BID and both pathways lead to activation of caspase-3.

Page 58: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

45

Figure 3.3 Differences in Apoptotic Protein Expression in TAg-RB Tumors at PND84.

Cells were manually counted and an average number of apoptotic cells per section were calculated in order to

quantify expression of five apoptotic proteins (caspase 3, 8, 9, TRAIL and Bax) active in TAg-RB tumors at

PND84. When averaged, results revealed between five to ten times less apoptotic activity in TAg-RB tumors null

for Cdh11 (Student‟s T-test: p values = 0.014, 0.029, 0.014, 0.008, and 0.029, respectively).

3.1.3 Analysis of Cadherin-11 in TAg-RB Derived Cell Lines

To support the in vivo data from the TAg-RB tumors, in vitro work was performed with cell lines

previously described (Section 2.8), derived from primary TAg-RB tumors. Various cell lines

were harvested to isolate protein and Western Blot analysis was performed to determine Cdh11

expression. Six candidate cell lines were chosen to analyze for Cdh11 expression, including

Page 59: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

46

T+532, T+539b, T+572a, T+794a, T+818a, and T+827b. Protein analysis revealed varied Cdh11

expression, supporting evidence that TAg-RB tumors parallel human retinoblastoma genomic

and expression changes (Marchong, Yurkowski, et al, PLOs Genetics (in press), Dimaras et al.,

2008), as 4 of 6 or 67% of cell lines showed a decrease, little or no Cdh11 expression (Figure

3.4).

Figure 3.4 Cdh11 Protein Expression in TAg-RB Derived Cell Lines.

Cdh11 protein expression was analyzed via Western blot using six different cell lines all derived from primary TAg-

RB tumors. Expression was varied in all six cell lines ranging from abundant Cdh11 expression, to decreased or

little Cdh11 expression, paralleling human retinoblastoma cell lines.

3.1.4 Apoptotic Protein Expression in a TAg-RB Derived Cell Line

In order to support the idea that the suppressive mechanism of Cdh11 in TAg-RB tumors

involved the promotion of apoptosis, cell line T+539b, derived from primary TAg-RB tumors

(Section 2.10) was chosen as a candidate line to use in knockdown experiments, on the basis that

it showed abundant Cdh11 expression via Western blot (Figure 3.4). This experiment was done

in order to establish and confirm a direct effect of Cdh11 on apoptosis, by directly inhibiting

Page 60: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

47

Cdh11 by siRNA (Sigma, MSS202865-87). After 10 days treatment, Western blot analysis was

done to look the change in pro-apoptotic protein, caspase-3 expression. Effective knockdown of

Cdh11 was assessed by Western Blot and showed significant knockdown by siRNA, by 67.8%

(siRNA MSS202867) (Figure 3.5). Accordingly, when comparing the expression of these

proteins to the siRNA control lane, caspase-3 expression was also decreased by 53.1%. This

data indicated Cdh11 had an effect on apoptosis in vitro, and showed support for the

observations seen in vivo (Figure 3.3), that Cdh11 is promoting apoptosis within TAg-RB

tumors.

Figure 3.5 Cdh11 Knockdown Leads to Decreased Caspase-3 Expression.

Cdh11 was knocked down using stealth siRNA (MSS202867, Invitrogen) in a cell line derived from TAg-RB

tumors, T +539. Upon knockdown of Cdh11, caspase-3 expression was significantly decreased compared to

control.

Page 61: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

48

3.1.5 Cdh11 and Proliferation in TAg-RB Tumors

Evidence supports the notion that Cdh11 is promoting apoptosis within TAg-RB tumors.

However, development of tumors is often distinguished as a fine balance between apoptosis and

proliferation, and so experiments were pursued to determine the effect of loss of Cdh11 on

proliferation within TAg-RB tumors. Therefore, these experiments were necessary to specify the

role of Cdh11 in the progression of these tumors. Preliminary evidence suggested Cdh11 was

not affecting proliferation in TAg-RB tumors through immunohistochemistry using PCNA as a

marker for proliferation (Marchong, Yurkowski, et al, PLOs Genetics (in press)), and so to

expand and confirm these initial results, quantification with BrdU was performed. Mice were

sacrificed at PND84 (3 months), the same timepoint used for apoptotic protein expression

experiments. For these experiments 6 Cdh11-/-

;TAg+/-

and 6 Cdh11+/+

;TAg+/-

mice were used in

the analysis. Four hours before sacrificing these mice, an IP injection of BrdU was given. Eyes

were sectioned as previously described and every 10th

slide was chosen in analysis. This gave a

total of 24 sections measured and analyzed per mouse. Immunohistochemistry was used to look

at proliferating cells by staining for BrdU. The area of the positively stained cells (examples

outlined in blue), measured in pixels, was taken and used as a percentage of tumor area (outlined

in yellow), as described by previous methods used in the lab (Figure 3.6) (Dimaras et al., 2009).

Upon quantification, no significant difference was seen in the amount of BrdU positive cells

between the two genotypes (Figure 3.7), suggesting that Cdh11 was not affecting proliferation

within the tumors. By these results, Cdh11 appears to be affecting tumor growth and exerting a

suppressive function specifically through affecting apoptotic activity.

Page 62: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

49

Figure 3.6 Quantifying the Percentage of BrdU Proliferating Cells as a Function of Tumor Area.

A) Eyes were horizontally sectioned onto slides and every 10th

slide was chosen to use in analysis. A total of 24

sections were analyzed. B) Selected sections were stained for BrdU using DAB for visualization in bright field.

Area of proliferating cells was found by using Image J software. Images were analyzed in an 8-bit grey scale, and

the area of BrdU positive cells (red) was found by measuring in pixels. Area of proliferating cells was taken as a

percentage of total tumor area (in yellow).

Page 63: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

50

Figure 3.7 Average Area of Proliferating Cells Per Tumor Area at PND84.

6 Cdh11+/+

;TAg+/-

and 6 Cdh11-/-

;TAg+/-

PND84 eyes were stained for BrdU. Proliferation was analyzed by

measuring BrdU positive cells (in pixels) as a percentage of tumor area. An average was found for each genotype.

The percentage of BrdU positive cells as a function of tumor area is shown with no significant difference in the

amount of staining for the proliferation marker seen between the two genotypes (p value = 0.121).

Page 64: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

51

3.2 Loss of Cdh11 in TAg-RB Tumors Increases Invasion Potential

3.2.1 Knockdown of Cdh11 in a TAg-RB Cell Line Leads to an Increase of EMT

Markers and Metalloproteases

It was confirmed by the above experiments, that Cdh11 is acting as a tumor suppressor in TAg-

RB tumors through promotion of apoptosis, and that when Cdh11 is lost, tumors grow at a faster

rate due primarily to decreased cell death. We then questioned if the loss of Cdh11 would affect

the invasion potential of these tumors, as when Cdh11 was lost, if these tumors would take on a

more aggressive role. It is well established that altered expression of cadherin molecules affect

tumorigenesis and influence invasion. Often the loss of cell-cell adhesion and the alteration of

signaling transduction pathways lead to malignant transformation as well as enhanced migration,

invasion and metastasis (Cavallaro and Christofori, 2004; Yilmaz and Christofori, 2009). To

examine if Cdh11 would have a similar role in retinoblastoma, RT-PCR was performed to

analyze mRNA expression levels in vitro using candidate TAg-RB cell lines to observe the

mRNA expression of various EMT markers, and MMPs upon knockdown of Cdh11 using

siRNA. The cell line T+539b, positive for Cdh11 as described above (Figure 3.4, Section 2.8),

was chosen for these experiments. Cdh11 was knocked down in line T+539b and at a timepoint

of 10 days, RNA was isolated and RT-PCR was done in order to analyze the levels of the

different invasion markers. Cdh11 knockdown was confirmed via Western blot as described

above (Figure 3.5). Two different EMT markers were analyzed; TWIST and the SNAi2, the

mouse homolog of Slug. End-point RT-PCR revealed both of these markers had low expression

in line T+539b and when treated with the control siRNA, but were upregulated after knockdown

with siRNA (Figure 3.8A). Both these molecules have been widely published in the literature to

increase during EMT and essential to metastasis formation and invasion (Kurrey et al., 2005;

Yang et al., 2004). End-point RT-PCR for MMPs also revealed an up-regulation in MMP-2 and

MMP-9 upon knockdown of Cdh11 in T+539b (Figure 3.8B). A recent study saw up-regulation

Page 65: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

52

of both MMP-2 and MMP-9 in human samples of retinoblastoma, in a higher percentage of

invasive tumors compared to non-invasive tumors (Adithi et al., 2007). Our results demonstrate

up-regulation of two MMPs, and two EMT markers, indicating an increase in invasion potential

of TAg-RB tumors, upon the loss of Cdh11.

Figure 3.8 Knockdown of Cdh11 Leads to Increased mRNA Expression of EMT and Invasion

Markers.

Cdh11 was knocked down with siRNA (Invitrogen) and RT-PCR analysis was done to look at mRNA expression

levels of invasion markers after 10 days treatment. Both EMT markers analyzed, TWIST and SNAi2 showed

upregulation after knockdown. Additionally, two MMPs analyzed, MMP-2 and MMP-9 showed upregulation after

Cdh11 knockdown.

Page 66: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

53

3.2.2 β-catenin mRNA and Protein Levels Increase Upon Knockdown of Cdh11

The loss of Cdh11 was seen to lead to an up-regulation of certain invasion markers in vitro. A

question remained to how Cdh11 loss could be linked to both this increase of invasion markers

and promotion of apoptosis. β-catenin is the primary protein that interacts with Cdh11. It is well

established to have dual roles within the cell, in adhesion and in a variety of downstream

signaling events, most notably in Wnt signaling. The loss of Cdh11 cell-cell adhesion could

quite possibly affect the levels of free ϐ-catenin within the cell, and lead to altered downstream

signaling. Therefore, to us to look at ϐ-catenin levels upon the loss of Cdh11, Cdh11 was

knocked down in cell line T+539b, and the mRNA and protein levels of ϐ-catenin were analyzed,

by RT-PCR and Western blot, respectively. At 10 days with siRNA, mRNA and protein samples

showed increased expression of ϐ-catenin and the knockdown of Cdh11 (Figure 3.9A, B). Many

downstream effects could result from this increase and would warrant further study.

Page 67: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

54

Figure 3.9 Cdh11 Knockdown Leads to Upregulation of ϐ-catenin mRNA and Protein Expression

Levels.

A) Cdh11 was knocked down using three different stealth siRNAs (Invitrogen) targeted to Cdh11, in TAg-RB

derived cell line T+539. Upon knockdown of Cdh11 (MSS202865 – siRNA #1, MSS202866 – siRNA #2,

MSS202867 – siRNA #3) expression levels of ϐ-catenin analyzed via Western blot were increased in 2 out of 3

Cdh11 siRNA treated samples. B) Cdh11 was knocked down with siRNA (Invitrogen). Following knockdown, RT-

PCR performed showed an increase in ϐ-catenin mRNA levels in TAg-RB cell line T+539.

Page 68: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

55

Chapter 4

Discussion

4.1 Cdh11 as a Tumor Suppressor in Retinoblastoma Progression

Earlier studies have implicated involvement of Cdh11 in Retinoblastoma progression. Early

cytogenetic studies showed recurrent chromosomal abnormalities in human retinoblastoma, and

chromosome 16 was the most common genomic loss [reviewed in(Potluri et al., 1986)]. More

recent CGH studies corroborate these findings, and revealed that 32% (58/179) of tumors

showed loss of either the entire chromosome 16 or 16q, with all but 7 tumors showing a minimal

region of loss at 16q22 [reviewed in(Corson and Gallie, 2007)]. Marchong et al., showed copy

number loss for Cdh11 in retinoblastoma based on LOH and QM-PCR techniques. In addition,

some tumors analyzed showed exonic deletion of Cdh11 and three tumors showed potential

intragenic mutations, although no further analysis was done past this point. Copy number loss

was confirmed by another recent study in which allelic loss of Cdh11 was observed in 45% of 20

tumors (Bowles et al., 2007). Expression analysis of Cdh11 revealed loss or decrease in 91% of

tumors and cell lines. The same study also revealed decreased expression in advanced TAg-RB

tumors (Marchong et al., 2004). While early tumors of TAg-RB mice showed high expression of

Cdh11, tumors by 21 weeks showed decreased Cdh11 expression, and 38% of TAg-RB

advanced tumors showed complete loss/decrease of Cdh11 mRNA expression. Supporting the

link of loss of Cdh11 associating with malignancy in retinoblastoma, levels of expression in

adjacent retinoma were equivalent to normal retina (Dimaras et al., 2008). The observation of

loss of Cdh11 expression in late stage tumors supports the hypothesis that Cdh11 loss is a late

event based on frequency and correlation with other genomic changes (Bowles et al., 2007).

