GENETIC VARIATION AND COMPLEX DISEASE: THE EXAMINATION OF AN X-LINKED DISORDER AND A MULTIFACTORIAL DISEASE

DISSERTATION

Presented in Partial Fulfillment of the Requirements for

the Degree Doctor of Philosophy in the Graduate

School of The Ohio State University

By

Catherine Elise Cottrell

*****

The Ohio State University 2007

Dissertation Committee: Approved by Dr. Julie M. Gastier-Foster, Advisor

Dr. John Bauer ______Dr. Gail Herman Advisor Graduate Program in Dr. Susan R. Mallery Integrated Biomedical Sciences

ABSTRACT

Two genetic studies are presented in this dissertation. The studies share a common theme of the influence of genetic variability in complex disease. The first study is entitled, “The Association of Antioxidant-Related Gene Polymorphisms and Second

Primary Malignant Neoplasms in Pediatric Hodgkin .”

Improved treatment strategies in pediatric (HL) have

resulted in a cure rate approaching 95%, yet the development of a second primary

malignant neoplasm (SMN) is a risk for long-term survivors. In a pediatric HL

population, between 6-26% of patients will develop a SMN within 30 years following

treatment. The etiology of HL is still largely unknown, as is the cause of certain types of

SMNs.

Oxidative stress has been linked to the development of cancer due to the

damaging effects of reactive species (ROS) on DNA, , and proteins. During

periods of extended oxidative stress, the damaging effects of ROS are likely to increase

which in turn raises the risk of genetic change. In a multi-stage model of tumorigenesis,

genetic change is considered to be the initiating event of cancer development. Under

normal circumstances, antioxidant enzymes within the cell mitigate the effects of ROS by

converting reactive species into non-toxic molecules. Mechanisms within the cell

maintain a steady state balance between ROS and antioxidant enzyme levels. We

ii hypothesize that individuals predisposed to lower levels of antioxidant enzyme activity

due to polymorphic variants within those genes may be at risk for increased damage

caused by ROS. Although decreased amounts of antioxidant enzymes have been found in

a variety of cancers, a correlation between polymorphisms in antioxidant genes and

predisposition to secondary cancer has not been studied.

The purpose this study was to assess the association of antioxidant gene alleles with the risk of developing a SMN in HL survivors. DNA samples were obtained from

768 HL patients enrolled in the Childhood Cancer Survivor Study (CCSS). The samples were genotyped for 90 polymorphisms in antioxidant related genes including SOD, GPX,

NOS, CAT, and CYP2C9. Statistical analysis methods to determine risk of developing a

SMN included association, haplotype, and multiple regression models.

In our HL cohort, 131 patients developed a SMN. An additional 117 patients

developed nonmelanoma skin cancer, and 34 patients developed 2 SMNs. Out of the 36

SNPs that were included in the final analysis, 4 SNPs in the GPX1, GPX3, GPX4, and

SOD2 genes, were potentially suggestive (p<0.05) of an association between genotype

and the development of a SMN. A general trend observed in the genotyping data

suggested that an increased risk of SMN was conferred by the presence of the minor

allele, while a decreased risk of SMN was conferred by the presence of the major allele in

the population. Replication studies are necessary, though it is notable that polymorphisms within the GPX family may be associated with the development SMN in our cohort. Additionally, ratios were only moderately increased in this population suggesting that the development of a SMN in a pediatric HL is likely multi-factorial in nature. The elucidation of critical genes involved in SMN development could influence

iii patient follow-up and intervention strategies, and potentially aid in the prevention of

SMN.

The second study in this thesis is entitled, “Atypical X-Inactivation in an X;1

Translocation Patient Diagnosed with Otopalatodigital Syndrome.” X-chromosome inactivation (XCI) is an epigenetic process used to regulate gene dosage in mammalian

females by silencing one X-chromosome. While the pattern of XCI is typically random

in normal females, abnormalities of the X-chromosome may result in skewing due to

disadvantaged cell growth. We describe a female patient with an X;1 translocation

[46,X, t(X;1)(q28;q21)dn] and unusual pattern of XCI who was clinically diagnosed with

Otopalatodigital syndrome (OPD) type 1. There was complete skewing of XCI in the

patient, along with the atypical findings of an active normal X-chromosome and an

inactive derivative X. An X-linked disorder, OPD1 is characterized by multiple

congenital anomalies including skeletal abnormalities, craniofacial defects, and hearing

loss. Mutations within the FLNA gene (Xq28) are known to cause OPD, though none

were detected in our patient. Additionally, no abnormalities in FLNA mRNA or protein

were detected in our patient. Characterization of the translocation revealed that the

patient’s Xq28 breakpoint interrupts the DKC1 gene, located 400kB distal to FLNA.

Molecular analysis of the breakpoint region revealed functional disomy of Xq28 genes

distal to DKC1. No monosomy of 1q genes was detected. Possible explanations for the

patient’s phenotype include a position effect due to the translocation breakpoint, an

undetected FLNA-related mutation, or altered gene dosage due to consequences of

atypical XCI.

iv

Dedicated to Andy Thank you for your love, constant support, and words of encouragement.

&

To my parents, Charles and Denise Thank you for inspiring me, fostering a sense of creativity, and for a lifetime of love and support.

v

ACKNOWLEDGMENTS

It is with deepest gratitude that I would like to thank my advisor, Dr. Julie

Gastier-Foster for her dedication and commitment to me on both a personal and professional level. You are a constant source of inspiration and an exceptional mentor.

Thank you for your patience, guidance, and support over the years. Most of all, I thank you for your time, for the countless hours spent teaching me, and for the direction you provided so that I could mature on both a scientific and personal level.

I would like to thank my committee members, Dr. Gail Herman, Dr. John Bauer, and Dr. Susan Mallery for your commitment to teaching, dedication to students, and exceptional guidance.

I would like to express my sincere appreciation for Dr. Steve Qualman. Thank you for guiding me to choose a wonderful lab in which to complete my dissertation.

Thank you for introducing me to Julie, because without her none of this would be possible. Finally, thank you most of all for your dedication, your commitment to research and teaching, and your passion in supporting Children’s Hospital.

I would like to thank Dr. Allan Yates and Christine Kerr for your support of the

IBGP program, for your devotion to the students, and for your sincere desire to see each of us succeed.

vi I would especially like to thank all of the technologists and staff within the

Molecular and Cytogenetics Laboratories at Children’s. Thank you for accepting me into

the lab, and for sharing your time and resources with me. Thank you especially to Jessica

who made me feel welcome when I first began and for your help along the way. Thanks

to Beth and Morgan for teaching me the basics of FISH, cell harvests and for your

expertise in taking wonderful photos on the scope. A special thank you goes to Eileen for

your unwavering support, your friendship, and your sense of humor. I don’t think I could have made it through this without you.

Finally, I would like to thank my family and friends for your support throughout

this process. Thank you to Jen for understanding what I was going though when no one

else did. Thank you for graduating ahead of me, so then I could ask you all sorts of

questions on finishing up. Thank you for extended chats over coffee and for going to

Buckeye football games with me. Thank you to Colleen for being a great sister. Thank you for your support and kind words, and for feeding the cats when I was working late or away on weekends. Thank you especially to Andy for your love and encouragement.

Thank you for driving long hours to see me, for cheering me up during hard times, and for always making me laugh, especially when this day seemed very far away. Thank you to my parents for sparking my interest in science early on. Thank you for those kitchen science experiments, grade school science fair ideas, and for taking me to your lab when I was very young. Most of all, thank you for your love and guidance over the years.

vii

VITA

August 3, 1979...... Born – Columbus, OH

June 2001...... B.S. Molecular Genetics The Ohio State University

2001 – 2007...... Graduate Fellow The Ohio State University.

AWARDS

2004 ...... Graduate Student Research Award Columbus Children’s Research Institute Annual Research Conference

FELLOWSHIPS

2001 ...... University Fellowship The Ohio State University

2001-2005 ...... Molecular Life Sciences Fellowship The Ohio State University

2002-2007 ...... General Mason Fellowship Columbus Children’s Hospital

FIELD OF STUDY

Major Field: Integrated Biomedical Sciences Specialization: Genetics

viii

TABLE OF CONTENTS

P a g e

Abstract...... ii

Dedication...... v

Acknowledgments ...... vi

Vita ...... viii

List of Tables...... xiv

List of Figures ...... xv

Introduction ...... 1

Section I: The Association of Antioxidant Related Gene Polymorphisms and Second Primary Malignant Neoplasm in a Pediatric Hodgkin’s Lymphoma Population

Chapters:

1. Hodgkin Lymphoma and the Childhood Cancer Survivor Study ...... 5

1.1 Introduction to Hodgkin Lymphoma ...... 5

1.2 Classification of Hodgkin Lymphoma ...... 6

1.3 Risk Factors and Causative Agents in Hodgkin Lymphoma ...... 6

1.4 Treatment of Hodgkin Lymphoma: Maximizing Efficacy while Minimizing Risk ...... 8

ix 1.5 The Childhood Cancer Survivor Study ...... 9

1.5.1 Study Design and Eligibility ...... 9

1.5.2 Study Participation ...... 10

1.5.3 HL Patient Demographics within the CCSS ...... 11

1.6 Genetic Susceptibility to SMN in Cancer Survivors ...... 11

2. Introduction to Reactive Species, Antioxidant Enzymes and the Relationship between Oxidative Stress and Malignancy ...... 20

2.1 Reactive Species and Antioxidants ...... 20

2.1.1 Reactive Oxygen Species ...... 20

2.1.2 Antioxidant Enzymes ...... 21

2.1.3 Reactive Nitrogen Species ...... 22

2.2 Types of Antioxidant Enzymes ...... 23

2.2.1 Superoxide Dismutase ...... 23

2.2.2 Glutathione Peroxidase...... 23

2.2.3 Catalase...... 25

2.3 Oxidative Stress and Cancer ...... 25

2.3.1 A Multi-Stage Model of Carcinogenesis ...... 25

2.3.2 Initiation ...... 26

2.3.3 Promotion ...... 26

2.3.4 Reactive Species: A Balancing Act of Tumor Promotion or Tumor Suppression . . . 26

2.3.5 Progression ...... 27

2.3.6 Antioxidant Enzymes and Malignancy...... 27

x 3. The Association of Antioxidant Related Gene Polymorphisms and Second Primary Malignant Neoplasms in a Pediatric Hodgkin Lymphoma Population ...... 37

3.1 Abstract ...... 37

3.2 Introduction ...... 39

3.3 Materials and Methods ...... 41

3.3.1 The Childhood Cancer Survivor Study ...... 41

3.3.2 Buccal Cell Collection ...... 42

3.3.3 Genotyping Methodology...... 43

3.3.4 Statistical Analysis Methods...... 45

3.4 Results ...... 47

3.5 Discussion ...... 51

Section II: Atypical X Chromosome Inactivation in an X;1 Translocation Patient Diagnosed with Otopalatodigital Syndrome

4. X Chromosome Inactivation and X;Autosome Translocations ...... 88

4.1 X Chromosome Inactivation ...... 88

4.1.1 Introduction ...... 88

4.1.2 Genes which Escape X-Inactivation ...... 89

4.2 X;Autosome Translocations ...... 90

4.2.1 Introduction to Translocations ...... 90

4.2.2 Balanced X;Autosome Translocations ...... 91

4.2.3 Phenotypic Consequences Translocation Breakpoint Location . . .91

4.2.4 XCI in a t(X;A) Carrier ...... 92

4.2.5 The Impact of the Mode of Inheritance in a t(X;A) Carrier...... 93

xi 5. Otopalatodigital Syndrome Spectrum Disorders and Filamin A ...... 94

5.1 Introduction to Otopalatodigital Syndrome ...... 94

5.2 Phenotypic Features of OPD ...... 94

5.3 Molecular Characterization of the OPD-Spectrum Disorders ...... 95

5.4 Filamin A Protein ...... 97

5.5 OPSD Genotype – Phenotype Correlations ...... 99

5.6 X-Chromosome Inactivation and OPSD...... 99

6. Atypical X Chromosome Inactivation in an X;1 Translocation Patient Diagnosed with Otopalatodigital Syndrome ...... 105

6.1 Abstract ...... 105

6.2 Introduction ...... 106

6.3 Materials and Methods ...... 108

6.3.1 Subjects and Consent ...... 108

6.3.2 Tissue Sources, Cell Lines, DNA and RNA Preparations ...... 108

6.3.3 Molecular and Cytogenetic Breakpoint Mapping ...... 109

6.3.4 X-Inactivation Analysis ...... 109

6.3.5 Expression – RT-PCR ...... 110

6.3.6 Sequencing ...... 110

6.3.7 Northern Blot ...... 111

6.3.8 Expression Array Analysis ...... 111

6.3.9 Comparative Genomic Hybridization ...... 112

6.3.10 Western Blot Analysis ...... 112

6.4 Results ...... 113

xii 6.4.1 Clinical Report ...... 113

6.4.2 Breakpoint Characterization ...... 114

6.4.3 X-Chromosome Inactivation Status ...... 115

6.4.4 RT-PCR Analysis ...... 115

6.4.5 Molecular Analysis of FLNA ...... 116

6.4.6 Expression Array Analysis ...... 118

6.4.7 Comparative Genomic Hybridization ...... 118

6.5 Discussion ...... 118

Appendix ...... 154

References ...... 155

xiii

LIST OF TABLES

Table Page

1.1 Composition of Participants within the CCSS Cohort ...... 16

1.2 Type of SMN in HL Survivors within the CCSS Cohort...... 18

3.1 Type of SMN/NMSC within the Subset of Genotyped HL Survivors. . .66

3.2 SNPs Included in the Final Statistical Analysis of the HL Cohort . . . . . 68

3.3 Associations between SMN or NMSC and Genotype by Cox Regression Analysis ...... 71

3.4 A Summarized List of Associations by Cox Regression Analysis . . . . . 76

3.5 Associations between SMN Classification and Genotype by Cox Regression Analysis ...... 78

3.6 A Summary of Association Results by Cox Regression Analysis Classified by Type of SMN or NMSC...... 81

3.7 Associations between SMN or NMSC and Genotype by Poisson Multivariate Regression Analysis...... 83

3.8 Haplotype Analysis using Poisson Regression Modeling ...... 85

5.1 Phenotypic Features of FLNA-Related Disorders ...... 103

6.1 Primer Sequences...... 145

6.2 Refinement of the X;1 Translocation Breakpoint by FISH Mapping. . .148

6.3 RT-PCR Analysis ...... 150

6.4 Combined RT-PCR and Array Results for Xq28 Genes...... 152

xiv

LIST OF FIGURES

Figure Page

1.1 Annual Incidence Rates of Hodgkin Lymphoma ...... 14

2.1 Chemical Reactions Involving ROS and Antioxidant Enzymes...... 29

2.2 Reactive Species Interact to Form an Oxidizing Free Radical ...... 31

2.3 A Multi-Stage Model of Tumorigenesis Relative to Oxidative Stress. . .33

2.4 Cellular Response to ROS During Times of Oxidative Stress...... 35

3.1 SNPlex Genotyping Chemistry ...... 58

3.2 GeneMapper Graphical Plots ...... 60

3.3 GeneMapper Samples Plot ...... 62

3.4 Causes of SNP Attrition in the SNPlex Genotyping Method...... 64

5.1 A Model of the Filamin A Dimer and Location of Pathogenic Mutations ...... 101

6.1 X;1 Translocation Breakpoint...... 125

6.2 Clinical Features of the Patient...... 127

6.3 Fluorescence In-Situ Hybridization Analysis...... 129

6.4 Map of the Xq28 Breakpoint Region...... 131

6.5 X-Chromosome Inactivation Analysis...... 133

6.6 Disomic Expression of the FLNA Gene...... 135

xv

6.7 Sequence Analysis of a 3’UTR FLNA Polymorphism ...... 137

6.8 Mfold Predictions of Optimal Secondary Structure...... 139

6.9 Northern Blot Analysis of FLNA Transcripts...... 141

6.10 Western Blot Analysis of Filamin A ...... 143

xvi

LIST OF ABBREVIATIONS

α Alpha

β Beta

AB Applied Biosystems

°C degrees Celsius

CAT Catalase

CCSS Childhood Cancer Survivor Study

CI Confidence Interval

der(X) Derivative X Chromosome

GEE Generalized Estimating Equation

GPX Glutathione Peroxidase

GST Glutathione S-Transferase

H2O2 Hydrogen Peroxide

HL Hodgkin Lymphoma

HRS Hodgkin Reed-Sternberg Cell

NHL Non-Hodgkin Lymphoma

NMSC Non-Melanoma Skin Cancer

NO• Nitric Oxide

xvii NOS Nitric Oxide Synthase

O2 Oxygen

•– O2 Superoxide Anion

•OH Hydroxy Radical

ONOO– Peroxynitrite Anion

OPD Otopalatodigital Syndrome

OPSD Otopalatodigital Syndrome Spectrum Disorders

RNS Reactive Nitrogen Species

ROS Reactive Oxygen Species

RS Reactive Species

SIR Standardized Incidence Ratio

SMN Second Primary Malignant Neoplasm

SMR Standardized Mortality Ratio

SNP Single Nucleotide Polymorphism

SOD Superoxide Dismutase t(X;A) X;Autosome Translocation

XCI X-Chromosome Inactivation

Xi Inactive X-Chromosome

Xa Active X-Chromosome

xviii

INTRODUCTION Thesis Composition and Objectives

Beginning with the Watson and Crick discovery of the double helix in 1953, major milestones in the field of genetics have slowly begun to reveal the secrets of the genome. More recent advances such as the completion of the Human Genome Project

(2003) resulted in a thorough guide to our genetic code, and subsequent studies using

HapMap data have been used to assess population variance. From data compiled so far, findings have been particularly striking in regard to genetic variation. Results suggest that the genomes between two individuals may differ by at least 1%. This indicates that the human genome contains far more variability than previously thought. Variability at the level of a single nucleotide polymorphism, a short tandem repeat, or even a chromosomal rearrangement adds to the complexity and function of the human genome.