Page 69: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

56

CDH13 was also listed as a potential tumor suppressor as the second most frequently deleted

marker was in intron 2 of the CDH13 gene. However, it was ruled out as a candidate gene as it

showed normal levels of expression in all retinoblastoma primary tumors and cell lines. Another

study analyzed the 16q region through LOH and CGH techniques, studying 58 retinoblastoma

tumors (Gratias et al., 2007), and defined a minimal region of loss at 16q24. They analyzed

Cdh13 and found no mutations, suggesting no involvement in retinoblastoma, confirming the

previous findings (Marchong et al., 2004).

As a candidate gene in retinoblastoma progression, preliminary assessment of the tumor

suppressor role of Cdh11 was done in TAg-RB mice. When TAg-RB+/-

mice were crossed with

Cdh11+/+

and Cdh11-/-

mice to examine tumorigenesis, tumor volumes were calculated from a

single cell at post-natal day 8 (PND8), to a large tumor at PND84. Volumes were larger overall

in Cdh11-/-

mice compared to those with normal Cdh11 alleles and tumor growth was faster

when quantified between PND28 and PND84 (Marchong, Yurkowski, et al, PLOs Genetics (in

press)). This suggested Cdh11 was acting as a tumor suppressor. Tumor growth can be defined

as a fine balance between cell proliferation and cell death and therefore, both possibilities were

considered. Preliminary experiments showed no statistical differences in PCNA, and did show a

statistical difference in caspase-3 staining when extrapolated to the entire tumor. These studies

required further work to 1) confirm the role of Cdh11 in Retinoblastoma as a tumor suppressor

and 2) to define the mechanism by which this was occurring. The work presented in the present

study achieved both of these goals.

4.2 Cdh11 Acts as a Tumor Suppressor by Promoting Apoptosis

The current study focused on defining the mechanism of suppressive action of Cdh11 in

retinoblastoma and to further identify its suppressive role in late progression of the disease.

Page 70: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

57

Further information was necessary to confirm that this suppressive role was tied to apoptotic

activity. Every experiment in the present study used the TAg-RB murine model. These mice

develop tumors resembling human retinoblastomas and past studies in the lab have shown that

expression changes in this model parallel human retinoblastoma (Dimaras et al., 2008; Marchong

et al., 2004). Even cell lines derived from primary TAg-RB tumors parallel expression changes

in vitro, with 67% of lines tested showing decreased or no expression of Cdh11 (this study).

This model is also the most widely used model for pre-clinical studies, making it an ideal model

for our purposes, in which crossing it with Cdh11 knockout mice allowed us to study the effect

of Cdh11 on late stage tumor development and progression.

Our analysis began with mapping out the acting apoptotic pathway in TAg-RB mice. This was

done by analyzing the presence of various pro and anti-apoptotic factors, verified by

immunohistochemical positive staining. Our first inclination was to assume a purely extrinsic

mechanism, due to the inactivation of p53 in this model. In contrast, we saw proteins activated

involved in both the intrinsic mitochondrial apoptotic pathway and the extrinsic death receptor

activated pathway. Apoptosis is initiated via the extrinsic pathway, through the initiation of

TRAIL, a TNF death receptor superfamily member. TRAIL mediated induction of apoptosis has

been shown to be preferential in a wide variety of tumor cells, but not in normal cells, in vitro

(Falschlehner et al., 2009). Limited studies looking at apoptosis have been done in human

retinoblastoma cells, which show a varied pattern of apoptosis. Both p53 dependent and

independent mechanisms have been reported in different tumors, with the majority of cases

leading to activation of caspase-3 (Sitorus et al., 2009). We speculate the same idea is paralleled

in TAg-RB tumors, where we also see activation of both pathways. In TAg-RB mice, we

observed merging of the two pathways, through caspase-8 cleavage and activation of tBID.

Page 71: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

58

Since tumor “growth” results from an imbalance between cell death and proliferation, we

examined in detail the contributions of cell proliferation (Figure 3.7) and cell death (Figure 3.3)

in TAg-RB mice with normal Cdh11 alleles vs. mice with mutated Cdh11 alleles. Our data

indicate that when Cdh11 is lost, cell death is deficient by five to ten times the amount, while

proliferation remains unchanged; suggesting that the tumor suppressor function of Cdh11 is

mediated through apoptosis rather than cell proliferation. This idea is further supported by our in

vitro data, where we showed a direct link between apoptosis and Cdh11, as there was a

significant decrease in caspase-3 expression of 67.8% in Cdh11 knockdown experiments (Figure

3.5), in lines derived from primary TAg-RB tumors. To examine proliferation, BrdU was used

as a marker in vivo, assayed by immunohistochemistry to look at differences between genotypes.

Quantification revealed no significant differences between TAg-RB mice with normal Cdh11

alleles, and mice null for Cdh11. These results further confirmed our hypothesis that Cdh11 was

acting as a tumor suppressor through promoting apoptosis, and not through inhibition of cell

division. Furthermore, these results support and coincide with a previous report that yielded

similar results in TAg-RB mice when proliferation and apoptosis were examined using PCNA

and caspase-3 as markers, respectively (Marchong, Yurkowski, et al, PLOs Genetics (in press)).

However, we must remember that while apoptosis is a linear process, proliferation is

exponential. Therefore, experiments to quantitate proliferation over time might reveal

significant effect of loss of Cdh11, as suggested by a trend in our studies of markers of

proliferating cells at isolated time points.

Cadherin molecules are well known to manipulate various cellular functions, including

apoptosis. The majority of studies have focused on N-cadherin, which has been seen to be

upregulated during tumorigenesis in many different tumor types (Yilmaz and Christofori, 2009).

Studies have shown that N-cadherin mediated cell adhesion actually prevents apoptosis in

Page 72: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

59

different cells types, and loss of this cell-cell adhesion via N-cadherin results in an increase of

apoptotic signaling and cell death (Erez et al., 2004; Hermiston and Gordon, 1995; Peluso, 1997;

Peluso et al., 1996). However, E-cadherin, which is often lost during tumorigenesis in a variety

of tumor types, has been seen to have a similar effect on apoptosis as presented in this study. A

recent study showed that E-cadherin restoration in human melanoma cell lines, led to increased

levels of activated caspase-3 and caspase-8, and sensitization of apoptosis (Kippenberger et al.,

2006). Current literature does not reveal studies directly linking Cdh11 to apoptotic activity.

My thesis then presents a novel function for Cdh11, in promoting apoptosis. We speculate that

both cadherin type and tissue type may lead to differential roles for cadherins in apoptotic

activity. Interestingly, most studies have implicated a role for N-cadherin in preventing

apoptosis, a cadherin molecule usually upregulated in cancer. Kippenberger et al, implicated E-

cadherin in promoting apoptosis, a molecule typically lost in tumorigenesis. Cdh11 has been

seen to be both lost and gained in the progression of different cancers, and in retinoblastoma, this

thesis shows that it acts to promote apoptosis, leading to the Cdh11 tumor suppressor role in this

cancer. Further studies done in cell lines and tissues of different origin then the retina could

clear up if the role of Cdh11 is context dependent.

4.3 Loss of Cdh11 and Invasion

After defining the mechanism of suppressive action of Cdh11 in the progression of

retinoblastoma, we were curious to further define its role in tumorigenesis. Experiments were

performed to analyze Cdh11 in invasion of retinoblastoma, and determine if this loss would lead

to more aggressive tumors. Our results indicate an upregulation of various invasion markers,

Twist and SNai2, as well as MMPs, MMP-2 and MMP-9. Often the loss of cell-cell adhesion

and the alteration of signaling transduction pathways lead to malignant transformation as well as

enhanced migration, invasion and metastasis (Cavallaro and Christofori, 2004; Yilmaz and

Page 73: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

60

Christofori, 2009). For example, downregulation of E-cadherin has been observed in invasive

and metastatic forms of many cancers, such as breast and gastric carcinomas (Margineanu et al.,

2008). Furthermore, recent data has correlated this change in phenotype to an upregulation in

MMPs (Llorens et al., 1998; Nawrocki-Raby et al., 2003). An increase in MMPs has been seen

in retinoblastoma, where expression of MMP-2 and MMP-9 were significantly correlated to

invasive tumors (Adithi et al., 2007). In addition, more invasive phenotypes correlate with an

upregulation of EMT markers, a hallmark in malignant transformation and metastasis formation

(Karreth and Tuveson, 2004), which include a wide variety of molecules from transcription

factors like Twist and Slug to growth factors and candidates in downstream effector pathways

(Yang et al., 2004; Yilmaz and Christofori, 2009).

A recent study supports our hypothesis that Cdh11 acts as a tumor suppressor in retinoblastoma

progression and that loss of Cdh11 leads to increased invasion potential (Laurie et al., 2009).

Cell lines were generated from a Chx10-Cre;RbLox/+;p107-/-;p53Lox/Lox murine model of

retinoblastoma. In culture, two of these cell lines, SJmRBL-3 and SJmRBL-8, underwent

genetic changes affecting cell adhesion. Gene expression microarrays showed changes in Cdh11

and N-cadherin mRNA and protein expression. Xenografts of both newborn rats and SCID mice

revealed highly invasive tumors, invading the optic nerve, anterior chamber, choroid and

subretinal space upon transplantation of SJmRBL-3 and SJmRBL-8. Further experiments

injected both Y79 and SJmRBL-8 cells, into adult SCID mice. These cells had been transfected

with Cdh11/N-cadherin or a control vector. Only eyes in xenografts with the control vector

revealed extensive optic nerve invasion. Either cadherin was sufficient to cause an increased

cell-adhesion phenotype resulting in decreased invasion properties of transfected tumor cells.

This established a functional significance in murine retinoblastoma invasion, mediated by

cadherin cell-adhesion. Furthermore, FISH performed on human retinoblastoma whole eye

Page 74: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

61

sections showed selective loss of Cdh11 in some samples with invasion of the optic nerve and

choroid.

4.4 Cdh11 in Cancer Progression

Cdh11 has an interesting role in cancer progression, as it has been seen to be both down

regulated and up regulated in different cancers. In retinoblastoma, Cdh11 acts as a tumor

suppressor, as implicated by earlier studies (Marchong et al., 2004; Marchong, Yurkowski, et al,

PLOs Genetics (in press)), and further shown by evidence presented in the current study. Here,

we have provided data showing that this effect is mediated through the promotion of apoptosis.

Additionally, the loss of Cdh11 correlates to up-regulation of EMT markers and MMPs,

increasing the invasion potential of these tumors. Studies done in osteosarcoma have shown a

similar tumor suppressor role for Cdh11. Multiple studies witnessed decreased expression of

Cdh11 in cell lines of primary tumors and even lower expression in metastatic cell lines, where

the loss of Cdh11 was even suggested as a prognostic marker (Nakajima et al., 2008; Zou et al.,

2008). Decreased Cdh11 expression has also been seen in astrocytomas and a subset of colon

cancers (Braungart et al., 1999; Zhou and Skalli, 2000). These types of changes in Cdh11

expression are reminiscent of changes seen in E-cadherin expression in a variety of cancers,

where the loss of E-cadherin is often correlated to aggressive and invasive cancer (Prasad et al.,

2009; von Burstin et al., 2009).

Upregulation of Cdh11 is also seen in certain cancers, such as breast and prostate cancers,

including invasive and metastatic forms (Chu et al., 2008; Nagaraja et al., 2006; Sarrio et al.,

2008). In this context, the role of Cdh11 is similar to the role of N-cadherin in cancer, which is

upregulated in a variety of cancers, especially those undergoing EMT (Gravdal et al., 2007; Nagi

et al., 2005). This could suggest binding partners for Cdh11 similar to N-cadherin in these

Page 75: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

62

cancers. Additionally it is often correlated with a downregulation of E-cadherin (Yilmaz and

Christofori, 2009). Interestingly, in retinoblastoma, we observe decreased expression of Cdh11

(Marchong et al., 2004) and in invasive retinoblastoma, an increased expression of N-cadherin

(Mohan et al., 2007).

These observations lead us to speculate why Cdh11 has such opposing roles in cancer

progression, having either a suppressive or oncogenic function. One possible explanation is that

the role of Cdh11 depends on tissue type and microenvironment, two ideas now heavily studied

in tumorigenesis. It appears that Cdh11 has more of a suppressive role in tumors that develop in

looser attached tissues, while an oncogenic role emerges in tighter attached tissues. This idea is

supported by a study in which cell density was observed to affect the expression of Cdh11 in

MDA-MB-231 cells, a mesenchymal-like breast cancer cell line. In this study, the expression of

Cdh11 depended on the density of cells plated. At increased cell densities, Cdh11 expression

was increased in comparison to low density (Farina et al., 2009).

Tumor microenvironment has emerged to become a hugely discussed topic in the field of

tumorigenesis. The tumor microenvironment is a completely unique environment that emerges

in the course of tumor progression as a result of interactions with the host. It is created and

shaped by the tumor which orchestrates molecular and cellular events taking place in

surrounding tissues. This leads to the implication that unique differences in tumor

microenvironment could be responsible for regulation of expression of various molecules, like

cadherins, leading to altered signaling pathways, cell-cell adhesion and the extracellular matrix.

For instance, a recent study showed that stable ectopic expression of interleukin 6 (IL-6) in

MCF-7 breast adenocarcinoma cells actually led to an EMT phenotype, with the downregulation

of E-cadherin, upregulation of N-cadherin, and an upregulation of EMT markers Snail and Twist

Page 76: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

63

(Sullivan et al., 2009). Other studies demonstrated that collagen type I, produced by pancreatic

tumors, influences the invasiveness of pancreatic carcinoma cells (Koenig et al., 2006; Shintani

et al., 2006). One study focused on the influence collagen type I had on disrupting the E-

cadherin adhesion complex within these cells which led to increased cell proliferation (Koenig et

al., 2006), and the other observed a response to collagen type I through signaling of JNK1

leading to an upregulation of N-cadherin and increased motility (Shintani et al., 2006). Other

factors have been implicated to have similar roles as well, including TGFϐ, reducing E-cadherin

expression in colon cancer cells (Bates and Mercurio, 2005). The implication of these and

additional microenvironmental factors being involved in regulation of cadherin molecules points

to multiple mechanisms impeding cell-cell adhesion in tumorigenesis.

4.5 Future Directions

4.5.1 The Mechanism of Influence of Cdh11 on Apoptosis and Invasion Potential

Cadherin levels have been heavily discussed in the literature as having numerous cellular effects

in both healthy tissue, developing tissue and in tumorigenesis. However, the direct link of

Cdh11 to apoptotic activity is rather novel. Generally, studies have looked at N-cadherin levels

and linked the loss of cadherin mediated cell-cell adhesion to an increase in apoptotic activity

(Erez et al., 2004). However, loss of E-cadherin in cancer has been shown to have the opposite

effect, and restoration of E-cadherin levels actually increased the levels of activated caspase-3

and caspase-8, sensitizing cells to apoptosis (Kippenberger et al., 2006). This study showed a

significant decrease of apoptosis upon the loss of Cdh11.

Cadherin molecules have numerous roles and huge impact in tissues and disease largely due to

their catenin binding partners, linking them to a wide variety of intracellular signaling pathways,

as well as their attachment to the cytoskeleton and their ability to bind to neighbouring cells and

Page 77: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

64

interact with other cell surface molecules. These characteristics enable multiple pathways, which

may include cross-talk, to be severely altered upon loss or gain of these molecules. Additionally,

micro-environmental factors add in another level of endless possibilities to the effects of

cadherins on cellular processes. These are reasons why it would be interesting to take the next

step in evaluating how the loss of Cdh11 cell-cell adhesion actually influences apoptosis and

invasion potential in retinoblastoma. Because Cdh11 binds to various catenin molecules in order

to assemble the AJs, various pathways could be altered that cumulatively lead to our

observations. One idea is that the loss of Cdh11 would increase ϐ-catenin levels, leading to

increased Wnt signaling, transcription of target genes, and decreased apoptosis or even increased

invasion potential. Wnt proteins have been linked to apoptosis, through both ϐ-catenin

dependent and independent mechanisms, inhibiting it through downstream signaling and by an

increase in Bcl-2 expression (Almeida et al., 2005). My preliminary results looking at

expression of ϐ-catenin after Cdh11 knockdown indeed show an increase in both protein and

mRNA levels. Whether this would affect Wnt signaling or not needs to be determined. Studies

have shown increased level of ϐ-catenin does not always indicate activation of Wnt, as shown

recently in a study in osteosarcoma (Cai et al., 2009). Furthermore, even if Wnt signaling was

activated due to increased ϐ-catenin, we have to keep in mind a recent study showing that Wnt

signaling, while enhancing tumorigenesis in multiple cancers, has tumor suppressive properties

in retinoblastoma (Tell et al., 2006). In addition, it is possible other cellular pathways are

affected by the loss of Cdh11, and in turn, may be linked to the increase we see in expression of

β-catenin. Cellular pathways such as the PI3K/Akt have been linked to β-catenin stabilization

(Fang et al., 2007), and are suggested to have roles in various cancers (Agarwal et al., 2005;

Shukla et al., 2007). Furthermore, deregulations of this pathway have been linked to cell

survival [reviewed in (Song et al., 2005)]. It is also plausible that Cdh11 could be affecting the

Page 78: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

65

levels of other molecules it interacts with like p120, known to interact with RhoGTPases. Upon

loss of cadherins, p120ctn accumulates in the cytoplasm, repressing RhoA and activating Cdc42

and Rac1, which modulate the cytoskeleton and increase migration and invasiveness

(Christofori, 2006; Stemmler, 2008). To further understand how Cdh11 is actually affecting

apoptotic activity and invasion potential, future studies have to be done. Experiments involved

in directly look at downstream targets of pathways such as Wnt signaling, as well as microarray

analysis after shRNA inhibition of Cdh11 would provide clues into this mechanism.

Understanding these mechanisms will give important insight into cadherin molecules and their

links to various cellular pathways, as well as insight into tumorigenesis of retinoblastoma and

other cancers, where similar pathways may be affected.

4.5.2 Mechanism of Cdh11 Downregulation in Retinoblastoma

It is thought that the loss of RB1 facilitates and acts as a prelude to genomic instability, linked to

the role of pRB in regulation of S-phase and mitotic progression (Knudsen et al., 2006). Thus, it

is plausible that the loss of 16q and Cdh11 is a consequence of genomic instability secondary to

RB1 loss. In order to further understand Cdh11 in retinoblastoma tumorigenesis, the next step is

to identify how it is lost. To date, the mechanism of Cdh11 loss is unknown, apart from a

reduction in copy number from 2 to 1. Previous work in our lab has suggested that the loss of

Cdh11 is a late event in retinoblastoma progression (Bowles et al., 2007; Dimaras et al., 2008).

Some insight can be taken from what has been seen as mechanisms for downregulation of E-

cadherin. Germline, somatic, and missense mutations identified in the E-cadherin gene have

been observed in a variety of cancers, including gastric and breast (Gayther et al., 1998; Hajra

and Fearon, 2002). Epigenetic modifications such as DNA hypermethylation of the promoter is

observed in numerous carcinomas (Hu et al., 2002; Zhao et al., 2007), and transcriptional

repression via Snail and Slug have been suggested (Halbleib and Nelson, 2006). Post-

Page 79: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

66

transcriptional modifications such as phosphorylation of ϐ-catenin leading to disassembly of the

AJ (Hu et al., 2001), and cleavage through MMPs have also been implicated in enhancing

tumorigenesis and metastasis (Noe et al., 2001). To date, studies have shown some

hypermethylation in colon cancer cases (Braungart et al., 1999), but the promoter is not well

defined, so the mechanism of silencing is still unknown (Kashima et al., 1999; Zhou and Skalli,

2000). Some studies looking into the mechanism of loss of Cdh11 could include mutational

analysis within the gene, looking for somatic, germline or epigenetic modifications. Cytogenetic

studies could reveal translocations.

4.6 Summary and Significance

Our lab has contributed significantly to understanding tumorigenesis toward retinoblastoma. As

a proto-typical model of cancer, understanding the progression of retinoblastoma could have

profound implications in the study of cancer genetics and biology. In the recent years, our lab

has been successful in taking information from earlier genomic studies and defining candidate

genes that have oncogenic and tumor suppressive properties in retinoblastoma. My thesis

focused on defining the tumor suppressor role of Cdh11 in retinoblastoma progression. Earlier

evidence had supported the idea that Cdh11 acts as a tumor suppressor, and my work focused

around defining the specific mechanism in which it exerted this suppressive function. I found

that Cdh11 was promoting apoptosis within these tumors since when it was lost, a significant

decrease in apoptotic activity resulted, supported by both in vitro and in vivo analysis.

Furthermore, I saw no significant effect on proliferation, specifying the mechanism to promoting

apoptosis. I also explored the idea that since the loss of Cdh11 leads to faster growing tumors, if

they may be more aggressive and possibly lead to invasion. I showed the expression of various

invasion markers increased after specific knockdown of Cdh11. I also showed increased levels

of ϐ-catenin expression after Cdh11 knockdown, which opens the door for new studies to link

Page 80: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

67

Cdh11 to apoptosis and invasion. Identifying and specifying the tumor suppressive role of

Cdh11 in retinoblastoma, will lead not only to further understanding of retinoblastoma

tumorigenesis, but also of the role of Cdh11, perhaps in other cancers. Additionally, this study

could lead to developing more ideas of how cadherin molecules in general affect different

cellular mechanisms, signaling pathways and tissue specific diseases. As a cell surface

molecule, Cdh11 has great potential for therapy consideration once its role and interactions in

tumorigenesis are fully understood.

Page 81: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

68

References

Aberle H, Bauer A, Stappert J, Kispert A, Kemler R (1997). beta-catenin is a target for the

ubiquitin-proteasome pathway. EMBO J 16: 3797-804.

Aberle H, Butz S, Stappert J, Weissig H, Kemler R, Hoschuetzky H (1994). Assembly of the

cadherin-catenin complex in vitro with recombinant proteins. J Cell Sci 107 ( Pt 12): 3655-63.

Aberle H, Schwartz H, Kemler R (1996). Cadherin-catenin complex: protein interactions and

their implications for cadherin function. J Cell Biochem 61: 514-23.

Abramson DH (1999). Second nonocular cancers in retinoblastoma: a unified hypothesis. The

Franceschetti Lecture. Ophthalmic Genet 20: 193-204.

Abramson DH, Frank CM (1998). Second nonocular tumors in survivors of bilateral

retinoblastoma: a possible age effect on radiation-related risk. Ophthalmology 105: 573-9;

discussion 579-80.

Acehan D, Jiang X, Morgan DG, Heuser JE, Wang X, Akey CW (2002). Three-dimensional

structure of the apoptosome: implications for assembly, procaspase-9 binding, and activation.

Mol Cell 9: 423-32.

Adithi M, Nalini V, Kandalam M, Krishnakumar S (2007). Expression of matrix

metalloproteinases and their inhibitors in retinoblastoma. J Pediatr Hematol Oncol 29: 399-405.