It remains a challenge for investigators to decode the secrets of the genome, and in turn to decipher how variability influences phenotype. Of perhaps the greatest clinical relevance is to determine how variability influences the risk of genetic disease. Progress on this front could improve patient screening, genetic counseling, and comprehensive care.

This thesis is a compilation of two separate research projects that were completed

during the course of my graduate study. Though each project is vastly different, a

common theme of genetic variation in complex disease is present.

1 Section one of this thesis relates to the Hodgkin Lymphoma project. Background

information regarding HL, antioxidants, and SMNs are included in chapters 1 and 2 to

introduce the reader to the field of study prior to presenting the results and conclusions.

The HL project began in 2004 using samples that were collected from pediatric cancer

survivors enrolled in the Childhood Cancer Survivor Study (CCSS). Cancer survivors within the CCSS represent one of the largest and most well characterized cohorts within

North America. HL was studied exclusively, as survivors have the greatest risk of

developing a SMN following cancer treatment, and because we were granted access to

patient buccal cell DNA from the CCSS. While previous genetic studies of HL and SMN

focused on genes involved in DNA repair and detoxification, we chose to focus on a

genetic analysis of polymorphisms within antioxidant related genes. The results of the

study are presented in chapter 3.

Section two of my thesis relates to the OPD project. This research project has

evolved greatly since its inception in 2002. At that time the causative gene for OPD

syndrome had not yet been cloned, but was localized to Xq28. By chance, a patient

diagnosed with OPD1 and having an X;1 translocation was currently being followed by

the genetics clinic and presented the opportunity to attempt to clone the gene. The

project began following parental and IRB approval. Concurrent with our efforts to clone

the gene, another research group in Europe was working toward the same goal. In 2003

Robertson et al. reported mutations within the FLNA gene to be causative for the OPD

spectrum disorders. Based on this finding, the focus of our project shifted to determine

the etiology of OPD1 in our patient. An introduction to X chromosome inactivation and

X;autosome translocations are presented in chapter 4, while background information

2 regarding OPD and Filamin A is presented in chapter 5. The results of this study are presented in Chapter 6.

The objectives of this thesis are to provide a thorough explanation of each independent project, to draw relevant conclusions from the results, and to identify future aims of this research.

3 SECTION I

THE ASSOCIATION OF ANTIOXIDANT RELATED GENE POLYMORPHISMS AND SECOND PRIMARY MALIGNANT NEOPLASM IN A PEDIATRIC HODGKIN LYMPHOMA POPULATION

4

CHAPTER 1

INTRODUCTION TO HODGKIN LYMPHOMA AND THE CHILDHOOD CANCER SURVIVOR STUDY

1.1: Introduction to Hodgkin Lymphoma

Hodgkin Lymphoma (HL) is a malignancy of the lymphatic system that first

presents as a painless swelling of the lymph nodes. Fever, night sweats, loss and

itching are other common symptoms. Disease progression occurs through the lymphatic

vessels as malignant cells spread into other lymph nodes or adjacent organs. The age of

incidence of HL follows a bimodal distribution which peaks first in young adults (15-34

years) and again in older individuals (>60 years) (Figure 1.1).1 Children and adolescents under the age of 20 represent approximately 12% of all HL diagnoses.1 Presently, the

five year event free survival of HL patients is greater than 90%.2, 3

The etiology of HL is unknown but may be caused by interactions between genes, the environment and infectious agents. Risk factors for the disease include age, exposure to Epstein-Barr virus, immune deficiency or a family history of HL. Unlike many other leukemias or , an underlying cytogenetic abnormality is not detected in HL patients, though this may be due to ascertainment bias. The neoplastic component of HL is believed to be the Hodgkin and Reed-Sternberg (HRS) cell, which represents only 0.1-

5 1.0% of cells in tumor tissue.4, 5 The cellular origin of the HRS cell was only recently elucidated, as immunological studies revealed a clonal rearrangement of the immunoglobulin genes indicative of B-cell origin.6-8 HRS cells are thought to be highly interactive within the cellular environment. It has been hypothesized that the ability of

HRS cells to mimic antigen-presenting immune cells results in the pathogenesis of HL.9

1.2: Classification of Hodgkin Lymphoma

Two major subtypes of HL exist under the World Health Organization’s

classification scheme; classical HL and nodular lymphocyte predominant HL.10, 11

Classical HL is further divided into four groups including, nodular sclerosing (60% of pediatric cases), mixed cellularity (30% of pediatric cases), lymphocyte rich (10% of pediatric cases), and lymphocyte depleted (extremely rare in children).12 In most cases, all forms of classical HL are treated similarly, usually including multi-drug and radiation.13, 14 Nodular lymphocyte predominant HL represents only 5% of HL cases and typically presents early on with only local involvement. Very few or no HRS cells are seen in this type of HL.

1.3: Risk Factors and Causative Agents in Hodgkin Lymphoma

No definitive causative agents have been identified in HL, though certain risk factors have been linked to the disease. Exposure to the Epstein-Barr virus is associated with about 40% of HL cases, but it is unclear how the virus is involved in the development of disease.15 Serology studies have demonstrated that HL patients have

higher antibody titers to EBV as compared to controls.16 EBV RNA and protein have been found to localize to HRS cells in HL patients and the virus is known to be transcriptionally active.17 Additional studies have demonstrated that EBV encodes an

6 oncogene (LMP-1) capable of transforming cells in-vitro which gives some insight as to

how the virus may function to promote a malignant phenotype in vivo.18

Family history is another potential risk factor in the development of HL. In a study by

Mack et al., concordance for HL was demonstrated in monozygotic twins, but not in dizygotic twins, which suggests a genetic component is involved in the etiology of disease. The study revealed that monozygotic twins had a 99 fold excess risk for disease

(Standardized Incidence Ratio (SIR) 99; 95% Confidence Interval (CI) 48-182).19 In an analysis of the Childhood Cancer Survivor Study cohort, non-twin full siblings of a HL survivor had a 2 fold increased risk for the development of disease (SIR 2.3; 95% CI 1.6-

3.1).20

A dysregulation of immune function is associated with HL. Patients affected with

HIV/AIDS and those taking immunosuppressive drugs because of organ transplant or

auto-immune disease are at greater risk for HL.10 Additionally, HL patients have been shown to demonstrate a lack of immune response known as anergy. This was first observed as HL patients failed to generate a cuteneous reaction to tuberculin despite having an active form of the disease.21 The frequency of anergy has been correlated with

HL disease severity in patients, with a lack of immune response observed more

frequently in patients in the later stages of disease. Upon remission of HL, it has been

documented that patients recover the ability to react to an antigen.22 The delayed immune response in HL patients is of clinical importance as patients are extremely susceptible to the development of infection. Despite extensive study, it is unclear whether impaired cellular immunity serves as a predisposing factor or result of HL.22

7 1.4: Treatment of HL: Maximizing Efficacy While Minimizing Risk

With 5 year event free survival rates of greater than 90%, challenges in the treatment of HL have shifted to minimize secondary effects of without reducing overall efficacy. Late effects of treatment are associated with increased morbidity and mortality of HL survivors, and frequently include infertility, cardiopulmonary disease, and second primary malignant neoplasms (SMN), in addition to other complications.23

The highest mortality risk in HL patients is due to a SMN.24 Reports of cumulative risk

for a SMN vary from 6% to 26%.14, 23, 25-32 The difference in risk is likely due to self-

reporting of SMNs, varying length of follow-up time, inclusion or exclusion of non-

melanoma skin cancer as a SMN, and differences in therapy regimens.

HL survivors face treatment-related sequelae caused by radiation and

chemotherapy agents used to treat their primary disease. With several decades of

treatment and research experience, investigators have become acutely aware of the

complications associated with late effects and have devised strategies to minimize

exposure to toxic agents. It is particularly desirable to limit exposure of the most

damaging agents including mechlorethamine and other alkylating drugs, ,

and radiation. Elimination or dosage reduction of certain drugs may improve sequelae;

such as second hematological malignancy (associated with mechlorethamine), sterility or

premature ovarian failure (associated with alkylating agents), and cardiopulmonary

disease (associated with anthracyclines).33-36 Radiotherapy treatment effects such as growth impairment of bone and soft tissue, thyroid dysfunction, and second malignancy

(particularly breast cancer) could be ameliorated by limiting radiation dose and field or by omission of this modality altogether.27, 37-40

8 treatment strategies for pediatric HL utilize combination chemotherapy

and low dose radiotherapy to involved lymph nodes.41 Low risk patients may now be placed on abbreviated chemotherapy regimens with radiotherapy omitted if complete response occurs.14 Intermediate and high risk patients are placed into very intensive

chemotherapy regimens with or without radiation treatment.41

1.5: The Childhood Cancer Survivor Study

Funded by the National Cancer Institute, the Childhood Cancer Survivor Study

(CCSS) was initiated in 1993 to examine the long term effects of cancer and its treatment

in a cohort of pediatric survivors. Objectives of the study were twofold: to accrue

knowledge in order to facilitate the design of new treatment protocols and intervention

strategies, and to educate survivors about their diagnosis, treatment, appropriate follow-

up and long term health risks.

1.5.1: Study Design and Eligibility

The CCSS represents the collaborative effort of 27 participating clinical centers

in the US and Canada and was established to follow a cohort of pediatric and adolescent

survivors of cancer. In order to assemble the cohort, participating institutions contacted

eligible patients who met the following criteria: (a) diagnosis of leukemia, central

nervous system malignancy, HL, non-HL, neuroblastoma, soft tissue sarcoma, kidney or

bone cancer; (b) date of diagnosis between January 1, 1970 and December 31, 1986; (c)

age 0-21 at the time of diagnosis; (d) survival 5 years past initial diagnosis; (e) English or

Spanish speaking (for data collection purposes); and (f) a US or Canadian resident.

Patient contact and recruitment began in 1994.42

9 Following recruitment into the study, survivors or family members (if the patient

was deceased, or a minor) were sent a comprehensive baseline questionnaire containing

289 questions. Subject matter was wide-ranging and included questions about patient

demographics, health information, depression, pain, primary cancer recurrence,

development of a SMN, marital status, pregnancy history, education, employment,

insurance, and family history. Siblings were also contacted from a random sampling of

study participants in order to serve as a comparison group.

1.5.2: Study Participation

Based on the initial subject registry completed by the clinical centers, a total of

20,276 survivors met eligibility requirements for the cohort. Of that group 17,280

subjects were successfully contacted, with the remaining survivors lost to follow-up. A

total of 14,054 survivors completed the study questionnaire representing 69.3% of the eligible cohort.42 Participating survivors were asked to sign a consent form to allow for medical record abstraction. Detailed information regarding cancer treatment, including chemotherapy, radiation, and surgical procedures was documented for each member of

the cohort. In the time since the baseline questionnaire was completed, additional follow-

up questionnaires have been sent to study participants to update cohort characteristics.

Demographic characteristics within the cohort indicated that greater than half (54%) of

CCSS participants were male, and the majority (87%) were classified as Caucasian.42

Detailed participant information including cancer diagnosis and treatment as well as cohort demographic data is shown in table 1.1.

10 1.5.3: HL Patient Demographics within the CCSS

HL patients represented 13% of the eligible CCSS cohort with a total of 2718

survivors. Of this group, 1906 HL survivors chose to participate in the study, and

complete medical records plus baseline data was available for 1815 HL survivors.42

Significantly increased morbidity and mortality was observed in the HL cohort.

Within this group, deaths occurred 8 times more often than expected relative to US

mortality rates (Standardized Mortality Ratio [SMR] 8.3; 95% CI 7.4-9.2)24. Aside from primary cancer recurrence, the highest rate of mortality was due to a SMN. Out of all cancer diagnoses within the CCSS, patients with HL were found to be at the greatest risk for death caused by a SMN (SMR 24.0; 95%CI 19.2-29.7).24

Of 1815 HL survivors within the CCSS cohort, 111 (6%) had a confirmed

SMN.23 Observed SMNs included leukemia, non-Hodgkin lymphoma (NHL), CNS tumor, breast cancer, bone cancer, sarcoma, thyroid cancer and melanoma (Table 1.2).

The most common solid tumor in HL survivors was breast cancer, representing 32% of all SMNs. Little is known about the differences between HL patients who develop a

SMN in comparison to those who do not.

1.6: Genetic susceptibility to SMN in Cancer Survivors

Aside from primary cancer recurrence, the greatest threat to pediatric cancer survivors is the development of a SMN. In a review of the CCSS cohort, 91.9% of individuals without a SMN were alive at time of study (2001), in comparison to 59.4% who developed a SMN.23 In order to improve screening practices, follow-up care and to enhance the quality of life for cancer survivors, investigators have attempted to determine the etiology of SMNs.

11 The development of some SMNs is thought to be related to a patient’s response to

therapy. Acute myelogenous leukemia (AML) is observed within 4-7 years of primary

cancer diagnosis and is associated with chemotherapy.23, 36 Solid tumors such as breast and thyroid cancer are associated with radiotherapy and typically occur greater than 5 years past primary cancer diagnosis. Non-Hodgkin Lymphoma does not appear to be associated more strongly with either chemotherapy or radiation, and like many other

SMNs, the etiology is unknown.23 Family history may also impact the risk of SMN in

cancer survivors. In a recent study by Nichols et al., a positive family history of cancer

(3 or more relatives with cancer and spanning 2 generations) was observed in 21% of HL

survivors who developed a SMN in comparison to 4% who did not develop a SMN.43

Findings such as these support the hypothesis that an underlying genetic predisposition to malignancy may exist.

Therapy-related SMNs have been studied to determine if there is an underlying genetic susceptibility in affected patients. Of particular interest are genes involved in

DNA repair and drug . In a study by Mertens et al, polymorphisms within the

XRCC1 and glutathione S-transferase (GST) genes were studied in relation to radiation therapy related SMNs in HL patients enrolled in the CCSS.44

XRCC1 functions within the DNA base excision repair pathway to correct

damaged bases and to repair single strand breaks.45 Individual bases may be damaged by

deamination, alkyation, or oxidation caused by a number of factors, including radiation

and xenobiotic exposure (particularly alkylating agents).45, 46 Polymorphisms within

XRCC1 resulting in amino acid substitutions, including Arg399Gln, have been previously

12 studied. Results have been contradictory, with the glutamine allele associated with a

decreased DNA repair capacity in some studies and a protective effect in others.47-50

GST genes code for enzymes involved in conjugation reactions to detoxify the highly reactive intermediate species formed during drug metabolism. The GST gene family includes 5 isoenzymes, two of which (GSTM1 and GSTT1) are commonly deleted resulting in no functional protein production. In a Caucasian population, 50% of individuals have a homozygous deletion of GSTM1, and 20% of individuals have a homozygous deletion of GSTT1.51, 52

The genotype of the XRCC1 polymorphism (Arg399Gln) and the presence or absence of GSTM1 and GSTT1 alleles were determined in HL survivors.44 A

nonsignificant increased risk of breast cancer was observed in HL survivors heterozygous

for the glutamine allele (Arg/Gln) at position 399 in XRCC1. Individuals with a homozygous deletion of GSTM1 were at an elevated risk of SMN with an odds ratio of

1.5 (95% CI 1.0-2.3). Increased risk for SMN in relation to XRCC1, GSTM1, and GSTT1 genotype was low, causing the authors to conclude that sensitivity to DNA damage is likely multigenic in this cohort.