Agarwal A, Das K, Lerner N, Sathe S, Cicek M, Casey G et al (2005). The AKT/I kappa B

kinase pathway promotes angiogenic/metastatic gene expression in colorectal cancer by

activating nuclear factor-kappa B and beta-catenin. Oncogene 24: 1021-31.

Alexander NR, Tran NL, Rekapally H, Summers CE, Glackin C, Heimark RL (2006). N-

cadherin gene expression in prostate carcinoma is modulated by integrin-dependent nuclear

translocation of Twist1. Cancer Res 66: 3365-9.

Almeida M, Han L, Bellido T, Manolagas SC, Kousteni S (2005). Wnt proteins prevent

apoptosis of both uncommitted osteoblast progenitors and differentiated osteoblasts by beta-

catenin-dependent and -independent signaling cascades involving Src/ERK and

phosphatidylinositol 3-kinase/AKT. J Biol Chem 280: 41342-51.

Balmer A, Munier F, Gailloud C (1991). Retinoma. Case studies. Ophthalmic Paediatr Genet

12: 131-7.

Balmer A, Zografos L, Munier F (2006). Diagnosis and current management of retinoblastoma.

Oncogene 25: 5341-9.

Bates RC, Mercurio AM (2005). The epithelial-mesenchymal transition (EMT) and colorectal

cancer progression. Cancer Biol Ther 4: 365-70.

Page 82: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

69

Baudis M, Cleary ML (2001). Progenetix.net: an online repository for molecular cytogenetic

aberration data. Bioinformatics 17: 1228-9.

Baudry D, Faussillon M, Cabanis MO, Rigolet M, Zucker JM, Patte C et al (2002). Changes in

WT1 splicing are associated with a specific gene expression profile in Wilms' tumour. Oncogene

21: 5566-73.

Beavon IR (2000). The E-cadherin-catenin complex in tumour metastasis: structure, function and

regulation. Eur J Cancer 36: 1607-20.

Becker I, Becker KF, Rohrl MH, Minkus G, Schutze K, Hofler H (1996). Single-cell mutation

analysis of tumors from stained histologic slides. Lab Invest 75: 801-7.

Becker KF, Atkinson MJ, Reich U, Becker I, Nekarda H, Siewert JR et al (1994). E-cadherin

gene mutations provide clues to diffuse type gastric carcinomas. Cancer Res 54: 3845-52.

Behrens J (1999). Cadherins and catenins: role in signal transduction and tumor progression.

Cancer Metastasis Rev 18: 15-30.

Behrens J, von Kries JP, Kuhl M, Bruhn L, Wedlich D, Grosschedl R et al (1996). Functional

interaction of beta-catenin with the transcription factor LEF-1. Nature 382: 638-42.

Bellahcene A, Bachelier R, Detry C, Lidereau R, Clezardin P, Castronovo V (2007).

Transcriptome analysis reveals an osteoblast-like phenotype for human osteotropic breast cancer

cells. Breast Cancer Res Treat 101: 135-48.

Bendel-Stenzel MR, Gomperts M, Anderson R, Heasman J, Wylie C (2000). The role of

cadherins during primordial germ cell migration and early gonad formation in the mouse. Mech

Dev 91: 143-52.

Benedict WF, Banerjee A, Mark C, Murphree AL (1983). Nonrandom chromosomal changes in

untreated retinoblastomas. Cancer Genet Cytogenet 10: 311-33.

Billen LP, Shamas-Din A, Andrews DW (2008). Bid: a Bax-like BH3 protein. Oncogene 27

Suppl 1: S93-104.

Birchmeier W, Behrens J (1994). Cadherin expression in carcinomas: role in the formation of

cell junctions and the prevention of invasiveness. Biochim Biophys Acta 1198: 11-26.

Blaschuk OW, Sullivan R, David S, Pouliot Y (1990). Identification of a cadherin cell adhesion

recognition sequence. Dev Biol 139: 227-9.

Bookstein R, Lee WH (1991). Molecular genetics of the retinoblastoma suppressor gene. Crit

Rev Oncog 2: 211-27.

Bowles E, Corson TW, Bayani J, Squire JA, Wong N, Lai PB et al (2007). Profiling genomic

copy number changes in retinoblastoma beyond loss of RB1. Genes Chromosomes Cancer 46:

118-29.

Page 83: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

70

Braungart E, Schumacher C, Hartmann E, Nekarda H, Becker KF, Hofler H et al (1999).

Functional loss of E-cadherin and cadherin-11 alleles on chromosome 16q22 in colonic cancer. J

Pathol 187: 530-4.

Brodeur GM (2003). Neuroblastoma: biological insights into a clinical enigma. Nat Rev Cancer

3: 203-16.

Bussemakers MJ, Van Bokhoven A, Tomita K, Jansen CF, Schalken JA (2000). Complex

cadherin expression in human prostate cancer cells. Int J Cancer 85: 446-50.

Cai Y, Mohseny AB, Karperien M, Hogendoorn PC, Zhou G, Cleton-Jansen AM (2009).

Inactive Wnt/beta-catenin pathway in conventional high-grade osteosarcoma. J Pathol.

Cao L, Faha B, Dembski M, Tsai LH, Harlow E, Dyson N (1992). Independent binding of the

retinoblastoma protein and p107 to the transcription factor E2F. Nature 355: 176-9.

Carleton M, Mao M, Biery M, Warrener P, Kim S, Buser C et al (2006). RNA interference-

mediated silencing of mitotic kinesin KIF14 disrupts cell cycle progression and induces

cytokinesis failure. Mol Cell Biol 26: 3853-63.

Carro MS, Spiga FM, Quarto M, Di Ninni V, Volorio S, Alcalay M et al (2006). DEK

Expression is controlled by E2F and deregulated in diverse tumor types. Cell Cycle 5: 1202-7.

Cavalieri F, Goldfarb M (1988). N-myc proto-oncogene expression can induce DNA replication

in Balb/c 3T3 fibroblasts. Oncogene 2: 289-91.

Cavallaro U, Christofori G (2004). Multitasking in tumor progression: signaling functions of cell

adhesion molecules. Ann N Y Acad Sci 1014: 58-66.

Cavallaro U, Niedermeyer J, Fuxa M, Christofori G (2001). N-CAM modulates tumour-cell

adhesion to matrix by inducing FGF-receptor signalling. Nat Cell Biol 3: 650-7.

Cavenee WK, Dryja TP, Phillips RA, Benedict WF, Godbout R, Gallie BL et al (1983).

Expression of recessive alleles by chromosomal mechanisms in retinoblastoma. Nature 305:

779-84.

Cepko CL, Austin CP, Yang X, Alexiades M, Ezzeddine D (1996). Cell fate determination in the

vertebrate retina. Proc Natl Acad Sci U S A 93: 589-95.

Chan HS, Gallie BL, DeBoer G, Haddad G, Ikegaki N, Dimitroulakos J et al (1997). MYCN

protein expression as a predictor of neuroblastoma prognosis. Clin Cancer Res 3: 1699-706.

Chan HS, Gallie BL, Munier FL, Beck Popovic M (2005). Chemotherapy for retinoblastoma.

Ophthalmol Clin North Am 18: 55-63, viii.

Page 84: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

71

Chantada GL, Fandino AC, Raslawski EC, Manzitti J, de Davila MT, Casak SJ et al (2005).

Experience with chemoreduction and focal therapy for intraocular retinoblastoma in a

developing country. Pediatr Blood Cancer 44: 455-60.

Chaum E, Ellsworth RM, Abramson DH, Haik BG, Kitchin FD, Chaganti RS (1984).

Cytogenetic analysis of retinoblastoma: evidence for multifocal origin and in vivo gene

amplification. Cytogenet Cell Genet 38: 82-91.

Chellappan SP, Hiebert S, Mudryj M, Horowitz JM, Nevins JR (1991). The E2F transcription

factor is a cellular target for the RB protein. Cell 65: 1053-61.

Chen D, Gallie BL, Squire JA (2001). Minimal regions of chromosomal imbalance in

retinoblastoma detected by comparative genomic hybridization. Cancer Genet Cytogenet 129:

57-63.

Chen D, Livne-bar I, Vanderluit JL, Slack RS, Agochiya M, Bremner R (2004). Cell-specific

effects of RB or RB/p107 loss on retinal development implicate an intrinsically death-resistant

cell-of-origin in retinoblastoma. Cancer Cell 5: 539-51.

Chen WC, Obrink B (1991). Cell-cell contacts mediated by E-cadherin (uvomorulin) restrict

invasive behavior of L-cells. J Cell Biol 114: 319-27.

Chintagumpala M, Chevez-Barrios P, Paysse EA, Plon SE, Hurwitz R (2007). Retinoblastoma:

review of current management. Oncologist 12: 1237-46.

Chow RL, Lang RA (2001). Early eye development in vertebrates. Annu Rev Cell Dev Biol 17:

255-96.

Christofori G (2003). Changing neighbours, changing behaviour: cell adhesion molecule-

mediated signalling during tumour progression. EMBO J 22: 2318-23.

Christofori G (2006). New signals from the invasive front. Nature 441: 444-50.

Chu K, Cheng CJ, Ye X, Lee YC, Zurita AJ, Chen DT et al (2008). Cadherin-11 promotes the

metastasis of prostate cancer cells to bone. Mol Cancer Res 6: 1259-67.

Clarke AR, Maandag ER, van Roon M, van der Lugt NM, van der Valk M, Hooper ML et al

(1992). Requirement for a functional Rb-1 gene in murine development. Nature 359: 328-30.

Clevers H (2006). Wnt/beta-catenin signaling in development and disease. Cell 127: 469-80.

Cobrinik D (2005). Pocket proteins and cell cycle control. Oncogene 24: 2796-809.

Cohen GM (1997). Caspases: the executioners of apoptosis. Biochem J 326 ( Pt 1): 1-16.

Colman DR (1997). Neurites, synapses, and cadherins reconciled. Mol Cell Neurosci 10: 1-6.

Comings DE (1973). A general theory of carcinogenesis. Proc Natl Acad Sci U S A 70: 3324-8.

Page 85: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

72

Corn PG, Heath EI, Heitmiller R, Fogt F, Forastiere AA, Herman JG et al (2001). Frequent

hypermethylation of the 5' CpG island of E-cadherin in esophageal adenocarcinoma. Clin Cancer

Res 7: 2765-9.

Corn PG, Smith BD, Ruckdeschel ES, Douglas D, Baylin SB, Herman JG (2000). E-cadherin

expression is silenced by 5' CpG island methylation in acute leukemia. Clin Cancer Res 6: 4243-

8.

Corson TW, Gallie BL (2006). KIF14 mRNA expression is a predictor of grade and outcome in

breast cancer. Int J Cancer 119: 1088-94.

Corson TW, Gallie BL (2007). One hit, two hits, three hits, more? Genomic changes in the

development of retinoblastoma. Genes Chromosomes Cancer 46: 617-34.

Corson TW, Huang A, Tsao MS, Gallie BL (2005). KIF14 is a candidate oncogene in the 1q

minimal region of genomic gain in multiple cancers. Oncogene 24: 4741-53.

Corson TW, Zhu CQ, Lau SK, Shepherd FA, Tsao MS, Gallie BL (2007). KIF14 messenger

RNA expression is independently prognostic for outcome in lung cancer. Clin Cancer Res 13:

3229-34.

Dannenberg JH, Schuijff L, Dekker M, van der Valk M, te Riele H (2004). Tissue-specific tumor

suppressor activity of retinoblastoma gene homologs p107 and p130. Genes Dev 18: 2952-62.

Davidoff AM (2009). Wilms' tumor. Curr Opin Pediatr 21: 357-64.

Davis MA, Ireton RC, Reynolds AB (2003). A core function for p120-catenin in cadherin

turnover. J Cell Biol 163: 525-34.

de Bruin A, Wu L, Saavedra HI, Wilson P, Yang Y, Rosol TJ et al (2003). Rb function in

extraembryonic lineages suppresses apoptosis in the CNS of Rb-deficient mice. Proc Natl Acad

Sci U S A 100: 6546-51.