The Mertans et al. study mirrors the results of many similar studies with few

significant findings correlating genotype and increased risk of SMN in a variety of

primary cancer backgrounds.47, 53-59 This would lead us to believe that risk of malignancy

is dependent on the additive effects of multiple genes in a variety of pathways including

DNA repair, detoxification, drug metabolism, and potentially antioxidant related genes.

13

Figure 1.1: Annual Incidence Rates of Hodgkin Lymphoma Age is a risk factor in the development of Hodgkin Lymphoma. Age of incidence occurs in a bimodal distribution with adolescents and young adults (15-34y) and individuals over the age of 60 years at the greatest risk. Incidence rates among men and women are nearly equal until age 35, at which point the disease prevalence decreases in women and remains constant in men. (Created using SEER incidence data1)

14 Figure 1.1

15

Table 1.1: Composition of Participants within the CCSS Cohort Demographic characteristics and cancer diagnosis and treatment information is listed for participants in the CCSS cohort. Participants included pediatric cancer survivors who met CCSS eligibility criteria and completed a baseline questionnaire for enrollment into the cohort. (Demographic data available from www.stjude.org/ccss)

16 Table 1.1 Composition of Participants within the CCSS cohort

Participant Breakdown Characteristic Number and (Percentage)

Total number of Participants 14054 (100)

Sex Male 7541 (54) Female 6513 (46)

Race Caucasian 11679 (87) Hispanic 635 (5) African American 291 (2) Asian/Pacific Islander 165 (1) Other 615 (5)

Diagnosis ALL 4233 (29) AML 357 (3) Other Leukmia 137 (2) Astrocytomas 1158 (8) Medulloblastoma/PNET 366 (3) Other CNS tumors 305 (2) Hodgkin Disease 1906 (14) Non-Hodgkin Disease 1054 (7) Kidney tumors 1214 (9) Neuroblastoma 928 (7) Soft tissue sarcoma 1222 (9) Ewing's sarcoma 393 (3) Osteosarcoma 727 (5) Other bone tumors 53 (<1)

Age at Diagnosis <1 975 (7) 1-3 3539 (25) 4-7 3157 (22) 8-10 1538 (11) 11-14 2373 (17) 15-20 2472 (18)

Treatment Modality Chemotherapy + Radiation + Surgery 5491 (44) Chemotherapy + Radiation 1443 (12) Chemotherapy + Surgery 2237 (18) Radiation + Surgery 1477 (12) Chemotherapy Only 797 (6) Surgery Only 901 (7) Radiation Only 32 (<1)

17

Table 1.2: Type of SMN in HL Survivors within the CCSS Cohort The distribution by type of second and subsequent malignant neoplasms in HL survivors within the CCSS cohort is shown in the table. Radiation induced cancers such as breast and thyroid carcinoma are the most common SMNs in survivors of HL. This table represents SMNs within the CCSS HL cohort as of 2001.

18 Table 1.2

Second and Subsequent Primary Malignant Neoplasms in HL Survivors within the CCSS cohort

Second Malignant Neoplasm Number of Cases

Breast Cancer 35 Thyroid Cancer 17 Sarcoma 9 Leukemia 9 Non-Hodgkin Lymphoma 7 Melanoma 6 Bone Cancer 5 CNS Tumor 5 Other Malignancies 18 Total 111

Subsequent Malignant Neoplasm Number of Cases

Breast Cancer 4 Thryroid Cancer 3 Non-Hodgkin Lymphoma 1 Melanoma 1 Other Malignancies 1 Total 10

19

CHAPTER 2

INTRODUCTION TO REACTIVE SPECIES, ANTIOXIDANT ENZYMES, AND THE RELATIONSHIP BETWEEN OXIDATIVE STRESS AND MALIGNANCY

2.1: Reactive Species and Antioxidants

2.1.1: Reactive Oxygen Species

Reactive oxygen species (ROS) are natural byproducts of cellular metabolism that

have the potential to damage DNA, oxidize proteins, and initiate peroxidation,

resulting in mutagenesis or cell death.60 Common forms of ROS include superoxide

•– • anion (O2 ), hydroxyl radical ( OH), and hydrogen peroxide (H2O2). Both exogenous and endogenous sources contribute to the generation of ROS. Cellular sources of ROS include mitochondria, peroxisomes, cytochrome P450 metabolism, and inflammatory cytokine activation.61 Because mitochondria serve as the site of oxidative phosphorylation, they are thought to generate the greatest amount of ROS within the cell.62 Superoxide radicals are regularly produced in mitochondria as a result of the electron transport chain and may subsequently be converted to H2O2 or other forms of

ROS.63 Exogenous sources, such as carcinogens, xenobiotics, and radiation, are also associated with the generation of ROS.61, 64-67

20 Chemotherapeutic drugs, such as those used in the treatment of HL, are known to

generate ROS during metabolism. The class of drugs (which includes

), are particularly associated with ROS generation because they are known

redox-cycling agents.68, 69 Anthracyclines are quinone based drugs, and during

activation, the quinone molecule serves an as electron acceptor and is reduced.

Biological reducing agents such as NADPH, NADH, glutathione, and flavoproteins serve

as electron donors and become oxidized during the reaction.69 The reduced form of anthracycline may become re-oxidized in the presence of molecular oxygen by the donation of an electron to O2, resulting in the generation of superoxide anion. This redox

cycle continues as long as reducing agents and molecular oxygen are available.

Continued redox cycling may be cytotoxic due to the increasing amounts of ROS, and

depletion of reducing agents, such as glutathione, one of the most important molecules

involved in the cellular antioxidant system.68

2.1.2: Antioxidant Enzymes

The levels and actions of ROS are controlled by the presence of antioxidant enzymes within the cell. These enzymes function to mitigate the effects of ROS by detoxifying reactive species so that they can safely be removed from the body.

Antioxidants include the family of superoxide dismutases (SODs), glutathione peroxidases (GPXs), and catalase (CAT) enzymes. For proper biological function to occur, it is critically important to maintain a steady state balance between ROS production and neutralization by antioxidant enzymes. A process known as oxidative stress occurs when ROS overwhelm the capabilities of antioxidant enzymes leading to oxidative damage.

21 Antioxidant enzymes drive chemical reactions within the cellular environment to

convert ROS into non-toxic molecules. Chemical reactions involving ROS and

antioxidant enzymes are shown in figure 2.1. While each enzyme has a specialized

function, the system is complex and highly integrated. SOD enzymes function by

•– catalyzing the dismutation of the superoxide anion (O2 ) to form H2O2 and O2. GPXs convert H2O2 to H2O, by the oxidation of glutathione to glutathione disulfide, and

61, 70, 71 catalase converts H2O2 to H2O and O2. Although substrate affinities, conversion

rates, and subcellular distributions differ, GPX and catalase have somewhat overlapping functions, and the loss of catalase may be partially compensated by GPX activity.72

Additionally, dietary intake of antioxidants, such as vitamins C and E, as well as carotenoids and flavonoids, may help counteract the damaging effects of ROS.73-75

2.1.3: Reactive Nitrogen Species

Reactive nitrogen species (RNS) are another type of reactive molecule produced during normal physiological processes. Examples of RNS include, nitric oxide (NO•), a signaling molecule generated by nitric oxide synthase (NOS), as well as peroxynitrite anion (ONOO-), a powerful oxidizing free radical.61 Though NO• is a short-lived molecule (with a half life of mere seconds), it serves as a critical messenger involved in a

wide variety of biological processes.76 It is an endogenously produced gas involved in regulating neurotransmission, immune function, blood , and smooth muscle relaxation.61, 77, 78 In combination with superoxide anion, NO• has the potential to form

ONOO-(figure 2.2). This reaction typically occurs during inflammation, as immune cells

•– • produce an oxidative burst (releasing O2 and NO ) when they come into contact with an antigen.79 The reactive potential of peroxynitrite anion is much greater than that of NO•.

22 As an oxidizing free radical, peroxynitrite anion is capable of damaging DNA and

inducing lipid peroxidation. A process known a nitrosative stress occurs when the steady

state balance between RNS production and neutralization is unstable leading to an

overload of RNS and the potential for cellular damage.80 As both reactive oxygen

species and reactive nitrogen species have the potential to cause oxidative damage, they

may be referred to collectively as reactive species (RS).

2.2: Types of Antioxidant Enzymes

2.2.1: Superoxide Dismutase

SOD is an essential antioxidant enzyme that defends cells against potentially

damaging superoxide radicals. There are three known human isoforms of SOD, two of

which are copper-zinc containing enzymes (CuZnSOD), and one manganese containing

enzyme (MnSOD). SOD1 (CuZnSOD) is found in the cytoplasm and nucleus in the form

of a dimer.81 SOD2 (MnSOD) is a tetrameric protein that functions in the mitochondria,

and SOD3 (CuZnSOD) is a tetrameric, extracellular form of the enzyme82, 83 While each enzyme performs a critical function, SOD2 is particularly important due to its location within the mitochondria. Over 95% of cellular oxygen is metabolized in the

mitochondria during oxidative phosphorylation, making these organelles particularly

susceptible to oxidative stress.84, 85

2.2.2: Glutathione Peroxidase

Members of the GPX family are selenoproteins, meaning they incorporate selenocysteine into their primary protein structure. Incorporation occurs at the active site of the protein, and selenocysteine is known to act as an efficient biological catalyst.86-88

23 The main function of GPX is to catalyze the reduction of hydroperoxides to water and the

respective alcohols, while oxidizing glutathione (GSH) to glutathione disulfide (GSSG).

GSH is an important antioxidant that functions as a scavenger for ROS, including

• - 89 OH, ONOO , H202 and lipid peroxyl radicals. It is synthesized in the liver from

glutamate, cysteine, and glycine to form a tripeptide. Within the cellular environment,

glutathione cycles between a reduced form (GSH) and an oxidized form (GSSG) with the ratio of GSH to GSSG greater than 500 to1.90 Glutathione sulfide reductase is responsible for converting GSSG to GSH, and uses NADPH as a cofactor in this reaction.91 GSH and GSSG serve as the major redox couple within the cell, and are

important determinants in the total cellular antioxidant capacity.89, 90

In humans, four distinct isoforms of GPX have been identified. Expression of

each isoform is ubiquitous, though levels vary by tissue type.71 GPX1 is found in the cytoplasm and mitochondria of most cells, and is highly expressed in the liver, kidney,

92 lung, and red blood cells. This isoform functions to catalyze the reduction of H2O2 and some organic peroxides and is similar in both function and homology to GPX2 and

GPX3. The cytosolic isoform, GPX2, is found mainly in the liver and gastrointestinal tract, and the unique distribution of this enzyme suggests that it may protect against ingested lipid hydroperoxides.92, 93 Produced mainly by the kidney, GPX3, is secreted

into the extracellular environment and is most highly detected in plasma.94 Finally,

GPX4 is found in the cytoplasm, nucleus, and mitochondria of many cells, and is most highly expressed in the testis.95

Studies involving GPX4 have demonstrated that it differs from the other GPX enzymes in several important ways. Unlike the other isoforms, GPX4 is capable of 24 directly reducing phospholipid hydroperoxide within the cell, including lipid peroxides

derived from cholesterol.96, 97 Additionally, it functions as a monomer, rather than as a tetramer as seen in the other isoforms.98 Finally, GPX4 is translated as either a long or short form of the protein, with the long form containing a mitochondrial targeting sequence at the N terminus of the protein.99, 100 Once imported into the mitochondrion, the long form of the protein is modified to remove the targeting sequence, resulting in a protein of equal length to the short form. Expression of the short form was found to occur in the nucleus, endoplasmic reticulum and cytosol, but was not observed in the

101 mitochondrion. While all GPX isoforms and catalase share a common substrate, H2O2,

GPX is considered to be the major enzyme responsible for the reduction of lipid and organic peroxides.71

2.2.3: Catalase

Catalase is an antioxidant enzyme that is concentrated mainly in peroxisomes (an organelle involved in the breakdown of fatty acids). This enzyme functions as a tetramer and contains a ferriprotoporphyrin group in each of the four subunits71 As the name implies, this enzyme catalyzes the conversion of H2O2 to water and oxygen. While catalase is clearly a critical enzyme, GPX has the ability to compensate partially for low or absent catalase function.72

2.3: Oxidative Stress and Cancer

2.3.1: A Multi-Stage Model of Carcinogenesis

There is mounting evidence that persistent oxidative stress is a major contributor to the development of cancer.61, 65, 70-72 In a multi-stage model of carcinogenesis, the development of cancer is based on the cumulative effects of successive events within a

25 single cell.102, 103 The model may be described by three stages including the initiation,

promotion and progression of tumorigenesis. The effects of oxidative stress within a cell

can impact each stage of the model as shown in figure 2.3.

2.3.2: Initiation

Genetic change is the initiating event in the multi-stage model of carcinogenesis.

During periods of oxidative stress, damage to DNA has the potential to result in genetic

change. Oxidizing free radicals may cause irreparable damage including base

modification, DNA strand breaks, and DNA-protein cross-links.60, 104 The presence of modified base products, such as 7,8-dihydro-8-oxy-2’-deoxyguanosine (8-OH-dG) serve as good indicators of oxidative base damage. Studies involving cancers of the breast, lung, and kidney all report significant increases of 8-OH-dG in tumor vs. normal tissues from the same patient.73, 74, 105

2.3.3: Promotion

Following cellular damage, an initiated cell may gain a pre-neoplastic phenotype due to the dysregulation of cell signaling pathways and tumor suppressors, and an inhibition of apoptosis. The clonal expansion of initiated cells represents the promotion stage of carcinogenesis. Promotion is considered to be a reversible event, as a constant exposure to tumor promoting factors is required to induce a malignant phenotype.61, 106

2.3.4: RS: A Balancing Act of Tumor Promotion or Tumor Suppression

The effects of oxidative damage within a cell represent a paradox between tumor

suppression and tumor promotion due to the nature of RS (Figure 2.4). The collective

cellular environment (including cell type, levels of antioxidants, types of RS, relative

amounts of RS, and total exposure) ultimately sways the balance in one direction.70 On

26 one end of the spectrum, tumor suppression may occur because of the severe oxidative

damage that results from an overload of RS. Suppression of a malignant phenotype

occurs due to senescence, necrosis, or induction of apoptosis in affected cells.

Alternately, a low to moderate increase in RS may have a tumor promoting effect.

Escalating cellular damage is caused by oxidative stress, but not to the point of inducing

apoptosis. In turn, oxidative damage may result in the dysregulation of cellular processes

and contribute to proliferation and inhibition of apoptosis.107

2.3.5: Progression

The final stage in the model of carcinogenesis is progression. Pre-neoplastic cells

from the promotion stage undergo further genetic damage and are no longer able to

respond to growth inhibition or apoptotic signals.61 Evidence suggests that the involvement of RS in tumorigenesis may also include roles in angiogenesis and metastasis. RS may activate matrixmetalloproteinases, which degrade the extracellular matrix and contribute to the invasive potential of the tumor.108, 109 RS are also known to regulate vascular endothelial growth factor signaling in relation to angiogenesis.110, 111

2.3.6: Antioxidant Enzymes and Malignancy

In relation to oxidative stress and cancer, a variety of studies involving antioxidant enzyme levels and malignancy have been performed. In studies of SOD activity, tumor cells nearly always show a decrease in SOD2 expression, and quite often a decrease in SOD1 as well.112-114 In knock-out experiments, mice lacking SOD1

demonstrated an increased rate in the development of liver cancer, while SOD2 null mice exhibit a neonatal lethal phenotype.115, 116 Interestingly, mice with a single copy of SOD2 survive, but are at increased risk for the development of cancer later in life.117 Catalase

27 activity is generally lower in tumor cells than in normal tissue, while glutathione

peroxidase activities have been found to be variable.113, 118 In mice lacking the catalase enzyme, an increased rate of spontaneous mammary tumor formation has been observed.119 Additionally, in a simultaneous knock-out experiment of both GPX1 and

GPX2, mice were found to develop intestinal cancer.120 While a causal relationship

between oxidants and cancer has yet to be proven, it is important to note the many

correlations that have been observed to date. Further research in this field will improve

our understanding of the role of oxidative stress in the development of cancer.

28

Figure 2.1: Chemical Reactions Involving ROS and Antioxidant Enzymes In reaction [1] superoxide anion may be converted to a secondary ROS after catalysis by SOD during the dismustase reaction. H202 is a substrate for both the GPX and CAT enzymes as shown in reactions [2] and [3]. A redox reaction occurs in [3] as reduced glutathione (GSH) is converted to its oxidized form (GSSG) in turn reducing H202 to H2O.