Deshpande A, Hinds PW (2006). The retinoblastoma protein in osteoblast differentiation and

osteosarcoma. Curr Mol Med 6: 809-17.

DiCiommo D, Gallie BL, Bremner R (2000). Retinoblastoma: the disease, gene and protein

provide critical leads to understand cancer. Semin Cancer Biol 10: 255-69.

Dimaras H, Coburn B, Pajovic S, Gallie BL (2006). Loss of p75 neurotrophin receptor

expression accompanies malignant progression to human and murine retinoblastoma. Mol

Carcinog 45: 333-43.

Dimaras H, Khetan V, Halliday W, Orlic M, Prigoda NL, Piovesan B et al (2008). Loss of RB1

induces non-proliferative retinoma: increasing genomic instability correlates with progression to

retinoblastoma. Hum Mol Genet 17: 1363-72.

Dimaras H, Marchong MN, Gallie BL (2009). Quantitative analysis of tumor size in a murine

model of retinoblastoma. Ophthalmic Genet 30: 84-90.

Page 86: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

73

Dryja TP, Friend S, Weinberg RA (1986). Genetic sequences that predispose to retinoblastoma

and osteosarcoma. Symp Fundam Cancer Res 39: 115-9.

Dyer MA, Bremner R (2005). The search for the retinoblastoma cell of origin. Nat Rev Cancer

5: 91-101.

Dyson N, Buchkovich K, Whyte P, Harlow E (1989). Cellular proteins that are targetted by DNA

tumor viruses for transformation. Princess Takamatsu Symp 20: 191-8.

Eagle RC, Jr., Shields JA, Donoso L, Milner RS (1989). Malignant transformation of

spontaneously regressed retinoblastoma, retinoma/retinocytoma variant. Ophthalmology 96:

1389-95.

Erdmann B, Kirsch FP, Rathjen FG, More MI (2003). N-cadherin is essential for retinal

lamination in the zebrafish. Dev Dyn 226: 570-7.

Erez N, Zamir E, Gour BJ, Blaschuk OW, Geiger B (2004). Induction of apoptosis in cultured

endothelial cells by a cadherin antagonist peptide: involvement of fibroblast growth factor

receptor-mediated signalling. Exp Cell Res 294: 366-78.

Falschlehner C, Ganten TM, Koschny R, Schaefer U, Walczak H (2009). TRAIL and Other

TRAIL Receptor Agonists as Novel Cancer Therapeutics. Adv Exp Med Biol 647: 195-206.

Fang D, Hawke D, Zheng Y, Xia Y, Meisenhelder J, Nika H et al (2007). Phosphorylation of

beta-catenin by AKT promotes beta-catenin transcriptional activity. J Biol Chem 282: 11221-9.

Farina AK, Bong YS, Feltes CM, Byers SW (2009). Post-transcriptional regulation of cadherin-

11 expression by GSK-3 and beta-catenin in prostate and breast cancer cells. PLoS One 4:

e4797.

Faulkner-Jones BE, Godinho LN, Reese BE, Pasquini GF, Ruefli A, Tan SS (1999a). Cloning

and expression of mouse Cadherin-7, a type-II cadherin isolated from the developing eye. Mol

Cell Neurosci 14: 1-16.

Faulkner-Jones BE, Godinho LN, Tan SS (1999b). Multiple cadherin mRNA expression and

developmental regulation of a novel cadherin in the developing mouse eye. Exp Neurol 156:

316-25.

Felsani A, Mileo AM, Paggi MG (2006). Retinoblastoma family proteins as key targets of the

small DNA virus oncoproteins. Oncogene 25: 5277-85.

Friend SH, Bernards R, Rogelj S, Weinberg RA, Rapaport JM, Albert DM et al (1986). A human

DNA segment with properties of the gene that predisposes to retinoblastoma and osteosarcoma.

Nature 323: 643-6.

Page 87: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

74

Fujita Y, Krause G, Scheffner M, Zechner D, Leddy HE, Behrens J et al (2002). Hakai, a c-Cbl-

like protein, ubiquitinates and induces endocytosis of the E-cadherin complex. Nat Cell Biol 4:

222-31.

Fung YK, Murphree AL, T'Ang A, Qian J, Hinrichs SH, Benedict WF (1987). Structural

evidence for the authenticity of the human retinoblastoma gene. Science 236: 1657-61.

Gallie BL, Ellsworth RM, Abramson DH, Phillips RA (1982). Retinoma: spontaneous regression

of retinoblastoma or benign manifestation of the mutation? Br J Cancer 45: 513-21.

Gardner HA, Gallie BL, Knight LA, Phillips RA (1982). Multiple karyotypic changes in

retinoblastoma tumor cells: presence of normal chromosome No. 13 in most tumors. Cancer

Genet Cytogenet 6: 201-11.

Gayther SA, Gorringe KL, Ramus SJ, Huntsman D, Roviello F, Grehan N et al (1998).

Identification of germ-line E-cadherin mutations in gastric cancer families of European origin.

Cancer Res 58: 4086-9.

Getsios S, MacCalman CD (2003). Cadherin-11 modulates the terminal differentiation and

fusion of human trophoblastic cells in vitro. Dev Biol 257: 41-54.

Ghiotto F, Fais F, Bruno S (2009). BH3-only proteins: The death-puppeteer's wires. Cytometry

A.

Godbout R, Dryja TP, Squire J, Gallie BL, Phillips RA (1983). Somatic inactivation of genes on

chromosome 13 is a common event in retinoblastoma. Nature 304: 451-3.

Gottardi CJ, Wong E, Gumbiner BM (2001). E-cadherin suppresses cellular transformation by

inhibiting beta-catenin signaling in an adhesion-independent manner. J Cell Biol 153: 1049-60.

Grasemann C, Gratias S, Stephan H, Schuler A, Schramm A, Klein-Hitpass L et al (2005). Gains

and overexpression identify DEK and E2F3 as targets of chromosome 6p gains in

retinoblastoma. Oncogene 24: 6441-9.

Gratias S, Rieder H, Ullmann R, Klein-Hitpass L, Schneider S, Boloni R et al (2007). Allelic

loss in a minimal region on chromosome 16q24 is associated with vitreous seeding of

retinoblastoma. Cancer Res 67: 408-16.

Gravdal K, Halvorsen OJ, Haukaas SA, Akslen LA (2007). A switch from E-cadherin to N-

cadherin expression indicates epithelial to mesenchymal transition and is of strong and

independent importance for the progress of prostate cancer. Clin Cancer Res 13: 7003-11.

Green DR, Kroemer G (2004). The pathophysiology of mitochondrial cell death. Science 305:

626-9.

Gruneberg U, Neef R, Li X, Chan EH, Chalamalasetty RB, Nigg EA et al (2006). KIF14 and

citron kinase act together to promote efficient cytokinesis. J Cell Biol 172: 363-72.

Page 88: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

75

Guilford P, Hopkins J, Harraway J, McLeod M, McLeod N, Harawira P et al (1998). E-cadherin

germline mutations in familial gastric cancer. Nature 392: 402-5.

Guillemot F, Cepko CL (1992). Retinal fate and ganglion cell differentiation are potentiated by

acidic FGF in an in vitro assay of early retinal development. Development 114: 743-54.

Gumbiner BM (1995). Signal transduction of beta-catenin. Curr Opin Cell Biol 7: 634-40.

Gumbiner BM (1996). Cell adhesion: the molecular basis of tissue architecture and

morphogenesis. Cell 84: 345-57.

Hajra KM, Fearon ER (2002). Cadherin and catenin alterations in human cancer. Genes

Chromosomes Cancer 34: 255-68.

Hakem R, Hakem A, Duncan GS, Henderson JT, Woo M, Soengas MS et al (1998). Differential

requirement for caspase 9 in apoptotic pathways in vivo. Cell 94: 339-52.

Halbleib JM, Nelson WJ (2006). Cadherins in development: cell adhesion, sorting, and tissue

morphogenesis. Genes Dev 20: 3199-214.

Handschuh G, Candidus S, Luber B, Reich U, Schott C, Oswald S et al (1999). Tumour-

associated E-cadherin mutations alter cellular morphology, decrease cellular adhesion and

increase cellular motility. Oncogene 18: 4301-12.

Handschuh G, Luber B, Hutzler P, Hofler H, Becker KF (2001). Single amino acid substitutions

in conserved extracellular domains of E-cadherin differ in their functional consequences. J Mol

Biol 314: 445-54.

Harbour JW, Lai SL, Whang-Peng J, Gazdar AF, Minna JD, Kaye FJ (1988). Abnormalities in

structure and expression of the human retinoblastoma gene in SCLC. Science 241: 353-7.

Harrison OJ, Corps EM, Berge T, Kilshaw PJ (2005). The mechanism of cell adhesion by

classical cadherins: the role of domain 1. J Cell Sci 118: 711-21.

Hartland SN, Murphy F, Aucott RL, Abergel A, Zhou X, Waung J et al (2009). Active matrix

metalloproteinase-2 promotes apoptosis of hepatic stellate cells via the cleavage of cellular N-

cadherin. Liver Int 29: 966-78.

Hazan RB, Phillips GR, Qiao RF, Norton L, Aaronson SA (2000). Exogenous expression of N-

cadherin in breast cancer cells induces cell migration, invasion, and metastasis. J Cell Biol 148:

779-90.

Hazan RB, Qiao R, Keren R, Badano I, Suyama K (2004). Cadherin switch in tumor progression.

Ann N Y Acad Sci 1014: 155-63.

Hengartner MO (2000). The biochemistry of apoptosis. Nature 407: 770-6.

Herath NI, Leggett BA, MacDonald GA (2006). Review of genetic and epigenetic alterations in

hepatocarcinogenesis. J Gastroenterol Hepatol 21: 15-21.

Page 89: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

76

Hermiston ML, Gordon JI (1995). In vivo analysis of cadherin function in the mouse intestinal

epithelium: essential roles in adhesion, maintenance of differentiation, and regulation of

programmed cell death. J Cell Biol 129: 489-506.

Herzog S, Lohmann DR, Buiting K, Schuler A, Horsthemke B, Rehder H et al (2001). Marked

differences in unilateral isolated retinoblastomas from young and older children studied by

comparative genomic hybridization. Hum Genet 108: 98-104.

Hirano S, Nose A, Hatta K, Kawakami A, Takeichi M (1987). Calcium-dependent cell-cell

adhesion molecules (cadherins): subclass specificities and possible involvement of actin bundles.

J Cell Biol 105: 2501-10.

Hoffman I, Balling R (1995). Chromosomal localization of the murine cadherin-11. Mamm

Genome 6: 304.

Honjo M, Tanihara H, Suzuki S, Tanaka T, Honda Y, Takeichi M (2000). Differential expression

of cadherin adhesion receptors in neural retina of the postnatal mouse. Invest Ophthalmol Vis Sci

41: 546-51.

Horikawa K, Radice G, Takeichi M, Chisaka O (1999). Adhesive subdivisions intrinsic to the

epithelial somites. Dev Biol 215: 182-9.

Hu P, O'Keefe EJ, Rubenstein DS (2001). Tyrosine phosphorylation of human keratinocyte beta-

catenin and plakoglobin reversibly regulates their binding to E-cadherin and alpha-catenin. J

Invest Dermatol 117: 1059-67.

Hu XC, Loo WT, Chow LW (2002). E-cadherin promoter methylation can regulate its

expression in invasive ductal breast cancer tissue in Chinese woman. Life Sci 71: 1397-404.

Huber O, Bierkamp C, Kemler R (1996). Cadherins and catenins in development. Curr Opin Cell

Biol 8: 685-91.

Hulit J, Suyama K, Chung S, Keren R, Agiostratidou G, Shan W et al (2007). N-cadherin

signaling potentiates mammary tumor metastasis via enhanced extracellular signal-regulated

kinase activation. Cancer Res 67: 3106-16.