29 Figure 2.1

30

Figure 2.2: Reactive Species Interact to Form an Oxidizing Free Radical • •– Nitric oxide (NO ) in combination with superoxide anion (O2 ) forms the oxidatively active peroxynitrite anion (ONOO-). This reaction is known to occur during the oxidative burst triggered by an inflammatory reaction within the immune system.

31 Figure 2.2

· ·- - NO + O2 ONOO

32

Figure 2.3: A Multi-Stage Model of Tumorigenesis Relative to Oxidative Stress Oxidative damage from RS may serve as the initiating event in carcinogenesis. During the promotion stage, RS may cause either tumor promoting or tumor suppressing effects dependent on the level of oxidative stress. Following progression, RS may contribute to the invasiveness and metastic potential of tumor cells.

33 Figure 2.3

34

Figure 2.4: Cellular Response to ROS During Times of Oxidative Stress ROS are constantly generated during normal physiological processes in response to endogenous and exogenous stimuli. Inherently, a steady state balance is maintained within a cell as antioxidant enzymes counteract and neutralize ROS. During times of oxidative stress, the balance has shifted due to an increase in ROS production, or a decrease in antioxidant enzyme buffering capacity. As levels of ROS increase, so does the potential for oxidative damage to occur. While small amounts of damage are quickly mitigated by cellular repair processes, increasing damage over an extended time period is likely to be detrimental. Low to moderate levels of ROS may be tumor promoting as typical cellular processes are disrupted due to the cumulative effects of oxidative damage. The inappropriate activation of genes, phosphorylation of proteins, and increase in ROS availability for cell signaling may result in a proliferative phenotype. Alternately, when of ROS are too high, the results are catastrophic due to overwhelming oxidative damage and ultimately cell death.

35 Figure 2.4

36

CHAPTER 3

THE ASSOCIATION OF ANTIOXIDANT RELATED GENE POLYMORPHISMS AND SECOND PRIMARY MALIGNANT NEOPLASMS IN A PEDIATRIC HOGDKIN LYMPHOMA POPULATION

This work was performed in collaboration with Dr. Kim McBride (The Ohio State University/Nationwide Children’s Hospital), Dr. Stella Davies (Cincinnati Children’s Hospital Medical Center), Dr. John Whitton and Dr. Wendy Leisenring (Fred Hutchinson Cancer Research Center).

3.1: Abstract

Improved treatment strategies in pediatric Hodgkin Lymphoma (HL) have

resulted in a cure rate approaching 95%, yet the development of a second primary

malignant neoplasm (SMN) is a risk for long-term survivors. In a pediatric HL

population, between 6-26% of patients will develop a SMN within 30 years following

treatment. The etiology of HL is still largely unknown, as is the cause of certain types of

SMNs.

Oxidative stress has been linked to the development of cancer due to the damaging effects of reactive oxygen species (ROS) on DNA, lipids, and proteins. During periods of extended oxidative stress, the damaging effects of ROS are likely to increase which in turn raises the risk of genetic change. In a multi-stage model of tumorigenesis, genetic change is considered to be the initiating event of cancer development. Under

37 normal circumstances, antioxidant enzymes within the cell mitigate the effects of ROS by

converting reactive species into non-toxic molecules. Mechanisms within the cell

maintain a steady state balance between ROS and antioxidant enzyme levels. We

hypothesize that individuals predisposed to lower levels of antioxidant enzyme activity

due to polymorphic variants within those genes may be at risk for increased damage

caused by ROS. Although decreased amounts of antioxidant enzymes have been found in

a variety of cancers, a correlation between polymorphisms in antioxidant genes and

predisposition to secondary cancer has not been studied.

The purpose this study was to assess the association of antioxidant gene alleles with the risk of developing a SMN in HL survivors. DNA samples were obtained from

768 HL patients enrolled in the Childhood Cancer Survivor Study (CCSS). The samples were genotyped for 90 polymorphisms in antioxidant related genes including SOD, GPX,

NOS, CAT, and CYP2C9. Statistical analysis methods to determine risk of developing a

SMN included association, haplotype, and multiple regression models.

In our HL cohort, 131 patients developed a SMN. An additional 117 patients

developed nonmelanoma skin cancer, and 34 patients developed 2 SMNs. Out of the 36

SNPs that were included in the final analysis, 4 SNPs in the GPX1, GPX3, GPX4, and

SOD2 genes, were potentially suggestive (p<0.05) of an association between genotype

and the development of a SMN. A general trend observed in the genotyping data

suggested that an increased risk of SMN was conferred by the presence of the minor

allele, while a decreased risk of SMN was conferred by the presence of the major allele in

the population. Replication studies are necessary, though it is notable that polymorphisms within the GPX family may be associated with the development SMN in

38 our cohort. Additionally, hazard ratios were only moderately increased in this population suggesting that the development of a SMN in a pediatric HL is likely multi-factorial in

nature. The elucidation of critical genes involved in SMN development could influence

patient follow-up and intervention strategies, and potentially aid in the prevention of

SMN.

3.2: Introduction:

Though childhood cancer is a rare disease, more than 10,000 new cases will be

diagnosed in a pediatric population (0-14 years of age) within the United States this

year.121 Advances in chemotherapy and radiation treatment options have dramatically increased once poor cure rates, with 5-year survival rates now approaching 80% for all childhood cancer diagnoses. As the number of pediatric cancer survivors increases, investigators have begun to focus their efforts on reducing the long term treatment related side effects in patients. Late effects are wide ranging and may include cognitive impairment, developmental delay, vision and hearing problems, infertility, thyroid dysfunction, cardiovascular and respiratory complications, development of a second primary malignant neoplasm (SMN), psychological and social problems, educational setbacks, obstacles in obtaining employment and insurance, and an overall reduced quality of life.122

Pediatric cancer survivors may be particularly susceptible to the late effects of treatment for several reasons. Diagnosis at a young age requires that chemotherapy and/or radiation treatment occur during prime years of growth and development, with the potential for an increased risk to the developing child. High doses of radiation are known to be especially damaging to pediatric patients, resulting in impaired growth of soft tissue

39 and bone, in addition to thyroid dysfunction.37, 38 Furthermore, pediatric cancer survivors

still have the potential for a long life span, and with that comes the possible burden of late

effects at each stage of life.

Survivors of Hodgkin Lymphoma (HL) seem to be particularly susceptible to the

late effects of cancer therapy. The most common long term sequelae in HL patients

include thyroid dysfunction, sterility, cardiopulmonary disease, and SMN. Tailored

treatment regimens in HL patients have resulted in a 95% cure rate, yet morbidity and

mortality due to SMNs are the highest out of all pediatric cancers studied in the CCSS

cohort.23, 24 The cumulative incidence of developing a SMN within 20 years of original diagnosis was 7.6% in HL survivors.122 The optimization of treatment in order to

minimize late effects without reducing the efficacy of therapy has become a major goal of

pediatric cancer research.

While the development of some types of SMNs is attributed to cancer therapy, the

cause of other SMNs is unknown. It is probable that genetic background, as well as

environmental influences, play a role in developing a SMN. Previous studies have

examined polymorphic variants in genes involved in DNA repair, cytochrome P450

metabolism, and detoxification pathways in relation to SMNs in a variety of primary

cancer backgrounds.36, 44, 45 Only low to moderate increased risk was observed in these

studies; therefore, the development of a SMN was hypothesized to be multigenic in

nature.

Oxidative stress has been linked to the development of cancer due to the

damaging effects of reactive oxygen species (ROS) on DNA, lipids, and proteins. During

periods of extended oxidative stress, the damaging effects of ROS are likely to increase

40 which in turn raises the risk of genetic change. In a multi-stage model of tumorigenesis,

genetic change is considered to be the initiating event of cancer development. Under

normal circumstances, antioxidant enzymes within the cell mitigate the effects of ROS by

converting reactive species into non-toxic molecules. Mechanisms within the cell

maintain a steady state balance between ROS production and neutralization by

antioxidant enzymes. We hypothesize that individuals predisposed to lower levels of

antioxidant enzyme activity due to polymorphic variants within those genes may be at

risk for increased damage caused by ROS. Although decreased amounts of antioxidant

enzymes have been found in a variety of cancers, a correlation between polymorphisms

in antioxidant genes and predisposition to SMN in HL survivors has not been studied.

3.3: Materials and Methods

3.3.1: The Childhood Cancer Survivor Study

The Childhood Cancer Survivor Study (CCSS) was designed to examine the late

effects of cancer treatment among survivors of pediatric cancer. As a retrospective study,

it followed a cohort of 14,372 pediatric cancer survivors diagnosed between 1970-1986.

CCSS eligibility requirements included; (a) diagnosis of leukemia, CNS tumor,

Hodgkin’s lymphoma, non-Hodgkin’s lymphoma, kidney tumor, neuroblastoma, soft

tissue sarcoma, or bone tumor; (b) diagnosis and treatment at one of 27 participating

oncology centers in the US and Canada; (c) primary cancer diagnosis between January 1,

1970, and December 31, 1986; (d) age 0-21 at diagnosis; and (e) survival a minimum of 5

years past diagnosis. The cohort and study design has been previously described.42

Contact and recruitment of participants began in 1994. Eligible and consenting patients

(or parents) completed a 24 page questionnaire and agreed to have their medical records

41 abstracted. Detailed information regarding primary cancer diagnosis and treatment,

family history, quality of life, and patient health were collected to assess various

outcomes. Current areas of study involving the cohort include; chronic disease in survivors, genetics, cancer control/intervention, reproductive outcomes, second malignancies, epidemiology/statistics, and neurologic/psychosocial outcomes.

Because the CCSS cohort is so well characterized, it has become a useful tool for

genetic study. Attributes such as the large sample size, similar treatment regimen among

cancer survivors, and the inclusion of age matched healthy sibling controls help to

minimize bias in the cohort.

3.3.2: Buccal Cell Collection

The CCSS began collection of buccal cell samples from eligible individuals

within the cohort in 1999. In order to procure samples, CCSS participants were mailed a

specimen collection kit which included a cover letter explaining the study, a consent

form, an instruction sheet, a 45ml bottle of mouthwash (15% alcohol content), a

specimen collection container, mailing labels, and return postage.123, 124 Participants were advised to rinse with the mouthwash, and to return the used mouthwash to the specimen collection container. They were then instructed to return the container by mail to the designated CCSS laboratory. As of June 2005, 5790 members of the cohort had returned a buccal cell sample. DNA was isolated from the mouthwash by standard procedures using the Puregene kit (Qiagen, Valencia, CA). After quantification, a median of 40ug of

DNA per sample was obtained in the cohort.123 The molecular genetics laboratory at

Nationwide Children’s Hospital received 1ug of DNA (100ng/ul) from 768 HL patients enrolled in the CCSS.

42 3.3.3: Genotyping Methodology

The SNPlex High-Throughput genotyping system (Applied Biosytems, Foster

City, CA) was used to determine patient genotype at polymorphic sites near or within antioxidant related genes in our HL survivor cohort. Polymorphisms were chosen after

thorough review of the literature from relevant antioxidant related genes including;

SOD2, SOD3, GPX1, GPX2, GPX3, GPX4, CAT, and NOS3. Prior to genotyping, 114

potential SNPs were screened online using the SNPlex Assay Design Tool

(www.appliedbiosystems.com). The design tool was utilized to create multiplex SNP pools with the greatest combination of polymorphisms passing both genomic screening and assay rules. Following analysis, 25 SNPs failed screening and the resultant 89 SNPs were divided into multiplex probe pools containing 44 and 45 polymorphisms each. Each probe pool was used independently in SNPlex genotyping reactions.

The combination of an oligonucleotide ligation assay (OLA) and multiplex PCR form the basis of a SNPlex reaction (Figure 3.1). Allelic discrimination is achieved through the use of allele specific oligo (ASO) probes designed with 3’ end specificity relative to each SNP. During the OLA assay, a locus specific oligo (LSO) probe binds near site of the SNP, and in combination with the ASO probes, creates an OLA reaction product following a phosphorylation and ligation reaction. Universal priming sites on the

ASO and LSO probes are utilized during the multiplex PCR reaction to allow for target amplification. Polymorphic variants are distinguished using ZipChute probes which contain an allele specific zipcode sequence, a mobility modifier, and a fluorescent label.

Following electrophoresis on the Applied Biosystems (AB) 3730 genetic analyzer, base

43 calls were distinguished using the size and color variation provided by the ZipChute probes (Figures 3.2 and 3.3).

Due to the lower quality of buccal cell DNA, a moderate amount of DNA (100ng) was necessary for each multiplex reaction. DNA was dried overnight in 96-well plates prior to use. The OLA reaction, exonuclease purification, PCR amplification, and hybridization were performed according to published recommendations. Samples were electrophoresed on a 3730 Genetic Analyzer and analysis was performed using

GeneMapper 4.0 (Applied Biosystems, Foster City, CA). During analysis, samples were subjectively graded on quality, and scored as good, adequate, or poor. Scoring was based on a number of factors including; peak height relative to background, the number of successful SNP reactions per sample, and the appropriate findings in the positive and negative controls. All samples graded as poor quality were repeated.

DNA samples were run in duplicate to ensure accuracy in genotyping calls.

Because of the limited quantity and lower quality of the buccal cell DNA, samples were subjected to whole genome amplification (WGA) before running a second reaction.

WGA reactions were performed using the GenomePlex WGA kit (Sigma, St. Louis, MO) and results were validated to ensure bias (allelic dropout) was not occurring. In each

WGA reaction, 75ng of DNA served as the starting material. WGA reactions were run according to the published protocol, and reactions were purified using the Qiaquick 96- well PCR purification kit by silica gel membrane (Qiagen, Valencia, CA). At random, 10 samples were chosen from each plate in order to estimate DNA quantity. Approximately

150ng of WGA product was used per SNPlex reaction. Genotyping results were scored as before, and all poor quality samples were repeated. Additionally, approximately 15%

44 of all samples (unamplified and WGA) were genotyped a third time to ensure accuracy in

base calls.

Taqman allelic discrimination was used to genotype an SOD2 SNP (rs4880) that

was excluded from the original SNPlex multiplex pools due to genomic screening rules

(likely because of high GC content) (Applied Biosystems, Foster City, CA).

Approximately 30ng of unamplified DNA was used per reaction, and sequence verified

controls were run in each plate. A validated Taqman assay (C___8709053_10)

containing the gene specific primers and labeled probes was used for genotyping

(Applied Biosystems, Foster City, CA). Taqman reactions were run on the 7900HT

Sequence Detection System and analyzed by Sequence Detection Software 2.0 (Applied

Biosystems, Foster City, CA).

3.3.4: Statistical Analysis Methods

Statistical analysis was completed using tests of association, haplotype,

multivariate Poisson regression and Cox regression modeling (all analyses were

performed in collaboration with Dr. Kim McBride, Dr. Wendy Leisenring, and Dr. John

Whitton). For the purpose of these analyses, the development of a SMN was treated

separately from the development of a non-melanoma skin cancer (NMSC).

An association analysis was performed using Haploview software with HL

survivors sorted as either a case (with SMN or NMSC) or a control (no SMN or

NMSC).125 During the association test, the relationship between genotype and a given trait (SMN or NMSC) was studied. In order to correct against the bias of performing multiple tests on the dataset, a permutation test was also run using Haploview. In the

45 permutation test, case vs. control assignments were rearranged multiple times to

determine if an observed association with a SNP held up as a measure of signficance.126

Cox regression models were used to estimate the hazard ratio of developing a

SMN or NMSC in HL survivors by comparison of genotype.127 The hazard ratio is a measure of risk of a developing a SMN across genotypes as compared to a baseline (or reference) genotype with a risk of 1. The initial models looked at overall risk of SMN,

NMSC and SMN or NMSC, vs. no SMN/NMSC. Following the initial Cox regression analysis, additional models were created to estimate the hazard ratio of developing a certain type of SMN or NMSC vs no SMN/NMSC by comparison of genotype. The models were set up to include the following SMNs: breast, thyroid, breast or thyroid

(known radiation induced cancers), CNS and melanoma. NMSC was divided into squamous cell carcinoma and basal cell carcinoma for the purpose of this analysis.

The effects of treatment-related and patient-specific factors on the risk of

developing SMN or NMSC were also investigated. The use of Cox regression is not

appropriate in this case, as it is often difficult to define a time point at which a treatment

may have an effect on SMN/NMSC risk. For this reason, multivariate Poisson regression

analyses were used. The genotype hazard ratios were adjusted for the effects of

chemotherapy (Y/N), Radiation (Y/N), Sex (M/F) and age at HL diagnosis (0-9 y, 10-

16y, 17-21y).

The final analysis was performed by haplotype due to the low number of SNPs

per gene within our dataset. Theoretically, the association between a specific antioxidant

gene and risk of developing a SMN or NMSC could be detected by a SNP found to be in

linkage disequilibrium with a polymorphism included in our dataset. Because relatively

46 few SNPs per gene were included in the analysis, the results from haplotype blocks fell into groupings by gene. Haplotype analysis was performed using a generalized estimating equation (GEE). The GEE is a technique for modifying an analysis when results are not independent, and was used to correct the different probability estimates observed in individual haplotypes.128

In all analyses, genotyping data were assigned an arbitrary cutoff value of p<0.05 which represents only a suggestive association between SMN and genotype. It is important to note that many regressions were carried out, and we would expect 5% of them to meet this criterion. Uncorrected data is reported with the caveat that many analyses were performed on the dataset, and with the idea that p-values which are merely suggestive of a significant association still have value for downstream hypothesis generated research.

3.4: Results

Once genotype analysis was complete, CCSS statisticians at the Fred Hutchinson

Cancer Center (FHCC) provided a limited information data set on HL patients successfully genotyped during this research project. In our study group, 131 patients developed a SMN, and 117 were diagnosed with NMSC. A total of 34 patients were diagnosed with both a SMN and NMSC, with breast and thyroid (radiation therapy- related cancers) found to be the most frequent SMNs (Table 3.1). Typically, NMSC is treated as a separate group in comparison to other types of SMNs. This is due to the high frequency of NMSC in the general population, and subsequently because of the lack of accurate population incidence data regarding this malignancy.

47 Out of 768 HL patient samples, 754 samples (98%) were successfully genotyped.

After thorough review of SNPlex genotyping results, SNP dropout occurred at each new phase of design or analysis with a mean attrition rate of 23% (Figure 3.4). From an initial

SNP pool design containing 114 polymorphisms, 89 passed the strict AB screening guidelines. Upon validation, approximately 60 SNPs provided consistent and unambiguous results using both unamplified and WGA DNA. Lastly, before statistical analysis was performed, the SNP pool was further refined to eliminate polymorphisms which fell out of Hardy-Weinberg equilibrium, those more than 99% homozygous, and those missing a large amount of genotyping data. Following this approach, 35 SNPlex polymorphisms remained eligible for analysis (Table 3.2). A single Taqman SNP was also included in the final data analysis.

The association test compared genotypes of HL individuals who developed a

SMN (case) to HL patients who did not develop a SMN (control). A permutation test was used to correct for multiple testing bias. No significant relationship between SMN and genotype was found using this method.

Association results generated by Cox regression analysis with a cutoff of p<0.05 are listed in Table 3.3, with the results summarized in Table 3.4. Genotypes were compared to a reference group with a hazard ratio (HR) of 1 in order to determine if allele combinations were associated with an altered risk of SMN. Multiple reference groups were used to determine the impact of an allele in both a heterozygous or homozygous state.

Out of the 36 SNPs that were included in the final analysis, 4 SNPs were potentially suggestive of an association between genotype and the development of a

48 SMN. The SNPs were found to occur near or within the following genes, GPX1, GPX3,

GPX4, and SOD2. Increased hazard ratios appeared to be associated with the presence of the minor allele, while decreased hazard ratios were associated with the presence of the major allele.

Results from the Cox regression models that considered the type of SMN as an outcome during analysis are shown in table 3.5 for SNPs with a p-value <0.05 with a summary listed in table 3.6. Classification by type of SMN included breast and thyroid cancers as independent variables and in a combination (breast or thyroid) in order to better measure risk for radiation induced SMNs. Additional SMN and NMSC outcomes including CNS cancer, melanoma, basal cell carcinoma, and squamous cell carcinoma were considered in the analysis.

Radiation induced cancers (breast and thyroid) represented the most frequently observed SMNs with findings nearing significance. This is likely due to the small sample size of other SMNs observed in our cohort. A potential association between genotype and the development of breast or thyroid SMN was observed in 4 SNPs, including GPX1,

NOS3, CAT, and GPX3. For NMSC, a total of 2 SNPs (in GPX1 and GPX4) were suggestive of an association between genotype and the development of basal cell

carcinoma. Once again, the presence of a minor allele seemed to confer an increased risk

of SMN, while the presence of a major allele demonstrated a protective effect.

Multivariate Poisson regression analyses were used to adjust for the effects of treatment-related and additional factors on the risk of developing SMN or NMSC in our cohort. Genotype hazard ratios were adjusted for the effects of chemotherapy and radiation treatment, sex, and age at HL diagnosis. By this analysis, 2 SNPs (one each in

49 GPX4 and CAT) were suggestive (p<0.05) of an association between genotype and SMN.

These findings are presented in table 3.7. It is important to note that the reference

genotype used in the Poisson analysis was opposite that used in the Cox regression

models. Risk may be associated with an increase or decrease dependent on the reference genotype.

Poisson regressions were also used during haplotype analysis to adjust for the

effects of treatment, sex, and age at HL diagnosis. Analysis by haplotype grouped SNPs

according to the combination of alleles at linked loci. Polymorphisms were grouped by

haplotype as single genes, as the analyzable SNPs per gene were found to be in linkage

disequilibrium. The results were suggestive of an association between the GPX1 ATAC

haplotype and a slightly increased risk of SMN or NMSC. This analysis may have been complicated by the small number of analyzable SNPs per gene, as the results for other genes were found to be inconclusive (Table 3.8).

As a general trend observed in both Cox regression and multivariate Poisson analyses, in polymorphisms found to be suggestive of significance, the presence of a minor allele conferred an increased risk for SMN, while the presence of the major allele demonstrated a protective effect. While no single SNP appeared to be more highly suggestive of significance compared to others with p-values <0.05, it is notable that several polymorphisms from the GPX family approached significance during our analysis. Because multiple analyses were performed on the dataset, any significant findings may be due to chance. Additionally, caution must be used when interpreting data generated from small numbers of individuals due to the potential for bias.

50 3.5: Discussion

The development of a SMN in survivors of pediatric cancer is likely due to a

combination of genetic predisposition and exposure to genotoxic agents used during cancer therapy. The CCSS cohort serves as a valuable resource in which to study the etiology of SMNs without bias because it consists of a large number of members, with well characterized treatment regimens and similar backgrounds. HL survivors in particular, are useful to study because of the high incidence of SMNs, similar treatment regimen of patients, and the considerable number of participants in the cohort.

Out of 754 HL survivors within our cohort, 131 (17%) developed a SMN, and 117

(16%) developed NMSC. Thirty-four patients were diagnosed with both a SMN and

NMSC. Radiation induced cancers represented the most frequently observed SMNs, as

9% of HL survivors developed breast cancer, and 3% of survivors were diagnosed with thyroid cancer. In order to improve the quality of life for HL survivors, there is a pressing need to identify biomarkers associated with SMNs so that adequate patient follow-up can occur.

Polymorphisms in antioxidant related genes were the focus of study because of

the documented relationship between oxidative stress and malignancy. In isolation, it is

improbable that an antioxidant related polymorphism (such as one leading to a decrease

in enzyme activity) would confer much risk for the development of a SMN. This is

because antioxidant enzymes have somewhat overlapping functions and may be able to

compensate for a decrease. Additionally, unless the system is challenged by an overload

of RS, decreased activity may not have a physiological effect in a normal and healthy

individual.

51 The difficulty arises within the context of cancer, which necessitates that patients undergo chemotherapy and/or radiation treatments known to dramatically increase RS levels and oxidative damage to cells. Common HL including doxorubicin, , and radiation have been shown to increase cellular RS levels.64-67 It is notable that anthracyclines such as doxorubicin, can act as redox-cycling agents that generate

ROS and in turn deplete biological reducing agents in a continuing cycle.68 A cancer patient with a predisposition to lower levels of antioxidant enzyme activity, in combination with the oxidative stress brought on by therapy, may be at increased risk for treatment effects, especially the development of a SMN.

While the purpose of cancer therapy agents is to eradicate malignant cells, exposure of normal cells is unavoidable and may have devastating long terms effects in cancer survivors. In order to avoid treatment related sequelae, investigators are now focusing their effects on optimizing treatment, while reducing the risk to survivors.

There are several limitations to this study. Of primary concern is the small number of SNPs available in the final analysis of the antioxidant related candidate genes.

Incomplete SNP coverage within these genes may have led to an underestimate of

association between SMN and genotype. Acertainment bias is a concern within our

cohort on the basis of inclusion criteria, as only HL patients within the CCSS who

complied with the request to provide a buccal cell sample were included in our analysis.

Therefore, patients lost to follow-up, those deceased, or those who opted not to submit a

buccal cell sample were not included in our cohort. Of particular concern are HL patients

who are deceased due to a SMN, as the rate of SMN is likely underestimated in this

population. Additionally, it should be noted that in order to control for the possibility of

52 therapy related genetic change, we compared HL patients without a SMN (serving as controls) to those with a SMN (cases). Therefore, a polymorphism associated with the development of HL would not be detected in our analysis.

The observation of false-positive results is a concern in our study because of the large number of statistical comparisons performed. It is possible that some or all associations with p-values <0.05 in our study may be due to chance alone. The generation of a false-positive result due to chance is known as a type I error.129 There are methods to correct against the bias of performing multiple tests in order to prevent type I errors. A permutation test, such as the one performed during our case vs. control association analysis would be one such method. Additionally, it is possible to control for the false discovery rate (FDR) observed in multiple comparisons. The FDR is the expected proportion of false positive results among all results generated within a study.130, 131 When using this method, a q-value provides a measure of the significance

for each finding in relation to a FDR, much like a p-value provides a measure of

significance in relation to a false positive rate.131 The FDR method is useful because it

can control against increased false positives due to multiple testing bias without being so

stringent that findings of true significance are lost. A false-negative error, or type II

error, is the error of failing to observe a difference when one truly exists.129 Type II

errors are a major reason why the Bonferroni adjustment is not appropriate to correct for

multiple comparisons, as findings of true significance may be lost due to the extremely

stringent p-value requirements.132

The results from this study are potentially suggestive of an association between

antioxidant related polymorphisms and risk of SMN or NMSC, though replication studies

53 are necessary to determine true associations. Yet, the results still have value in that they

could serve as starting point for hypothesis driven research, particularly in SNPs yielding

low p-values (P<0.02) (personal communication from Dr. John Whitton). It is notable

that polymorphisms within the glutathione peroxidase (GPX) family may be associated

with the development SMN or NMSC in our cohort, and this finding could impact future

studies.

Members of the glutathione peroxidase (GPX) gene family are important

antioxidant enzymes which help to mitigate the damaging effects of reactive species.

GPXs function as a catalyst in an oxidation-reduction reaction to convert hydroperoxides

to H2O and alcohol [(H2O2 + 2GSH → 2H2O +GSSG) and (ROOH + 2GSH → ROH +

GSSG +H2O)]. During this reaction, glutathione (GSH) serves as a co-factor and is oxidized to form glutathione disulfide (GSSG). Four isoforms of GPX have been identified in humans. Expression of each isoform is ubiquitous, though levels may vary by tissue type.71 Though GPX enzymes and catalase have somewhat overlapping functions, GPX is considered to be the major enzyme responsible for the reduction of lipid and organic peroxides.71

The relationship between GPX enzyme function and abnormal phenotype has been previously studied. In one such study, mice lacking the GPX1 enzyme were found to have retarded growth and an increased level of lipid peroxidation in the liver.133 An additional study revealed that the simultaneous knockout of both GPX1 and GPX2 led to an increased risk of colitis and intestinal cancer in mice.120 Mice in this study exhibited sensitivity to oxidative stress, and were highly susceptible to bacterial-associated inflammation and tumor development in the gastrointestinal tract. GPX1 and GPX2

54 double knockout mice were not reported to have retarded growth, which may be due to

the differences in genetic background of the mouse strains. In studies of GPX4 null mice,

an embryonic lethal phenotype was observed. Abnormal embryos were found to lack

well organized structures, and failed to develop past embryonic day 8. This suggests that

GPX4 is an essential gene in mouse development. Cell lines derived from heterozygous

mice (GPX4+/-) with a single functional copy of the gene demonstrated an increased

sensitivity to oxidative stress.134 In entirety, these data suggest that the GPXs are critical enzymes necessary for normal physiological function and antioxidant defense within the cell.

In order to more fully elucidate the relationship between antioxidant genes and

SMN, future studies are necessary. Because many SNPs were not fully analyzable in our study, it would be prudent to take a step back and perform a genome-wide association analysis. By this method, we are not limited to studying only genes within the antioxidant network, but we can analyze genes involved in detoxification and drug

metabolism, as well as determine if associations between previously unexamined genes

and SMN exist. An association analysis using the Affymetrix Genome-Wide Human

SNP Array 6.0, provides the most comprehensive coverage of the human genome to date.

It contains 906,600 polymorphims and therefore reduces the risk of false-negative results

due to an underpowered study. It is an increasing affordable option when performing

moderate sized association studies. In order to continue the study of SMN in HL

survivors, I would perform an association analysis using the Affymetrix SNP Array 6.0 to

genotype the CCSS HL cohort. Ideally DNA should be collected from all members of

the cohort in order to increase the number of genotypable participants in the study.

55 The next step is to perform biological verification of the findings which were

suggestive of significance in the association analysis. In relation to our HL study, let us

assume that an association between GPX4 and SMN was demonstrated using the

Affymetrix SNP array. At this point it would be important to determine if a SNP is functionally significant in relation to oxidative stress response. A cell culture system utilizing a transfected GPX4 expression vector containing the SNP of interest would serve as a good resource in which to measure functional activity. Because the degree of homology between the human and murine GPX4 gene is very high (94%), studies of

functionality could be performed using mouse cell lines.135 A heterozygous GPX4 cell

line, such as the GPX4+/- embryonic fibroblast line generated by Yant et al. during the study of GPX4 knock out mice, would serve as a good starting point for studies of biological function.134 A design such as this might yield a more meaningful measure of

GPX4 activity and oxidative stress response, as levels would approximate that of the endogenous gene. Overexpression of GPX4 in cell culture systems has been shown to result in the inhibition of apoptosis, and therefore would not be representative typical biological activity.136-138

To begin, it is necessary to obtain a full length GPX4 expression clone available from a source such as Origene (Rockville, MD) or GeneCopoeia (Germantown, MD).

Next, the clone would be sequenced to confirm that it contained GPX4 sequence and to

determine if any polymorphisms were present in the gene. In order to measure the effect

of a specific polymorphism, site-directed mutagenesis would likely be performed in order

to introduce the SNP of interest into the clone. At this point both a wildtype and variant

form of the clone would be transfected into separate GPX4 heterozygous cell lines, and

56 measurements of gene expression (by a northern blot or quantitative PCR) and protein

expression (by western blot) would be performed. Additionally, a measurement of GPX

enzyme activity could be performed using a commercially available total GPX activity

assay (Total Glutathione Peroxidase Assay Kit, ZeptoMetrix Corp., Buffalo, NY). While

it might not be possible to directly measure GPX4 activity, the difference in total activity

between the wildtype and variant cell lines could be determined.

The oxidative stress response of the cell lines would also need to be measured.

For these studies, both the wildtype and variant cell lines would be exposed to an

oxidative stressing agent such as H2O2, paraquat, radiation, or even a chemotherapeutic drug associated with the generation of ROS such as doxorubicin or bleomycin.65, 134

Cellular response could be measured by population doubling times of the cultured cells, and by measuring for markers of apoptosis. Additionally it would be important to assay for the products of oxidative stress within the cell culture system. Because GPX4 catalyzes the reduction of phospholipid hydroperoxides, screening for evidence of lipid peroxidation would be an important measure of oxidative stress. This could be performed by a commercially available TBARS (Thiobarbituric Acid Reactive

Substances) assay which is designed to measure lipid peroxidation and can be performed on cultured cells (TBARS Assay Kit, ZeptoMetrix Corp., Buffalo, NY).139

The best possible outcome of this research would be to determine that a polymorphism, or biomarker, is clinically relevant in the development of SMN in HL patients. This would greatly improve patient screening strategies aimed at the prevention of a SMN, and improve the overall quality of life in cancer survivors.