Hurst CD, Tomlinson DC, Williams SV, Platt FM, Knowles MA (2008). Inactivation of the Rb

pathway and overexpression of both isoforms of E2F3 are obligate events in bladder tumours

with 6p22 amplification. Oncogene 27: 2716-27.

Hurwitz MY, Marcus KT, Chevez-Barrios P, Louie K, Aguilar-Cordova E, Hurwitz RL (1999).

Suicide gene therapy for treatment of retinoblastoma in a murine model. Hum Gene Ther 10:

441-8.

Ikeda S, Kishida M, Matsuura Y, Usui H, Kikuchi A (2000). GSK-3beta-dependent

phosphorylation of adenomatous polyposis coli gene product can be modulated by beta-catenin

and protein phosphatase 2A complexed with Axin. Oncogene 19: 537-45.

Page 90: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

77

Jacks T, Fazeli A, Schmitt EM, Bronson RT, Goodell MA, Weinberg RA (1992). Effects of an

Rb mutation in the mouse. Nature 359: 295-300.

Jeanes A, Gottardi CJ, Yap AS (2008). Cadherins and cancer: how does cadherin dysfunction

promote tumor progression? Oncogene 27: 6920-9.

Kanai Y, Ushijima S, Hui AM, Ochiai A, Tsuda H, Sakamoto M et al (1997). The E-cadherin

gene is silenced by CpG methylation in human hepatocellular carcinomas. Int J Cancer 71: 355-

9.

Kang SJ, Durairaj C, Kompella UB, O'Brien JM, Grossniklaus HE (2009). Subconjunctival

nanoparticle carboplatin in the treatment of murine retinoblastoma. Arch Ophthalmol 127: 1043-

7.

Kappes F, Burger K, Baack M, Fackelmayer FO, Gruss C (2001). Subcellular localization of the

human proto-oncogene protein DEK. J Biol Chem 276: 26317-23.

Karreth F, Tuveson DA (2004). Twist induces an epithelial-mesenchymal transition to facilitate

tumor metastasis. Cancer Biol Ther 3: 1058-9.

Kashima T, Kawaguchi J, Takeshita S, Kuroda M, Takanashi M, Horiuchi H et al (1999).

Anomalous cadherin expression in osteosarcoma. Possible relationships to metastasis and

morphogenesis. Am J Pathol 155: 1549-55.

Kashima T, Nakamura K, Kawaguchi J, Takanashi M, Ishida T, Aburatani H et al (2003).

Overexpression of cadherins suppresses pulmonary metastasis of osteosarcoma in vivo. Int J

Cancer 104: 147-54.

Kawaguchi J, Azuma Y, Hoshi K, Kii I, Takeshita S, Ohta T et al (2001). Targeted disruption of

cadherin-11 leads to a reduction in bone density in calvaria and long bone metaphyses. J Bone

Miner Res 16: 1265-71.

Kawaguchi J, Takeshita S, Kashima T, Imai T, Machinami R, Kudo A (1999). Expression and

function of the splice variant of the human cadherin-11 gene in subordination to intact cadherin-

11. J Bone Miner Res 14: 764-75.

Kawakami M, Staub J, Cliby W, Hartmann L, Smith DI, Shridhar V (1999). Involvement of H-

cadherin (CDH13) on 16q in the region of frequent deletion in ovarian cancer. Int J Oncol 15:

715-20.

Kemler R (1993). From cadherins to catenins: cytoplasmic protein interactions and regulation of

cell adhesion. Trends Genet 9: 317-21.

Kerbauy DB, Deeg HJ (2007). Apoptosis and antiapoptotic mechanisms in the progression of

myelodysplastic syndrome. Exp Hematol 35: 1739-46.

Page 91: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

78

Kerr JF, Winterford CM, Harmon BV (1994). Apoptosis. Its significance in cancer and cancer

therapy. Cancer 73: 2013-26.

Kerr JF, Wyllie AH, Currie AR (1972). Apoptosis: a basic biological phenomenon with wide-

ranging implications in tissue kinetics. Br J Cancer 26: 239-57.

Kiener HP, Lee DM, Agarwal SK, Brenner MB (2006). Cadherin-11 induces rheumatoid arthritis

fibroblast-like synoviocytes to form lining layers in vitro. Am J Pathol 168: 1486-99.

Kiener HP, Niederreiter B, Lee DM, Jimenez-Boj E, Smolen JS, Brenner MB (2009). Cadherin

11 promotes invasive behavior of fibroblast-like synoviocytes. Arthritis Rheum 60: 1305-10.

Kii I, Amizuka N, Shimomura J, Saga Y, Kudo A (2004). Cell-cell interaction mediated by

cadherin-11 directly regulates the differentiation of mesenchymal cells into the cells of the osteo-

lineage and the chondro-lineage. J Bone Miner Res 19: 1840-9.

Kim do H, Nagano Y, Choi IS, White JA, Yao JC, Rashid A (2008). Allelic alterations in well-

differentiated neuroendocrine tumors (carcinoid tumors) identified by genome-wide single

nucleotide polymorphism analysis and comparison with pancreatic endocrine tumors. Genes

Chromosomes Cancer 47: 84-92.

King KL, Cidlowski JA (1995). Cell cycle and apoptosis: common pathways to life and death. J

Cell Biochem 58: 175-80.

Kinzler KW, Vogelstein B (1996). Lessons from hereditary colorectal cancer. Cell 87: 159-70.

Kippenberger S, Loitsch S, Thaci D, Muller J, Guschel M, Kaufmann R et al (2006). Restoration

of E-cadherin sensitizes human melanoma cells for apoptosis. Melanoma Res 16: 393-403.

Kljavin IJ, Lagenaur C, Bixby JL, Reh TA (1994). Cell adhesion molecules regulating neurite

growth from amacrine and rod photoreceptor cells. J Neurosci 14: 5035-49.

Knudsen ES, Sexton CR, Mayhew CN (2006). Role of the retinoblastoma tumor suppressor in

the maintenance of genome integrity. Curr Mol Med 6: 749-57.

Knudson AG, Jr. (1971). Mutation and cancer: statistical study of retinoblastoma. Proc Natl

Acad Sci U S A 68: 820-3.

Kobielak A, Fuchs E (2004). Alpha-catenin: at the junction of intercellular adhesion and actin

dynamics. Nat Rev Mol Cell Biol 5: 614-25.

Koenig A, Mueller C, Hasel C, Adler G, Menke A (2006). Collagen type I induces disruption of

E-cadherin-mediated cell-cell contacts and promotes proliferation of pancreatic carcinoma cells.

Cancer Res 66: 4662-71.

Kouzarides T (1995). Transcriptional control by the retinoblastoma protein. Semin Cancer Biol

6: 91-8.

Page 92: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

79

Krueger A, Schmitz I, Baumann S, Krammer PH, Kirchhoff S (2001). Cellular FLICE-inhibitory

protein splice variants inhibit different steps of caspase-8 activation at the CD95 death-inducing

signaling complex. J Biol Chem 276: 20633-40.

Kurrey NK, K A, Bapat SA (2005). Snail and Slug are major determinants of ovarian cancer

invasiveness at the transcription level. Gynecol Oncol 97: 155-65.

Kusnetsova LE, Prigogina EL, Pogosianz HE, Belkina BM (1982). Similar chromosomal

abnormalities in several retinoblastomas. Hum Genet 61: 201-4.

Laurie N, Mohan A, McEvoy J, Reed D, Zhang J, Schweers B et al (2009). Changes in

retinoblastoma cell adhesion associated with optic nerve invasion. Mol Cell Biol 29: 6268-82.

Lee CH, Herman T, Clandinin TR, Lee R, Zipursky SL (2001). N-cadherin regulates target

specificity in the Drosophila visual system. Neuron 30: 437-50.

Lee DM, Kiener HP, Agarwal SK, Noss EH, Watts GF, Chisaka O et al (2007). Cadherin-11 in

synovial lining formation and pathology in arthritis. Science 315: 1006-10.

Lee EY, Chang CY, Hu N, Wang YC, Lai CC, Herrup K et al (1992). Mice deficient for Rb are

nonviable and show defects in neurogenesis and haematopoiesis. Nature 359: 288-94.

Lee WH, Bookstein R, Hong F, Young LJ, Shew JY, Lee EY (1987). Human retinoblastoma

susceptibility gene: cloning, identification, and sequence. Science 235: 1394-9.

Lee WH, Murphree AL, Benedict WF (1984). Expression and amplification of the N-myc gene

in primary retinoblastoma. Nature 309: 458-60.

Leone G, DeGregori J, Yan Z, Jakoi L, Ishida S, Williams RS et al (1998). E2F3 activity is

regulated during the cell cycle and is required for the induction of S phase. Genes Dev 12: 2120-

30.

Li G, Herlyn M (2000). Dynamics of intercellular communication during melanoma

development. Mol Med Today 6: 163-9.

Li P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, Alnemri ES et al (1997).

Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an

apoptotic protease cascade. Cell 91: 479-89.

Lillington DM, Kingston JE, Coen PG, Price E, Hungerford J, Domizio P et al (2003).

Comparative genomic hybridization of 49 primary retinoblastoma tumors identifies

chromosomal regions associated with histopathology, progression, and patient outcome. Genes

Chromosomes Cancer 36: 121-8.

Lipinski MM, Jacks T (1999). The retinoblastoma gene family in differentiation and

development. Oncogene 18: 7873-82.

Liu Q, Londraville R, Marrs JA, Wilson AL, Mbimba T, Murakami T et al (2008). Cadherin-6

function in zebrafish retinal development. Dev Neurobiol 68: 1107-22.

Page 93: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

80

Llorens A, Rodrigo I, Lopez-Barcons L, Gonzalez-Garrigues M, Lozano E, Vinyals A et al

(1998). Down-regulation of E-cadherin in mouse skin carcinoma cells enhances a migratory and

invasive phenotype linked to matrix metalloproteinase-9 gelatinase expression. Lab Invest 78:

1131-42.

Lo KC, Rossi MR, Eberhart CG, Cowell JK (2007). Genome wide copy number abnormalities in

pediatric medulloblastomas as assessed by array comparative genome hybridization. Brain

Pathol 17: 282-96.

Loo LW, Grove DI, Williams EM, Neal CL, Cousens LA, Schubert EL et al (2004). Array

comparative genomic hybridization analysis of genomic alterations in breast cancer subtypes.

Cancer Res 64: 8541-9.

Lowe SW, Lin AW (2000). Apoptosis in cancer. Carcinogenesis 21: 485-95.

Luo J, Lubaroff DM, Hendrix MJ (1999). Suppression of prostate cancer invasive potential and

matrix metalloproteinase activity by E-cadherin transfection. Cancer Res 59: 3552-6.

Maandag EC, van der Valk M, Vlaar M, Feltkamp C, O'Brien J, van Roon M et al (1994).

Developmental rescue of an embryonic-lethal mutation in the retinoblastoma gene in chimeric

mice. EMBO J 13: 4260-8.

MacCalman CD, Furth EE, Omigbodun A, Bronner M, Coutifaris C, Strauss JF, 3rd (1996).

Regulated expression of cadherin-11 in human epithelial cells: a role for cadherin-11 in

trophoblast-endometrium interactions? Dev Dyn 206: 201-11.

MacPherson D, Conkrite K, Tam M, Mukai S, Mu D, Jacks T (2007). Murine bilateral

retinoblastoma exhibiting rapid-onset, metastatic progression and N-myc gene amplification.

EMBO J 26: 784-94.

MacPherson D, Sage J, Kim T, Ho D, McLaughlin ME, Jacks T (2004). Cell type-specific

effects of Rb deletion in the murine retina. Genes Dev 18: 1681-94.

Mairal A, Pinglier E, Gilbert E, Peter M, Validire P, Desjardins L et al (2000). Detection of

chromosome imbalances in retinoblastoma by parallel karyotype and CGH analyses. Genes

Chromosomes Cancer 28: 370-9.

Manabe T, Togashi H, Uchida N, Suzuki SC, Hayakawa Y, Yamamoto M et al (2000). Loss of

cadherin-11 adhesion receptor enhances plastic changes in hippocampal synapses and modifies

behavioral responses. Mol Cell Neurosci 15: 534-46.

Marambaud P, Wen PH, Dutt A, Shioi J, Takashima A, Siman R et al (2003). A CBP binding

transcriptional repressor produced by the PS1/epsilon-cleavage of N-cadherin is inhibited by PS1

FAD mutations. Cell 114: 635-45.

Marchong MN, Chen D, Corson TW, Lee C, Harmandayan M, Bowles E et al (2004). Minimal

16q genomic loss implicates cadherin-11 in retinoblastoma. Mol Cancer Res 2: 495-503.

Page 94: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

81

Marees T, Moll AC, Imhof SM, de Boer MR, Ringens PJ, van Leeuwen FE (2008). Risk of

second malignancies in survivors of retinoblastoma: more than 40 years of follow-up. J Natl

Cancer Inst 100: 1771-9.

Margineanu E, Cotrutz CE, Cotrutz C (2008). Correlation between E-cadherin abnormal

expressions in different types of cancer and the process of metastasis. Rev Med Chir Soc Med

Nat Iasi 112: 432-6.

Markus MA, Reichmuth C, Atkinson MJ, Reich U, Hoffmann I, Balling R et al (1999).

Cadherin-11 is highly expressed in rhabdomyosarcomas and during differentiation of myoblasts

in vitro. J Pathol 187: 164-72.

Marthiens V, Gavard J, Padilla F, Monnet C, Castellani V, Lambert M et al (2005). A novel

function for cadherin-11 in the regulation of motor axon elongation and fasciculation. Mol Cell

Neurosci 28: 715-26.

Masai I, Lele Z, Yamaguchi M, Komori A, Nakata A, Nishiwaki Y et al (2003). N-cadherin

mediates retinal lamination, maintenance of forebrain compartments and patterning of retinal

neurites. Development 130: 2479-94.

McCusker C, Cousin H, Neuner R, Alfandari D (2009). Extracellular cleavage of cadherin-11 by

ADAM metalloproteases is essential for Xenopus cranial neural crest cell migration. Mol Biol

Cell 20: 78-89.

Mertens F, Mandahl N, Mitelman F, Heim S (1994). Cytogenetic analysis in the examination of

solid tumors in children. Pediatr Hematol Oncol 11: 361-77.

Mills MD, Windle JJ, Albert DM (1999). Retinoblastoma in transgenic mice: models of

hereditary retinoblastoma. Surv Ophthalmol 43: 508-18.

Mittnacht S (1998). Control of pRB phosphorylation. Curr Opin Genet Dev 8: 21-7.

Mohan A, Nalini V, Mallikarjuna K, Jyotirmay B, Krishnakumar S (2007). Expression of

motility-related protein MRP1/CD9, N-cadherin, E-cadherin, alpha-catenin and beta-catenin in

retinoblastoma. Exp Eye Res 84: 781-9.

Monahan TS, Andersen ND, Panossian H, Kalish JA, Daniel S, Shrikhande GV et al (2007). A

novel function for cadherin 11/osteoblast-cadherin in vascular smooth muscle cells: modulation

of cell migration and proliferation. J Vasc Surg 45: 581-9.

Morin PJ, Sparks AB, Korinek V, Barker N, Clevers H, Vogelstein B et al (1997). Activation of

beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC. Science 275:

1787-90.

Mrozek K (2008). Cytogenetic, molecular genetic, and clinical characteristics of acute myeloid

leukemia with a complex karyotype. Semin Oncol 35: 365-77.

Page 95: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

82

Munro SB, Turner IM, Farookhi R, Blaschuk OW, Jothy S (1995). E-cadherin and OB-cadherin

mRNA levels in normal human colon and colon carcinoma. Exp Mol Pathol 62: 118-22.

Nagaraja GM, Othman M, Fox BP, Alsaber R, Pellegrino CM, Zeng Y et al (2006). Gene

expression signatures and biomarkers of noninvasive and invasive breast cancer cells:

comprehensive profiles by representational difference analysis, microarrays and proteomics.

Oncogene 25: 2328-38.

Nagi C, Guttman M, Jaffer S, Qiao R, Keren R, Triana A et al (2005). N-cadherin expression in

breast cancer: correlation with an aggressive histologic variant--invasive micropapillary

carcinoma. Breast Cancer Res Treat 94: 225-35.

Nakajima G, Patino-Garcia A, Bruheim S, Xi Y, San Julian M, Lecanda F et al (2008). CDH11

expression is associated with survival in patients with osteosarcoma. Cancer Genomics

Proteomics 5: 37-42.

Nawrocki-Raby B, Gilles C, Polette M, Bruyneel E, Laronze JY, Bonnet N et al (2003).

Upregulation of MMPs by soluble E-cadherin in human lung tumor cells. Int J Cancer 105: 790-

5.

Nieman MT, Prudoff RS, Johnson KR, Wheelock MJ (1999). N-cadherin promotes motility in

human breast cancer cells regardless of their E-cadherin expression. J Cell Biol 147: 631-44.

Nishimura EK, Yoshida H, Kunisada T, Nishikawa SI (1999). Regulation of E- and P-cadherin

expression correlated with melanocyte migration and diversification. Dev Biol 215: 155-66.

Noe V, Fingleton B, Jacobs K, Crawford HC, Vermeulen S, Steelant W et al (2001). Release of

an invasion promoter E-cadherin fragment by matrilysin and stromelysin-1. J Cell Sci 114: 111-

118.

Nojima D, Nakajima K, Li LC, Franks J, Ribeiro-Filho L, Ishii N et al (2001). CpG methylation

of promoter region inactivates E-cadherin gene in renal cell carcinoma. Mol Carcinog 32: 19-27.

Nollet F, Kools P, van Roy F (2000). Phylogenetic analysis of the cadherin superfamily allows

identification of six major subfamilies besides several solitary members. J Mol Biol 299: 551-72.

Oeggerli M, Schraml P, Ruiz C, Bloch M, Novotny H, Mirlacher M et al (2006). E2F3 is the

main target gene of the 6p22 amplicon with high specificity for human bladder cancer. Oncogene

25: 6538-43.

Oka H, Shiozaki H, Kobayashi K, Inoue M, Tahara H, Kobayashi T et al (1993). Expression of

E-cadherin cell adhesion molecules in human breast cancer tissues and its relationship to

metastasis. Cancer Res 53: 1696-701.

Okazaki M, Takeshita S, Kawai S, Kikuno R, Tsujimura A, Kudo A et al (1994). Molecular

cloning and characterization of OB-cadherin, a new member of cadherin family expressed in

osteoblasts. J Biol Chem 269: 12092-8.

Page 96: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

83

Orlic M, Spencer CE, Wang L, Gallie BL (2006). Expression analysis of 6p22 genomic gain in

retinoblastoma. Genes Chromosomes Cancer 45: 72-82.

Pece S, Gutkind JS (2000). Signaling from E-cadherins to the MAPK pathway by the recruitment

and activation of epidermal growth factor receptors upon cell-cell contact formation. J Biol

Chem 275: 41227-33.

Peluso JJ (1997). Putative mechanism through which N-cadherin-mediated cell contact maintains

calcium homeostasis and thereby prevents ovarian cells from undergoing apoptosis. Biochem

Pharmacol 54: 847-53.

Peluso JJ, Pappalardo A, Trolice MP (1996). N-cadherin-mediated cell contact inhibits granulosa

cell apoptosis in a progesterone-independent manner. Endocrinology 137: 1196-203.

Perl AK, Wilgenbus P, Dahl U, Semb H, Christofori G (1998). A causal role for E-cadherin in

the transition from adenoma to carcinoma. Nature 392: 190-3.

Pishvaian MJ, Feltes CM, Thompson P, Bussemakers MJ, Schalken JA, Byers SW (1999).

Cadherin-11 is expressed in invasive breast cancer cell lines. Cancer Res 59: 947-52.

Pogosianz HE, Kuznetsova LE (1986). Nonrandom chromosomal changes in retinoblastomas.

Arch Geschwulstforsch 56: 135-43.

Potluri VR, Helson L, Ellsworth RM, Reid T, Gilbert F (1986). Chromosomal abnormalities in

human retinoblastoma. A review. Cancer 58: 663-71.

Prasad CP, Rath G, Mathur S, Bhatnagar D, Parshad R, Ralhan R (2009). Expression analysis of

E-cadherin, Slug and GSK3beta in invasive ductal carcinoma of breast. BMC Cancer 9: 325.

Ramburan A, Hadley GP, Govender D (2006). Expression of E-cadherin, cadherin-11, alpha-,

beta- and gamma-catenins in nephroblastomas: relationship with clinicopathological parameters,

prognostic factors and outcome. Pathology 38: 39-44.

Ramsay G, Stanton L, Schwab M, Bishop JM (1986). Human proto-oncogene N-myc encodes

nuclear proteins that bind DNA. Mol Cell Biol 6: 4450-7.

Rashid MG, Sanda MG, Vallorosi CJ, Rios-Doria J, Rubin MA, Day ML (2001).

Posttranslational truncation and inactivation of human E-cadherin distinguishes prostate cancer

from matched normal prostate. Cancer Res 61: 489-92.

Redston M (2003). Assembling a tumor progression model. Methods Mol Biol 223: 383-401.

Reynolds AB, Daniel JM, Mo YY, Wu J, Zhang Z (1996). The novel catenin p120cas binds

classical cadherins and induces an unusual morphological phenotype in NIH3T3 fibroblasts. Exp

Cell Res 225: 328-37.

Page 97: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

84

Richter S, Vandezande K, Chen N, Zhang K, Sutherland J, Anderson J et al (2003). Sensitive

and efficient detection of RB1 gene mutations enhances care for families with retinoblastoma.

Am J Hum Genet 72: 253-69.

Rios-Doria J, Day KC, Kuefer R, Rashid MG, Chinnaiyan AM, Rubin MA et al (2003). The role

of calpain in the proteolytic cleavage of E-cadherin in prostate and mammary epithelial cells. J

Biol Chem 278: 1372-9.

Robanus-Maandag E, Dekker M, van der Valk M, Carrozza ML, Jeanny JC, Dannenberg JH et al

(1998). p107 is a suppressor of retinoblastoma development in pRb-deficient mice. Genes Dev

12: 1599-609.

Roura S, Miravet S, Piedra J, Garcia de Herreros A, Dunach M (1999). Regulation of E-

cadherin/Catenin association by tyrosine phosphorylation. J Biol Chem 274: 36734-40.

Ruan G, Wedlich D, Koehler A (2006). Xenopus cadherin-6 regulates growth and epithelial

development of the retina. Mech Dev 123: 881-92.

Saramaki O, Visakorpi T (2007). Chromosomal aberrations in prostate cancer. Front Biosci 12:

3287-301.

Sarrio D, Rodriguez-Pinilla SM, Hardisson D, Cano A, Moreno-Bueno G, Palacios J (2008).

Epithelial-mesenchymal transition in breast cancer relates to the basal-like phenotype. Cancer

Res 68: 989-97.

Sato S, Nakamura Y, Tsuchiya E (1994). Difference of allelotype between squamous cell

carcinoma and adenocarcinoma of the lung. Cancer Res 54: 5652-5.

Shibata T, Ochiai A, Gotoh M, Machinami R, Hirohashi S (1996). Simultaneous expression of

cadherin-11 in signet-ring cell carcinoma and stromal cells of diffuse-type gastric cancer. Cancer

Lett 99: 147-53.

Shields CL, Shields JA, Needle M, de Potter P, Kheterpal S, Hamada A et al (1997). Combined

chemoreduction and adjuvant treatment for intraocular retinoblastoma. Ophthalmology 104:

2101-11.

Shintani Y, Hollingsworth MA, Wheelock MJ, Johnson KR (2006). Collagen I promotes

metastasis in pancreatic cancer by activating c-Jun NH(2)-terminal kinase 1 and up-regulating N-

cadherin expression. Cancer Res 66: 11745-53.