57

Figure 3.1: SNPlex Genotyping Chemistry An overview of SNPlex chemistry is presented in the figure. The combination of an oligonucleotide ligation assay (OLA) and multiplex PCR form the basis for the SNPlex reaction. Allele specific oligo (ASO) probes designed with 3’ end specificity enable allelic discrimination to be achieved. During the OLA assay, a locus specific oligo (LSO) probe binds near site of the SNP, and in combination with the ASO probes, creates an OLA reaction product following a phosphorylation and ligation reaction. Target amplification of the OLA reaction product is achieved through the use of universal priming sites on the ASO and LSO probes. Following electrophoresis on the AB 3730, base calls are distinguished by the differences in size and color provided by mobility modifiers and fluorescent labels on the ZipChute probes

58 Figure 3.1

Courtesy of Applied Biosystems

59

Figure 3.2: GeneMapper Graphical Plots The spatial separation of alleles allows for automated base calling during GeneMapper analysis. A polor plot (shown on the left) is a graphical representation of allele calls as grouped into clusters. SNP probes falling into tight clusters with clear separation generate the most unambiguous base calls. In a Cartesian plot (shown on the right), allele calls are grouped along the plot axes, with separation based on signal strength. In each plot, red circles represent allele 1 homozygotes, green triangles represent heterozygotes, and blue squares represent allele 2 homozygotes.

60

Figure 3.2 61

Figure 3.3: GeneMapper Samples Plot Through the use of ZipChute probes, (containing a fluorescent dye and a mobility modifier), electrophorectic separation of SNPlex products on the AB 3730 genetic analyzer allowed for accurate base calls of individual SNPs. The GeneMapper samples plot demonstrates how alleles are assigned on the basis of peak size and fluorescent reading. In the plot, peaks falling within the pink and gray bins are automatically assigned a base call as long as control parameters are satisfied. Three DNA samples with differing genotypes are shown in the plot.

62 Figure 3.3 63

Figure 3.4: Causes of SNP Attrition in the SNPlex Genotyping Method After extensive bioinformatics work and a review of the relevant literature, a starting pool of 114 polymorphisms was assembled. SNP dropout occurred during all steps of the genotyping process with only 31% of the original SNP pool available for the final data analysis. Attrition rates were nearly equal at each stage with a mean of 23%. The percentage of the remaining SNP pool relative to the starting pool is noted in parenthesis.

64 Figure 3.4

65

Table 3.1: Type of SMN/NMSC within the Subset of Genotyped HL Survivors The table shows the distribution SMNs and NMSCs diagnosed in 754 HL patients who were successfully genotyped during our study. Radiation therapy related cancers (breast and thyroid) were found to be the most frequent SMNs in the cohort. Basal cell carcinoma was the most frequently observed NMSC in the cohort. A total of 34 patients were diagnosed with both a SMN and NMSC. This data is representative only of HL survivors within the CCSS for whom buccal cell DNA was available for study.

66 Table 3.1

Second Primary Malignant Neoplasms within the Subset of 754 HL Survivors Successfully Genotyped for Antioxidant Related Polymorphisms

Second Malignant Neoplasm Number of Cases

Breast Cancer 70 Thyroid Cancer 20 Melanoma 6 CNS Tumor 5 Female Uro-Genital Tract 5 Adenocarcinoma 5 Lymphoma 4 Myeloid Leukemia 3 Other Malignancies 14 Total 131

Non-Melanoma Skin Cancer Number of Cases

Basal Cell Carcinoma 111 Squamous Cell Carcinoma 6 Total 117

67

Table 3.2: SNPs Included in the Final Statistical Analysis of the HL Cohort The table lists 36 polymorphisms which were included in the final statistical analysis of the HL cohort. The polymorphism location, dbSNP identifier, major allele, and cohort frequency are also listed.

68 Table 3.2

Major Allele and Location Relative to HL Cohort Gene Gene Coding Region db SNP ID Alleles Frequency

GPX1 (Chromosome 3) Upstream rs3811699 A/G A 0.70 Upstream rs3448 C/T C 0.73 Upstream rs8179164 A/T A 0.98 Upstream rs1987628 C/T C 0.70

GPX2 (Chromosome 14) 5' UTR rs2296327 A/G G 0.79 P126L rs17881652 C/T C 0.98 Intronic rs2412065 C/G G 0.79 Downstream rs11623705 G/T G 0.89 Downstream rs1064108 C/T C 0.70

GPX3 (Chromosome 5) Upstream rs3763015 A/G G 0.63 Intronic rs3792795 A/G G 0.92 3'UTR rs8177448 A/G G 0.99 3'UTR rs8177449 C/T C 0.92 Downstream rs8177834 A/G G 0.88

GPX4 (Chromosome 19) Upstream rs6843 A/G G 0.77 3'UTR rs713041 C/T C 0.55 Downstream rs2302109 G/A A 0.51

SOD2 (Chromosome 6) V16A rs4880 C/T T 0.53 Intron rs2758331 A/C C 0.54 Intron rs4523113 A/T T 0.76 Downstream rs5746136 A/G G 0.69

SOD3 (Chromosome 4) Upstream rs1089065 C/G C 0.57 Upstream rs800416 G/T G 0.99 Upstream rs1405689 A/G G 0.68 Upstream rs1089074 C/T C 0.99

Continued

69 Table 3.2 continued

Major Allele and Location Relative to HL Cohort Gene Gene Coding Region db SNP ID Alleles Frequency

CAT Upstream rs564250 C/T C 0.79 (Chromosome 11) Intronic rs2300182 A/T A 0.88 Intronic rs2300181 A/G G 0.71 Intronic rs17883260 C/T C 0.63 Downstream rs475043 A/G A 0.63 Downstream rs1535721 C/T C 0.78

NOS3 (Chromosome 7) Upstream rs11771844 A/G G 0.92 Upstream rs10277237 A/G G 0.77 Intronic rs1800783 A/T T 0.59

CYP2C9 (Chromosome 10) *10 E272G rs9332130 A/G A 0.99 *3 I359L rs1057910 A/C A 0.93

70

Table 3.3: Associations between SMN or NMSC and Genotype by Cox Regression Analysis Polymorphisms in antioxidant related genes were tested by Cox regression analysis to estimate the risk of developing a SMN or NMSC in HL survivors. Three outcomes were measured by this model including, SMN vs no SMN/NMSC, NMSC vs. no SMN/NMSC, and SMN/NMSC vs. no SMN/NMSC. Genotypes were compared to a reference with a hazard ratio (HR) of 1 in order to determine if allele combinations were associated with increased or decreased risk of SMN (p-values <0.05 are highlighted in bold).

71 Table 3.3

SMN or NMSC No SMN/NMSC Gene Chr SNP Outcome Genotype N % N % HR 95% CI p

GPX1 3 rs3448 SMN vs. no SMN/NMSC C/C 57 16.4 290 83.6 1 C/T 62 23.8 199 76.2 1.535 1.071 2.201 0.0196 T/T 10 18.2 45 81.8 1.228 0.627 2.405 0.5501 C/C or C/T 119 19.6 489 80.4 1 T/T 10 18.2 45 81.8 1.005 0.527 1.917 0.9888 C/C 57 16.4 290 83.6 1 C/T or T/T 72 22.8 244 77.2 1.483 1.048 2.101 0.0263 NMSC vs. no SMN/NMSC C/C 71 19.7 290 80.3 1 C/T 40 16.7 199 83.3 0.873 0.591 1.289 0.4945 T/T 5 10 45 90 0.598 0.241 1.484 0.2679 C/C or C/T 111 18.5 489 81.5 1 T/T 5 10 45 90 0.63 0.257 1.546 0.3132 C/C 71 19.7 290 80.3 1

72 C/T or T/T 45 15.6 244 84.4 0.831 0.57 1.209 0.333 SMN/NMSC vs. no SMN/NMSC C/C 108 27.1 290 72.9 1 C/T 91 31.4 199 68.6 1.22 0.922 1.615 0.1644 T/T 13 22.4 45 77.6 0.951 0.534 1.692 0.8638 C/C or C/T 199 28.9 489 71.1 1 T/T 13 22.4 45 77.6 0.872 0.497 1.529 0.6321 C/C 108 27.1 290 72.9 1 C/T or T/T 104 29.9 244 70.1 1.178 0.899 1.545 0.235

Continued

Table 3.3 continued

SMN or NMSC No SMN/NMSC Gene Chr SNP Outcome Genotype N % N % HR 95% CI p

GPX3 5 rs8177834 SMN vs. no SMN/NMSC A/A 5 41.7 7 58.3 1 A/G 28 20.6 108 79.4 0.388 0.15 1.006 0.0514 G/G 96 18.6 421 81.4 0.367 0.149 0.903 0.0292 A/A or A/G 33 22.3 115 77.7 1 G/G 96 18.6 421 81.4 0.859 0.578 1.276 0.4506 A/A 5 41.7 7 58.3 1 A/G or G/G 124 19 529 81 0.372 0.152 0.91 0.0303 NMSC vs. no SMN/NMSC A/A 2 22.2 7 77.8 1 A/G 19 15 108 85 0.474 0.11 2.046 0.3174 G/G 96 18.6 421 81.4 0.637 0.157 2.586 0.5277 A/A or A/G 21 15.4 115 84.6 1 G/G 96 18.6 421 81.4 1.271 0.785 2.06 0.3291 A/A 2 22.2 7 77.8 1

73 A/G or G/G 115 17.9 529 82.1 0.604 0.149 2.446 0.4796 SMN/NMSC vs. no SMN/NMSC A/A 5 41.7 7 58.3 1 A/G 42 28 108 72 0.42 0.166 1.064 0.0675 G/G 165 28.2 421 71.8 0.454 0.186 1.107 0.0824 A/A or A/G 47 29 115 71 1 G/G 165 28.2 421 71.8 1.013 0.73 1.404 0.9403 A/A 5 41.7 7 58.3 1 A/G or G/G 207 28.1 529 71.9 0.447 0.184 1.086 0.0755

Continued

Table 3.3 continued

SMN or NMSC No SMN/NMSC Gene Chr SNP Outcome Genotype N % N % HR 95% CI p

SOD2 6 rs5746136 SMN vs. no SMN/NMSC A/A 14 19.7 57 80.3 1 A/G 61 22.3 213 77.7 1.093 0.611 1.955 0.7651 G/G 55 17.4 262 82.6 0.749 0.416 1.35 0.3359 A/A or A/G 75 21.7 270 78.3 1 G/G 55 17.4 262 82.6 0.697 0.492 0.989 0.043 A/A 14 19.7 57 80.3 1 A/G or G/G 116 19.6 475 80.4 0.899 0.516 1.569 0.7089 NMSC vs. no SMN/NMSC A/A 13 18.6 57 81.4 1 A/G 46 17.8 213 82.2 0.909 0.491 1.682 0.7605 G/G 58 18.1 262 81.9 0.794 0.433 1.454 0.4545 A/A or A/G 59 17.9 270 82.1 1 G/G 58 18.1 262 81.9 0.856 0.592 1.237 0.4071 A/A 13 18.6 57 81.4 1

74 A/G or G/G 104 18 475 82 0.843 0.473 1.502 0.5614 SMN/NMSC vs. no SMN/NMSC A/A 22 27.8 57 72.2 1 A/G 95 30.8 213 69.2 1.079 0.678 1.715 0.7495 G/G 96 26.8 262 73.2 0.822 0.516 1.31 0.4106 A/A or A/G 117 30.2 270 69.8 1 G/G 96 26.8 262 73.2 0.774 0.589 1.016 0.0652 A/A 22 27.8 57 72.2 1 A/G or G/G 191 28.7 475 71.3 0.935 0.601 1.456 0.7672

Continued

Table 3.3 continued

SMN or NMSC No SMN/NMSC Gene Chr SNP Outcome Genotype N % N % HR 95% CI p

GPX4 19 rs6843 SMN vs. no SMN/NMSC A/A 12 30.8 27 69.2 1 A/G 38 19.1 161 80.9 0.651 0.34 1.248 0.1964 G/G 71 18.1 322 81.9 0.52 0.282 0.96 0.0367 A/A or A/G 50 21 188 79 1 G/G 71 18.1 322 81.9 0.731 0.508 1.052 0.092 A/A 1230.82769.21 A/G or G/G 109 18.4 483 81.6 0.56 0.308 1.017 0.057 NMSC vs. no SMN/NMSC A/A 12 30.8 27 69.2 1 A/G 36 18.3 161 81.7 0.682 0.337 1.38 0.2868 G/G 61 15.9 322 84.1 0.505 0.258 0.988 0.046 A/A or A/G 48 20.3 188 79.7 1 G/G 61 15.9 322 84.1 0.689 0.469 1.012 0.0574 A/A 1230.82769.21

75 A/G or G/G 97 16.7 483 83.3 0.557 0.29 1.071 0.0795 SMN/NMSC vs. no SMN/NMSC A/A 21 43.8 27 56.3 1 A/G 66 29.1 161 70.9 0.698 0.419 1.165 0.1693 G/G 113 26 322 74 0.547 0.336 0.891 0.0153 A/A or A/G 87 31.6 188 68.4 1 G/G 113 26 322 74 0.731 0.551 0.969 0.0292 A/A 2143.82756.31 A/G or G/G 179 27 483 73 0.594 0.37 0.955 0.0314

Table 3.4: A Summarized List of Assocation Results by Cox Regression Analysis A summarized list of findings (p <0.05) as determined by Cox regression analysis is presented in the table. Polymorphisms were correlated with both increased and decreased risk of SMN as shown by the hazard ratio (listed in bold).

76 Table 3.4

SMN or NMSC No SMN/NMSC Major Cohort Chr Gene SNP Allele Frequency Genotype Outcome N % N%HR95% CI p

3 GPX1 rs3448 C 0.73 C/T SMN vs. no SMN/NMSC 62 23.8 199 76.2 1.535 1.071 2.201 0.0196 C/T or T/T SMN vs. no SMN/NMSC 72 22.8 244 77.2 1.483 1.048 2.101 0.0263

5 GPX3 rs8177834 G 0.88 G/G SMN vs. no SMN/NMSC 96 18.6 421 81.4 0.367 0.149 0.903 0.0292 A/G or G/G SMN vs. no SMN/NMSC 12419529810.372 0.152 0.91 0.0303

19 GPX4 rs6843 G 0.77 G/G SMN vs. no SMN/NMSC 71 18.1 322 81.9 0.52 0.282 0.96 0.0367 G/G NMSC vs. no SMN/NMSC 61 15.9 322 84.1 0.505 0.258 0.988 0.046 G/G SMN/NMSC vs. no SMN/NMSC 11326322740.547 0.336 0.891 0.0153 G/G SMN/NMSC vs. no SMN/NMSC 11326322740.731 0.551 0.969 0.0292 A/G or G/G SMN/NMSC vs. no SMN/NMSC 17927483730.594 0.37 0.955 0.0314 77 6 SOD2 rs5746136 G0.69 G/G SMN vs. no SMN/NMSC 55 17.4 262 82.6 0.697 0.492 0.989 0.043

Table 3.5: Associations between SMN Classification and Genotype by Cox Regression Analysis Cox regression models were used to estimate the risk of developing a specific type of SMN by a comparison of genotypes among HL survivors. Genotypes were compared to a reference with a hazard ratio (HR) of 1 in order to determine if allele combinations were associated with increased or decreased risk of SMN The category of breast or thyroid cancer was also included in the analysis to determine the risk of radiation related SMNs in our cohort.

78 Table 3.5

Breast SMN No SMN/NMSC Chr Gene SNP Genotype N % N % HR 95% CI p

3 GPX1 rs3448 C/C 27 8.5 290 91.5 1 . . . C/T 36 15.3 199 84.7 1.905 1.155 3.14 0.0115 T/T 6 11.8 45 88.2 1.63 0.672 3.952 0.28 C/C or C/T 63 11.4 489 88.6 1 . . . T/T 6 11.8 45 88.2 1.188 0.513 2.749 0.6876 C/C 27 8.5 290 91.5 1 . . . C/T or T/T 42 14.7 244 85.3 1.86 1.146 3.018 0.012

5 GPX3 rs8177834 A/A 3 30 7 70 1 . . . A/G 14 11.5 108 88.5 0.302 0.087 1.055 0.0607 G/G 52 11 421 89 0.3 0.093 0.965 0.0435 A/A or A/G 17 12.9 115 87.1 1 . . . G/G 52 11 421 89 0.872 0.504 1.508 0.6232 A/A 3 30 7 70 1 . . . A/G or G/G 66 11.1 529 88.9 0.301 0.094 0.96 0.0425

11 CAT rs1535721 C/C 42 11.4 326 88.6 1 . . . C/T 19 9.3 186 90.7 0.76 0.442 1.307 0.3209 T/T 8 24.2 25 75.8 2.03 0.949 4.34 0.0679 C/C or C/T 61 10.6 512 89.4 1 . . . T/T 8 24.2 25 75.8 2.228 1.062 4.677 0.0342 C/C 42 11.4 326 88.6 1 . . . C/T or T/T 27 11.3 211 88.7 0.933 0.575 1.513 0.7786

Thyroid SMN No SMN/NMSC Chr Gene SNP Genotype N % N % HR 95% CI p

7 NOS3 rs1800783 A/A 2 2 98 98 1 . . . A/T 3 1.2 248 98.8 0.579 0.097 3.467 0.5497 T/T 11 5.7 182 94.3 2.739 0.607 12.36 0.1901 A/A or A/T 5 1.4 346 98.6 1 . . . T/T 11 5.7 182 94.3 3.933 1.366 11.323 0.0111 A/A 2 2 98 98 1 . . . A/T or T/T 14 3.2 430 96.8 1.522 0.346 6.699 0.5785

Continued

79 Table 3.5 continued

Breast or Thyroid SMN No SMN/NMSC Chr Gene SNP Genotype N % N % HR 95% CI p

3 GPX1 rs3448 C/C 36 11 290 89 1 . . . C/T 43 17.8 199 82.2 1.695 1.088 2.641 0.0197 T/T 7 13.5 45 86.5 1.389 0.618 3.125 0.4265 C/C or C/T 79 13.9 489 86.1 1 . . . T/T 7 13.5 45 86.5 1.08 0.498 2.341 0.8464 C/C 36 11 290 89 1 . . . C/T or T/T 50 17 244 83 1.644 1.071 2.525 0.0231

5 GPX3 rs8177834 A/A 4 36.4 7 63.6 1 . . . A/G 18 14.3 108 85.7 0.32 0.108 0.949 0.0399 G/G 64 13.2 421 86.8 0.306 0.111 0.844 0.0221 A/A or A/G 22 16.1 115 83.9 1 . . . G/G 64 13.2 421 86.8 0.838 0.516 1.362 0.4763 A/A 4 36.4 7 63.6 1 . . . A/G or G/G 82 13.4 529 86.6 0.309 0.113 0.846 0.0223

7 NOS3 rs1800783 A/A 14 12.5 98 87.5 1 . . . A/T 29 10.5 248 89.5 0.85 0.449 1.61 0.6181 T/T 40 18 182 82 1.473 0.798 2.717 0.2157 A/A or A/T 43 11.1 346 88.9 1 . . . T/T 40 18 182 82 1.649 1.069 2.543 0.0237 A/A 1412.59887.5 1 . . . A/T or T/T 69 13.8 430 86.2 1.123 0.631 1.998 0.694

11 CAT rs1535721 C/C 52 13.8 326 86.2 1 . . . C/T 24 11.4 186 88.6 0.785 0.484 1.273 0.3265 T/T 10 28.6 25 71.4 2.037 1.032 4.019 0.0402 C/C or C/T 76 12.9 512 87.1 1 . . . T/T 10 28.6 25 71.4 2.212 1.141 4.291 0.0188 C/C 52 13.8 326 86.2 1 . . . C/T or T/T 34 13.9 211 86.1 0.958 0.622 1.476 0.846

Continued

80

Table 3.6: A Summary of Association Results by Cox Regression Analysis Classified by Type of SMN or NMSC A summarized list of associations between genotype and SMN (p<0.05) are shown in the table. Cox regression models were used to estimate the risk of developing a specific type of SMN in the HL cohort as compared across genotype. Polymorphisms were correlated with both increased and decreased risk of SMN as shown by the hazard ratio (listed in bold).

81 Table 3.6

SMN No SMN/NMSC Major Cohort Chr Gene SNP Allele Frequency Genotype Type of SMN N % N % HR 95% CI p

3 GPX1 rs3448 C 0.73 C/T Breast SMN 36 15.3 199 84.7 1.905 1.155 3.14 0.0115 C/T or T/T Breast SMN 42 14.7 244 85.3 1.86 1.146 3.018 0.012 C/T Breast or thyroid SMN 43 17.8 199 82.2 1.695 1.088 2.641 0.0197 C/T or T/T Breast or thyroid SMN 50 17 244 83 1.644 1.071 2.525 0.0231

3 GPX1 rs1987628 C 0.7 T/T Basal cell carcinoma 15 26.8 41 73.2 1.734 1.006 2.991 0.0476

7 NOS3 rs1800783 T 0.59 T/T Breast or thyroid SMN 40 18 182 82 1.649 1.069 2.543 0.0237 T/T Thyroid SMN 11 5.7 182 94.3 3.933 1.366 11.323 0.0111

11 CAT rs1535721 C 0.78 T/T Breast SMN 8 24.2 25 75.8 2.228 1.062 4.677 0.0342 T/T Breast or thyroid SMN 10 28.6 25 71.4 2.037 1.032 4.019 0.0402 T/T Breast or thyroid SMN 10 28.6 25 71.4 2.212 1.141 4.291 0.0188 82

5 GPX3 rs8177834 G 0.88 A/G or G/G Breast SMN 66 11.1 529 88.9 0.301 0.094 0.96 0.0425 G/G Breast SMN 52 11 421 89 0.3 0.093 0.965 0.0435 G/G Breast or thyroid SMN 64 13.2 421 86.8 0.306 0.111 0.844 0.0221 A/G or G/G Breast or thyroid SMN 82 13.4 529 86.6 0.309 0.113 0.846 0.0223 A/G Breast or thyroid SMN 18 14.3 108 85.7 0.32 0.108 0.949 0.0399

19 GPX4 rs6843 G 0.77 G/G Basal cell carcinoma 59 15.5 322 84.5 0.49 0.25 0.959 0.0374

Table 3.7: Associations between SMN or NMSC and Genotype by Poisson Multivariate Regression Analysis Multivariate Poisson regression analyses were used to adjust for patient specific effects on the risk of developing a SMN or NMSC. Genotype hazard ratios were adjusted for treatment (chemotherapy and radiation Y/N), sex (M/F), and age at diagnosis of HL (0-9 y, 10-16y, 17-21y). Outcomes were compared across genotypes and findings (p <0.05) are presented in the table.

83 Table 3.7

SMN or NMSC Major Cohort Chr Gene SNP Allele Frequency Genotype Yes No Odds Ratio 95% CI p

15 GPX4 rs6843 G 0.77 G/G 109 302 1.000 1.000 1.000 - A/G 64 155 1.105 0.736 1.633 0.6240 A/A 20 24 2.027 1.042 3.613 0.0246

11 CAT rs1535721 C 0.78 T/T 14 23 1.000 1.000 1.000 - C/T 67 177 0.495 0.265 0.992 0.0348 C/C 124 305 0.534 0.301 1.036 0.0445

84

Table 3.8: Haplotype Analysis using Poisson Regression Modeling Haplotype analysis was used to assess the association of genotype by linked loci and the risk SMN or NMSC in the HL cohort. Haplotypes were grouped by gene and Poisson regression modeling was used to account for the effects of treatment related variables.

85 Table 3.8

Haplotype Analysis

GPX1 Chromosome 3 SMN or NMSC Parameter Yes No OR 95% CI p CHEMO_YN No . . 1 1 1 .

Yes . . 0.884 0.64 1.22 0.4523 RAD_YN No . . 1 1 1 . Yes . . 2.486 0.734 8.42 0.1434 SEX female . . 3.261 2.097 5.071 <.0001 male . . 1 1 1 . Age at Dx 0-9 . . 1 1 1 . 10-16 . . 3.93 1.326 11.648 0.0136 86 17-21 . . 4.814 1.594 14.54 0.0053 Major Allele and Haplotype HL Cohort SNP Order Alleles Frequency Haplotype A_C_A_C 89.7 214.5 1.082 0.968 1.208 0.1651 rs3811699 A/G A 0.70 A_T_A_C 52.2 124.9 1.128 1.019 1.248 0.0199 rs3448 C/T C 0.73 A_T_T_C 4 13.1 1.133 0.837 1.535 0.4194 rs8179164 A/T A 0.98 G_C_A_T 60.5 153 1 1 1 . rs1987628 C/T C 0.70

SECTION II

ATYPICAL X CHROMOSOME INACTIVATION IN AN X;1 TRANSLOCATION PATIENT DIAGNOSED WITH OTOPALATODIGITAL SYNDROME

87

CHAPTER 4

X-CHROMOSOME INACTIVATION AND X;AUTOSOME TRANSLOCATIONS

4.1: X-Chromosome Inactivation

4.1.1: Introduction

X chromosome inactivation (XCI) is an epigenetic process that occurs in mammalian females as a mechanism of gene dosage compensation.140, 141 The transcriptional silencing of most X-linked genes on one X chromosome occurs early during embryogenesis and is maintained through the cell lineage. XCI has evolved as a dosage compensation mechanism to minimize the differences in gene expression between

XY males and XX females.141

XCI is typically a random process in which either the maternal or paternal X chromosome is inactivated in a cell. Normal females are therefore mosaics in regard to the pattern of inactivation. When a non-random XCI pattern is observed (with greater than 80% of one X chromosome preferentially inactivated) a female may be described as

“skewed.” Skewing may occur due to chance alone, as only a limited number of precursor cells are present in the inner cell mass at the time XCI is initiated.142, 143 This is a rare finding though, as the distribution of XCI in females has been shown to follow a bell-shaped curve.144 The most common cause of non-random inactivation is due to an X

88 chromosome abnormality which confers a proliferative disadvantage in cells bearing an

active mutant X.145, 146 Skewing may be constitutional or tissue specific, depending on

the underlying cause.

The XIST gene, located within the X inactivation center of the chromosome

(Xq13.2), is responsible for initiating the process of XCI in mammalian females.147 XIST produces a transcript (X-inactive specific transcript) that functions in cis during the

inactivation process to coat the length of the inactive X chromosome (Xi) to maintain

gene silencing.148 Even autosomal material may be subject to silencing by XIST due to the spread of inactivation, as is the case in certain X;autosome translocations that retain

Xq13.2.149-151

4.1.2: Genes that Escape X-Inactivation

While a majority of genes are silenced on the Xi, it is estimated that 15% of genes escape XCI and remain active.152, 153 Genes within the pseudoautosomal regions (PAR) of the X chromosome are thought to escape inactivation, as a Y-chromosome counterpart

is present in males allowing for bi-allelic expression of these genes.141 Y homologues that escape XCI are also known to exist outside of the PAR region in the X conserved region (XCR) that is present in all mammals.154 Additionally, there are genes outside of

the PAR region, and with no known Y-homologue, that escape inactivation.

In genes known to escape XCI, expression from the Xi is typically thought to be lower than expression from the active X (Xa) though accurate measurement has proved difficult.152, 153, 155-157 In the most extensive characterization of the X chromosome to date, Willard and Carrel measured how often a gene escaped XCI using human-mouse somatic cell hybrid (SCH) lines retaining an Xi chromosome.153 While many genes

89 produced unambiguous results (either always or never escaping XCI), approximately

10% of genes showed variable patterns of inactivation dependent on the hybrid. Based

on these findings, the authors suggested that X-linked gene expression is a heterogenous

process and more variable than previously thought.

With the advent of array technology, a number of studies relating to the

expression status of X-linked genes have been performed.158-160 Several of these studies

have attempted to correlate expression array data with genes previously thought to escape

XCI, with varying results. In an analysis by Craig et al., expression array data were

found to correlate with a majority of genes previously thought to escape XCI.158

Alternately, in a study by Talebizadeh et al., the authors attempted to compare in-vivo expression array data with SCH data generated by Willard and Carrel.153, 160 While there was some agreement between the each system, the pattern was not consistent enough to allow for a direct correlation between SCH and array data. Results may have varied because the location and number transcripts measured per gene were not identical in each study. Furthermore, like many in-vitro systems, these data suggest that additional modifying processes may occur in-vivo that are not capable of being replicated in cell culture.

4.2: X;Autosome Translocations

4.2.1: Introduction to Translocations

A translocation is a rare event resulting in the transfer or exchange of material between two non-homologous chromosomes.161, 162 Many different forms of

translocations may occur, though reciprocal translocations may be among the most common. While the exact mechanism of formation is unknown, translocations are

90 thought to be the result of a recombination between non-allelic regions of homology, that

likely occur during meiosis.161 Translocations may be classified as de novo or inherited and may be present in either an unbalanced or balanced form.

4.2.2: Balanced X;Autosome Translocations

A balanced X;autosome translocation t(X;A)is a unique structural rearrangement that occurs as the result of a reciprocal exchange of genetic material, without apparent loss or gain, between the X chromosome and an autosome. Female carriers of a t(X;A) display phenotypic heterogeneity that may be classified into four broad categories; (1) a normal phenotype with or without a history of miscarriage; (2) gonadal dysfunction; (3) a known X-linked disorder; or (4) congenital abnormalities and developmental delay.161, 163

Factors such as the position of the translocation breakpoints, XCI status, and the mode of inheritance, all affect the phenotypic findings in a t(X;A) carrier.

4.2.3: Phenotypic Consequences of Translocation Breakpoint Location

The position of the breakpoint is a determining factor in t(X;A) carrier phenotype

relative to its impact on gene expression and chromatin environment. In respect to

gonadal dysfunction, studies have suggested that a critical region between Xq13 and

Xq26 is necessary for normal ovarian function, as breakpoints within this region are

associated with premature ovarian failure.164-166 Alternately, the phenotypic traits of a defined X-linked disorder may be observed in a female t(X;A) carrier if the breakpoint interrupts a critical gene or regulatory region on the X-chromosome. In instances such as this, the translocation carrier may be defined as a manifesting female, as she displays the traits of an X-linked disorder typically only seen in males.163 The degree to which the t(X;A) carrier is affected is likely to be modified by her XCI status. Manifesting females

91 with known X-linked disorders who carry a t(X;A) have been used in genetic mapping

experiments to aid in the cloning of disease genes. This approach has been used to clone

the causative genes for Duchenne muscular dystrophy, X-linked lissencephaly, and X-

linked mental retardation among others.167-171

Translocations may be associated with an increased risk of phenotypic abnormality even when the breakpoints do not directly interrupt a gene. This phenomenon is called a position effect, and is caused by a change in chromatin environment or interruption of a critical gene regulatory region.171-174 As a consequence of the translocation breakpoint, gene expression is altered resulting in phenotypic abnormalities. Position effects have been documented to cause a variety of human disease, including aniridia, campomelic dysplasia, holoprosencephaly, X-linked deafness, and sex reversal among many others.175-181 Position effect mutations are noteworthy because there are often great distances reported between the translocation breakpoint and the causative gene. Distances of several hundred kilobases have been commonly reported, while some position effect mutations function at distances of 1 Mb.173, 176 The exact mechanism of many position effect mutations is unknown. In the case of a translocation, a gene may become physically separated from a promoter, repressor or other regulatory element that acts in cis to regulate expression. Alternately, local chromatin environment may be disrupted by the chromosomal rearrangement resulting in a change in gene expression at an epigenetic level.173, 174

4.2.4: XCI in a t(X;A) Carrier

XCI is modifying factor in the phenotypic heterogeneity found in carriers of an

t(X;A). Skewed XCI is a frequent observation in balanced t(X;A) carriers, with the

92 derivative X chromosome typically found to remain active182, 183 Atypical XCI in female t(X;A) carriers has been documented and is often associated with an abnormal phenotype.184-187 Approximately 19% of t(X;A) carriers demonstrate a variable pattern of

XCI, while in approximately 5% of reported cases, the normal X chromosome remains

active in a majority of cells.188

An abnormal phenotype that is associated with atypical XCI may be due to monosomy of autosomal genes (due to the spread of XCI), and functional disomy of X- linked genes translocated onto the autosome. The spread of XCI into translocated autosomal material has been extensively studied. Results suggest that XIST is capable of acting in cis on autosomal material, resulting in gene silencing, although spreading may

be incomplete and discontinuous.151, 182, 189, 190

4.2.5: The Impact of Mode of Inheritance in a t(X;A) Carrier

The mode of inheritance can also impact phenotypic findings, as de novo t(X;A)

are significantly more likely to be associated with congenital abnormalities and

development delay.163, 183 The risk of an abnormal phenotype is higher in cases of a de novo translocation in comparison to carriers of inherited translocations.191 This` is always the case because a de novo structural rearrangement has not been previously demonstrated to be benign in a phenotypically normal parent. Further examination of an apparently balanced de novo t(X;A) may actually demonstrate that it is unbalanced at a molecular level. Additionally, there is always the risk that a critical gene or regulatory region may be interrupted, resulting in an abnormal phenotype. For all of these reasons, female carriers of an X;A translocation are at increased risk for phenotypic abnormalities.