Shukla S, Maclennan GT, Hartman DJ, Fu P, Resnick MI, Gupta S (2007). Activation of PI3K-

Akt signaling pathway promotes prostate cancer cell invasion. Int J Cancer 121: 1424-32.

Singh AD, Santos CM, Shields CL, Shields JA, Eagle RC, Jr. (2000). Observations on 17

patients with retinocytoma. Arch Ophthalmol 118: 199-205.

Sitorus RS, Gumay S, van der Valk P (2009). The apoptotic paradox in retinoblastoma. Ann N Y

Acad Sci 1171: 77-86.

Page 98: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

85

Snijders AM, Nowee ME, Fridlyand J, Piek JM, Dorsman JC, Jain AN et al (2003). Genome-

wide-array-based comparative genomic hybridization reveals genetic homogeneity and frequent

copy number increases encompassing CCNE1 in fallopian tube carcinoma. Oncogene 22: 4281-

6.

Sobrin L, Hayden BC, Murray TG, Cicciarelli N, Scott IU, Hernandez E et al (2004). External

beam radiation "salvage" therapy in transgenic murine retinoblastoma. Arch Ophthalmol 122:

251-7.

Song G, Ouyang G, Bao S (2005). The activation of Akt/PKB signaling pathway and cell

survival. J Cell Mol Med 9: 59-71.

Sparkes RS, Sparkes MC, Wilson MG, Towner JW, Benedict W, Murphree AL et al (1980).

Regional assignment of genes for human esterase D and retinoblastoma to chromosome band

13q14. Science 208: 1042-4.

Sprick MR, Weigand MA, Rieser E, Rauch CT, Juo P, Blenis J et al (2000). FADD/MORT1 and

caspase-8 are recruited to TRAIL receptors 1 and 2 and are essential for apoptosis mediated by

TRAIL receptor 2. Immunity 12: 599-609.

Squire J, Gallie BL, Phillips RA (1985). A detailed analysis of chromosomal changes in heritable

and non-heritable retinoblastoma. Hum Genet 70: 291-301.

Squire J, Goddard AD, Canton M, Becker A, Phillips RA, Gallie BL (1986). Tumour induction

by the retinoblastoma mutation is independent of N-myc expression. Nature 322: 555-7.

Stappert J, Kemler R (1994). A short core region of E-cadherin is essential for catenin binding

and is highly phosphorylated. Cell Adhes Commun 2: 319-27.

Stemmler MP (2008). Cadherins in development and cancer. Mol Biosyst 4: 835-50.

Strathdee G (2002). Epigenetic versus genetic alterations in the inactivation of E-cadherin. Semin

Cancer Biol 12: 373-9.

Suarez F, Jockovich ME, Hernandez E, Feuer W, Parel JM, Murray TG (2007). Paclitaxel in the

treatment of retinal tumors of LH beta-Tag murine transgenic model of retinoblastoma. Invest

Ophthalmol Vis Sci 48: 3437-40.

Sullivan NJ, Sasser AK, Axel AE, Vesuna F, Raman V, Ramirez N et al (2009). Interleukin-6

induces an epithelial-mesenchymal transition phenotype in human breast cancer cells. Oncogene

28: 2940-7.

Suyama K, Shapiro I, Guttman M, Hazan RB (2002). A signaling pathway leading to metastasis

is controlled by N-cadherin and the FGF receptor. Cancer Cell 2: 301-14.

Takeichi M (1995). Morphogenetic roles of classic cadherins. Curr Opin Cell Biol 7: 619-27.

Page 99: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

86

Tamura G, Sakata K, Nishizuka S, Maesawa C, Suzuki Y, Iwaya T et al (1996a). Inactivation of

the E-cadherin gene in primary gastric carcinomas and gastric carcinoma cell lines. Jpn J Cancer

Res 87: 1153-9.

Tamura G, Sato K, Akiyama S, Tsuchiya T, Endoh Y, Usuba O et al (2001). Molecular

characterization of undifferentiated-type gastric carcinoma. Lab Invest 81: 593-8.

Tamura S, Shiozaki H, Miyata M, Kadowaki T, Inoue M, Matsui S et al (1996b). Decreased E-

cadherin expression is associated with haematogenous recurrence and poor prognosis in patients

with squamous cell carcinoma of the oesophagus. Br J Surg 83: 1608-14.

Tanihara H, Sano K, Heimark RL, St John T, Suzuki S (1994). Cloning of five human cadherins

clarifies characteristic features of cadherin extracellular domain and provides further evidence

for two structurally different types of cadherin. Cell Adhes Commun 2: 15-26.

Tell S, Yi H, Jockovich ME, Murray TG, Hackam AS (2006). The Wnt signaling pathway has

tumor suppressor properties in retinoblastoma. Biochem Biophys Res Commun 349: 261-9.

Tepass U, Truong K, Godt D, Ikura M, Peifer M (2000). Cadherins in embryonic and neural

morphogenesis. Nat Rev Mol Cell Biol 1: 91-100.

Thiery JP (2002). Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer 2:

442-54.

Thompson CB (1995). Apoptosis in the pathogenesis and treatment of disease. Science 267:

1456-62.

Tomita K, van Bokhoven A, van Leenders GJ, Ruijter ET, Jansen CF, Bussemakers MJ et al

(2000). Cadherin switching in human prostate cancer progression. Cancer Res 60: 3650-4.

Ts'o MO, Fine BS, Zimmerman LE (1969). The Flexner-Wintersteiner rosettes in

retinoblastoma. Arch Pathol 88: 664-71.

Tsui JY, Dalgard C, Van Quill KR, Lee L, Grossniklaus HE, Edelhauser HF et al (2008).

Subconjunctival topotecan in fibrin sealant in the treatment of transgenic murine retinoblastoma.

Invest Ophthalmol Vis Sci 49: 490-6.

Valencia X, Higgins JM, Kiener HP, Lee DM, Podrebarac TA, Dascher CC et al (2004).

Cadherin-11 provides specific cellular adhesion between fibroblast-like synoviocytes. J Exp Med

200: 1673-9.

Van Aken EH, Papeleu P, De Potter P, Bruyneel E, Philippe J, Seregard S et al (2002). Structure

and function of the N-cadherin/catenin complex in retinoblastoma. Invest Ophthalmol Vis Sci 43:

595-602.

Page 100: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

87

van der Wal JE, Hermsen MA, Gille HJ, Schouten-Van Meeteren NY, Moll AC, Imhof SM et al

(2003). Comparative genomic hybridisation divides retinoblastomas into a high and a low level

chromosomal instability group. J Clin Pathol 56: 26-30.

Visser M, Sijmons C, Bras J, Arceci RJ, Godfried M, Valentijn LJ et al (1997). Allelotype of

pediatric rhabdomyosarcoma. Oncogene 15: 1309-14.

Vleminckx K, Vakaet L, Jr., Mareel M, Fiers W, van Roy F (1991). Genetic manipulation of E-

cadherin expression by epithelial tumor cells reveals an invasion suppressor role. Cell 66: 107-

19.

Vogelstein B, Kinzler KW (1993). The multistep nature of cancer. Trends Genet 9: 138-41.

Volk T, Cohen O, Geiger B (1987). Formation of heterotypic adherens-type junctions between

L-CAM-containing liver cells and A-CAM-containing lens cells. Cell 50: 987-94.

von Burstin J, Eser S, Paul MC, Seidler B, Brandl M, Messer M et al (2009). E-cadherin

regulates metastasis of pancreatic cancer in vivo and is suppressed by a SNAIL/HDAC1/HDAC2

repressor complex. Gastroenterology 137: 361-71, 371 e1-5.

von Lindern M, Fornerod M, Soekarman N, van Baal S, Jaegle M, Hagemeijer A et al (1992).

Translocation t(6;9) in acute non-lymphocytic leukaemia results in the formation of a DEK-CAN

fusion gene. Baillieres Clin Haematol 5: 857-79.

Walczak H, Krammer PH (2000). The CD95 (APO-1/Fas) and the TRAIL (APO-2L) apoptosis

systems. Exp Cell Res 256: 58-66.

Waldmann T, Scholten I, Kappes F, Hu HG, Knippers R (2004). The DEK protein--an abundant

and ubiquitous constituent of mammalian chromatin. Gene 343: 1-9.

Wheelock MJ, Johnson KR (2003). Cadherins as modulators of cellular phenotype. Annu Rev

Cell Dev Biol 19: 207-35.

Windle JJ, Albert DM, O'Brien JM, Marcus DM, Disteche CM, Bernards R et al (1990).

Retinoblastoma in transgenic mice. Nature 343: 665-9.

Wippold FJ, 2nd, Perry A (2006). Neuropathology for the neuroradiologist: rosettes and

pseudorosettes. AJNR Am J Neuroradiol 27: 488-92.

Wohrn JC, Puelles L, Nakagawa S, Takeichi M, Redies C (1998). Cadherin expression in the

retina and retinofugal pathways of the chicken embryo. J Comp Neurol 396: 20-38.

Workman ML, Soukup SW (1984). Chromosome features of two retinoblastomas. Cancer Genet

Cytogenet 12: 365-70.

Wu L, de Bruin A, Saavedra HI, Starovic M, Trimboli A, Yang Y et al (2003). Extra-embryonic

function of Rb is essential for embryonic development and viability. Nature 421: 942-7.

Page 101: Cdh11 Acts as a Tumor Suppressor in a Murine ... · ii Cdh11 Acts as a Tumor Suppressor in Murine Retinoblastoma Model by Facilitating Tumor Cell Death Christine Yurkowski Master

88

Wu Q, Li Z, Lin H, Han L, Liu S, Lin Z (2008). DEK overexpression in uterine cervical cancers.

Pathol Int 58: 378-82.

Wyllie AH, Kerr JF, Currie AR (1980). Cell death: the significance of apoptosis. Int Rev Cytol

68: 251-306.

Yang J, Mani SA, Donaher JL, Ramaswamy S, Itzykson RA, Come C et al (2004). Twist, a

master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 117: 927-39.

Yang Z, Zhang X, Gang H, Li X, Li Z, Wang T et al (2007). Up-regulation of gastric cancer cell

invasion by Twist is accompanied by N-cadherin and fibronectin expression. Biochem Biophys

Res Commun 358: 925-30.

Yilmaz M, Christofori G (2009). EMT, the cytoskeleton, and cancer cell invasion. Cancer

Metastasis Rev 28: 15-33.

Yoshida H, Kong YY, Yoshida R, Elia AJ, Hakem A, Hakem R et al (1998). Apaf1 is required

for mitochondrial pathways of apoptosis and brain development. Cell 94: 739-50.

Young RW (1985a). Cell differentiation in the retina of the mouse. Anat Rec 212: 199-205.

Young RW (1985b). Cell proliferation during postnatal development of the retina in the mouse.

Brain Res 353: 229-39.

Zhang J, Schweers B, Dyer MA (2004). The first knockout mouse model of retinoblastoma. Cell

Cycle 3: 952-9.

Zhao YF, Zhang YG, Tian XX, Juan D, Jie Z (2007). Aberrant methylation of multiple genes in

gastric carcinomas. Int J Surg Pathol 15: 242-51.

Zhou R, Skalli O (2000). Identification of cadherin-11 down-regulation as a common response of

astrocytoma cells to transforming growth factor-alpha. Differentiation 66: 165-72.

Zhou Y, Snead ML (2008). Derivation of cranial neural crest-like cells from human embryonic

stem cells. Biochem Biophys Res Commun 376: 542-7.

Zielinski B, Gratias S, Toedt G, Mendrzyk F, Stange DE, Radlwimmer B et al (2005). Detection

of chromosomal imbalances in retinoblastoma by matrix-based comparative genomic

hybridization. Genes Chromosomes Cancer 43: 294-301.

Zou CY, Wang J, Shen JN, Huang G, Jin S, Yin JQ et al (2008). Establishment and

characteristics of two syngeneic human osteosarcoma cell lines from primary tumor and skip

metastases. Acta Pharmacol Sin 29: 325-32.