93

CHAPTER 5

OTOPALATODIGITAL SYNDROME SPECTRUM DISORDERS AND FILAMIN A

5.1: Introduction to Otopalatodigital Syndrome

Otopalatodigital syndrome (OPD) is an X-linked semi-dominant disorder that was

first described by Taybi in 1962, and further characterized by Dudding in 1967.192, 193

The clinical features of OPD include hearing loss, cleft palate, dysmorphic facies, and

skeletal dysplasia. The classification of OPD syndrome is dependent on the severity of

disease, with type 1 being a milder form of the disorder [OMIM #311300] , and type 2

characterized as a more severe form that also includes CNS anomalies, cardiac defects,

severe skeletal malformations, and the potential for embryonic or neonatal lethality

[OMIM #304102].

5.2: Phenotypic Features of OPD

Males affected with OPD1 are described in the literature to have characteristic dysmorphic facies including a flattened nasal bridge, supraorbital hyperostosis, hypertelorism, microstomia, dental abnormalities, and cleft palate, along with conductive hearing loss, and generalized skeletal dysplasia.192, 193 Digital anomalies include a broadening of the thumbs and great toes, fifth finger clinodactyly, lengthened second fingers and toes, and a shortening of the great toe. Chest deformities and short stature are

94 also characteristics of the disorder. Mild mental retardation may be associated with

OPD1, though some researchers claim that intellect is unimpaired.194-198

Females affected with OPD1 exhibit variable expressivity of the disorder.199

Females may range from having a very mild, sub-clinical expression to a complete expression of the disorder, similar to that of affected males.196, 198, 199 X-chromosome inactivation (XCI) is likely to be the major modifier of expressivity in females.

5.3: Molecular Characterization of the OPD-Spectrum Disorders

OPD1 was first hypothesized to be an X-linked disorder based on the inheritance pattern observed in the pedigree of affected families.200 In order to further characterize the disorder, multi-generational OPD1 families were used in mapping studies to narrow the region of the causative gene by linkage analysis. Following this approach, the gene for OPD1 was linked to distal Xq and subsequently refined to Xq28.201, 202

Though OPD2 was long hypothesized to be allelic to OPD1 on the basis of

phenotypic overlap, the first evidence of allelism was demonstrated in 2001, as Robertson

reported the linkage of OPD2 to the Xq28 region.203 Similar lines of evidence suggested that two additional disorders, including Frontometaphyseal dysplasia (FMD) [OMIM

#305620], and Melnick-Needles Syndrome (MNS) [OMIM #309350] were allelic to

OPD.203-207 The major clinical features of each disorder are listed in table 5.1.

FMD is classified as an X-linked recessive disorder in which affected males demonstrate severe supraorbital hyperostosis and skeletal dysplasia.208, 209 Additional craniofacial features include hypertelorism, down slanting palpebral fissures, and dental anomalies. A bowing of the long bones and conductive deafness are also frequent observations in FMD males. Features unique to FMD, as compared to the related

95 disorders, include tracheal stenosis and joint contractures.194 FMD affected females demonstrate a pronounced supraorbital hyperostosis, hypertelorism, and joint contractures though variable expressivity of the disorder is observed.208, 210

MNS is an X-linked dominant disorder that is almost entirely female-limited due

to the embryonic lethal phenotype of nearly all affected males.211 Clinical features in

MNS females include supraorbital hyperostosis, micrognathia, prominent eyes, and full

cheeks. Additional features include irregularity of the long bones, and decreased lung

function due to abnormalities in the ribs and chest cavity.212 Males surviving to term have a severe phenotype that includes skeletal dysplasia, craniofacial anomalies, cardiac defects and obstructive uropathy.194 The phenotypic features of MNS males are quite

similar to those observed in OPD2 males.

A formal confirmation of allelism between OPD types 1 and 2, FMD, and MNS,

occured in 2003, as Robertson et al. reported that the causative mutations for each

disorder were found in the FLNA gene.213 All four disorders are now collectively

recognized as the Otopalatodigital syndrome spectrum disorders (OPSD).

The FLNA gene is located on Xq28 and codes for an actin binding protein known as filamin A. Mutations within the FLNA gene have been previously reported to cause periventricular nodular heterotopia (PH), a neuronal migration disorder.214 Classical bilateral PH [OMIM # 300049] is an X-linked dominant disorder associated with embryonic lethality in affected males.215 Females affected with the disorder typically

present with seizures, cerebellar hypoplasia, cardiac defects, and occasionally mild

mental retardation. In PH males surviving to term, phenotypic findings include seizures, severe mental retardation and multiple congenital anomalies.216

96 While the mode of pathogenesis is unknown, FLNA mutations within the OPSD

are thought to confer a gain-of-function effect on the protein product, filamin A.213 There are several lines of evidence to support this hypothesis. In OPSD mutations reported to date, all are missense mutations or in-frame deletions that conserve the translational

reading frame of FLNA.194 Additionally, western blot and immunostaining experiments have demonstrated that full length filamin A protein, capable of binding to actin, is produced in male OPSD patients.194 These findings are in contrast to the frameshift or

nonsense mutations observed in PH, which are reported to cause a loss-of-function due to

aberrant transcript production and a truncation of the filamin A protein.214, 217, 218

5.4: Filamin A Protein

Producing a transcript nearly 9 kb in length, FLNA codes for a 280 kDA protein known as filamin A.219, 220 As a cytoskeletal organizing protein, a major role of filamin A

is to cross-link actin into a network or stress fiber.221 Recently, the role of filamin A has been further elucidated with evidence of involvement in cell signaling and second

messenger pathways.222-224

The filamin A protein is composed of multiple domains and functions as a dimer

within the cell (Figure 5.1). At the amino-terminus of the protein is the actin binding

domain which consists of two calponin homology domains. The ability to bind actin is

mediated by the calponin homology domains and has been demonstrated in other proteins

such as dystrophin, α-actinin, and β-spectrin and utrophin.225, 226 Two rod domains comprise the remainder of the protein, and are composed of 24 structurally homologous repeats in the form of a β-pleated sheet.219, 227 The rod domains are joined by hinge regions, which also function as the site of calpain cleavage.228 At the carboxy terminus

97 of the protein, a dimerization domain exists in the 24th structural repeat.219, 229 Filamin A is highly homologous to two other actin binding proteins found in mammals, known as filamin B and filamin C. The composition of all three proteins is similar, with divergence found mainly in the hinge regions, and the 5’ and 3’UTRs.230 Among all 3 human

filamin proteins, overall amino acid sequence homology ranges between 60-80%.

Filamin A is a promiscuous binding partner capable of interacting with a wide

variety of proteins.222-224 Therefore, in addition to the primary role of organizing the

actin cytoskeleton, filamin A is thought to play an important role in cell signaling.

Molecules including protein kinase C, mitogen activated protein kinase SEK, and Smad

are known utilize filamin A as a scaffold or anchor, in order to participate in signal

transduction and phosphorylation reactions.224 Additionally, many of the proteins which

interact with filamin A are described as extracellular matrix receptors, such as β1-

integrin. When a cell is subjected to shear stress, β1-integrin associates with filamin A in order to produce a protective stiffening response.231 Filamin A is also known to be

associated with transmembrane proteins involved in cellular locomotion and adhesion.

This relationship has been observed among interactions between Ras-GTPases and

filamin A. Ras-related GTPases including Rac, Rho, Cdc42 and Ral A are all known to

bind with filamin A and function as molecular switches in the cell to regulate signal

transduction pathways in key cellular functions such proliferation or differentiation.232

There is evidence that filamin A may organize actin filaments as mediated by interactions with Ras-GTPase proteins in response to cellular stimuli.233-235

98 5.5: OPSD Genotype - Phenotype Correlations

Within the OPSD there is a relationship between disease phenotype and the

location of the pathogenic FLNA mutation (Figure 5.1). In cases of typical OPD1 and

OPD2, mutations have been reported in the actin binding domain of filamin A.194

Mutations causative for MNS demonstrate extreme specificity, as all reported patients were found to have a mutation within exon 22 of the FLNA gene (corresponding to filamin repeat 10). Finally, mutations in FMD were found to be the most variable, and were interspersed throughout filamin repeats 10, 14, 15 and 23. While the majority of

OPSD mutations exhibit clustering, this finding is in contrast to mutations causing PH, as they are found to be distributed throughout the entire FLNA gene.236

FLNA mutation screening studies have been performed on individuals diagnosed

with an OPSD. In one study, 70% of OPD2 patients were found to have a detectable

FLNA mutation.203 Screening in two FMD cohorts revealed that 43% and 57% of

patients had a mutation detectable during analysis.208, 213 Aside from possible misdiagnosis, these data imply that the pathogenesis of OPSD may be caused by mutations residing outside of the FLNA coding regions, with the potential involvement of other genes.

5.6: X-Chromosome Inactivation and OPSD

XCI pattern has been examined in relation to the OPSD, with non-random XCI

frequently observed in females with a heterozygous FLNA mutation. Skewing is biased toward the X chromosome carrying the mutant FLNA allele. This finding is in contrast to

XCI studies performed in patients with PH and a documented FLNA mutation, as a random pattern of X-inactivation was observed.214

99 Within the OPSD, the degree of skewing may be correlated with disease severity,

as Robertson et al. observed that the most marked skewing was associated with the more

severe phenotypes.213 This finding may be variable though, as no relationship between

degree of skewing and phenotype was found in a family diagnosed with MNS.212

Skewed XCI within the OPSD is thought to be caused by the selective growth disadvantage of cells carrying an active mutant FLNA allele.213 As hypothesized by

Zenker et al., OPSD females carrying milder FLNA mutations may demonstrate a more severe manifestation of the disease as a consequence of the less restrictive growth disadvantage on mutant cells. Reciprocally, a severe mutation would incur a greater selective disadvantage on cells with an active mutant allele resulting in a less pronounced phenotype.210

100

Figure 5.1 A Model of the Filamin A Dimer and Location of Pathogenic Mutations A filamin A dimer is depicted in the figure. Filamin A is composed of distinct protein domains, with important roles in actin binding, dimerization, and protein structure. Mutations within filamin A are causative for OPSD and PH. Pathogenic mutations are depicted relative to the location within the protein. While mutations in the OPSD seem to cluster into regions, this finding is not observed in relation to PH.

101 Figure 5.1

102

Table 5.1: Phenotypic Features of FLNA–Related Disorders OPD types 1 and 2, FMD, MNS, and PH are known to be allelic disorders. Common phenotypic features found within each disorder are listed in the table. Additional information regarding mode of inheritance, the location of the mutation, and disease pathogenesis are also included.

103 Table 5.1 Phenotypic Features of FLNA - Related Disorders Location XCI Mode of of FLNA Pattern in Disorder Inheritance Sex Phenotypic Features Mutations Pathogenesis Females

Prenatal or Hyper- Bilateral Agenesis of Neonatal Skeletal extensible Obstructive Hearing Cleft Craniofacial Cardiac Mental Nodular the Lethality Dysplasia Joints Uropathy Loss Palate Defects Defects Retardation Heterotopia Seizures Cerebellum X-linked semi- OPD1 dominant F - ●●/--●●/- ●● - ●/----ABD Gain of Function Skewed M - ●●● --●● ● ●●● ● ●/---- X-linked semi- ABD and OPD2 dominant F - ●● --●●/- ●● - ●/-- -RD Gain of Function Skewed M ●● ●●● ●/- ●● ●●● ●● ●●● ●● ●● --● X-linked ABD and FMD recessive F - ● --●●●● -- ---RD Gain of Function Skewed M ● ●●● ● ●●● ●●● - ●●● ● ---- X-linked

104 MNS dominant F - ●●● - ●● ●● - ●●● ●/-- - - -RD Gain of Function Skewed M ●●● ●●● ●/- ●●● Unknown - ●●● ●● ●● --- X-linked ABD and PH dominant F ------●● ●/- ●●● ●●● ●● RD Loss of Function Random M ●●● ●/------●● ●● ●●● ●●● ●●

● represent the frequency of the observed phenotypic finding ABD = Actin Binding Domain RD = Rod Domain

CHAPTER 6

ATYPICAL X-CHROMOSOME INACTIVATION IN AN X;1 TRANSLOCATION PATIENT DIAGNOSED WITH OTOPALATODIGITAL SYNDROME

This work was performed in collaboration with Dr. Andrew Lidral’s laboratory at the University of Iowa.

6.1 Abstract

X-chromosome inactivation (XCI) is an epigenetic process used to regulate gene dosage in mammalian females by silencing one X-chromosome. While the pattern of

XCI is typically random in normal females, abnormalities of the X-chromosome may result in skewing due to disadvantaged cell growth. We describe a female patient with an

X;1 translocation [46,X, t(X;1)(q28;q21)dn] and unusual pattern of XCI who was clinically diagnosed with Otopalatodigital syndrome (OPD) type 1. There was complete skewing of XCI in the patient in the tissues examined, along with the atypical findings of an active normal X-chromosome and an inactive derivative X. An X-linked disorder,

OPD1 is characterized by multiple congenital anomalies including skeletal abnormalities, craniofacial defects, and hearing loss. Mutations within the FLNA gene (Xq28) are known to cause OPD, though none were detected in our patient. Additionally, no abnormalities in FLNA mRNA or protein were detected in our patient. Characterization of the translocation revealed that the patient’s Xq28 breakpoint interrupts the DKC1

105 gene, located 400kB distal to FLNA. Molecular analysis of the breakpoint region

revealed functional disomy of Xq28 genes distal to DKC1. Monosomy of 1q genes was not detected. Possible explanations for the patient’s phenotype include a position effect due to the translocation breakpoint, an undetected FLNA-related mutation, or altered gene dosage due to consequences of atypical XCI.

6.2: Introduction

X-chromosome inactivation (XCI) is a critical epigenetic process that regulates

gene dosage in mammalian females. During early embryogenesis, the majority of X-

linked genes on one X-chromosome are transcriptionally silenced. XCI occurs randomly

in female somatic cells as a dosage compensation mechanism between XX females and

XY males, and is maintained through the cell lineage.140 Cytogenetic or molecular

abnormalities on the X-chromosome may influence XCI, resulting in a non-random or

“skewed” pattern due to a survival disadvantage of the affected cells.237 Skewed XCI is a

common finding in a balanced X;autosome translocation, as the derivative X chromosome [der(X)] typically remains active, while the normal X is inactivated. This pattern is observed due to a preferential loss of cells containing an imbalance in genetic

material.163

Otopalatodigital syndrome (OPD) is an X-linked semi-dominant disorder

characterized by multiple congenital anomalies including skeletal abnormalities,

craniofacial defects, and conductive hearing loss.192, 193 The clinical features of OPD syndrome vary with the severity of disease, and are classified as two types, OPD1 and

OPD2 [OMIM 311300; OMIM 304120]. OPD2 is the more severe and often lethal form, associated with cardiac defects, central nervous system conditions, and midline

106 defects.204, 206, 238 OPD types 1 and 2 have been grouped with similar disorders under the

broader category of OPD-spectrum disorders (OPSD), which also includes Melnick-

Needles syndrome (MNS) [OMIM 309350] and frontometaphyseal dysplasia (FMD)

[OMIM 305620].206, 213 OPD types 1 and 2 are semidominant, while MNS is dominant and FMD is recessive. These disorders were hypothesized to be allelic even before the causative mutations for all four phenotype classifications were reported in the FLNA

gene.204-206, 213

Located on Xq28, the FLNA gene produces a transcript that encodes a widely expressed 280kD protein, filamin A. As an actin binding protein, filamin A dimers crosslink actin in order to create three-dimensional structures which serve as scaffolds in the cytoskeleton. Filamin A is also known to be a promiscuous binding partner capable of interacting with over 45 proteins, suggesting that it serves multiple roles in cell signaling.224, 236

FLNA mutations in the OPSD are hypothesized to be gain-of-function.213 To date, all reported OPSD FLNA mutations are missense mutations or in-frame deletions that conserve the translational reading frame (www.hgmd.cf.ac.uk). Studies have shown that a full length filamin A protein, capable of associating with actin, is produced in males with a FLNA mutation.194 These findings are in contrast to FLNA mutations causing

periventricular heterotopia (PH) [OMIM 300049]. Mutations in PH (a neuronal

migration disorder) are described as loss-of-function, and are predominately frameshift or

nonsense mutations that produce an aberrant transcript and truncated protein.214

XCI has previously been studied in the OPSD, and in heterozygous affected females skewing is frequently observed.213 The X-chromosome carrying the normal

107 FLNA allele typically remains active, though the degree of skewing is variable.212 This finding is in contrast to FLNA mutations leading to PH, in which heterozygous females display a random XCI pattern.

In this report, we describe a female patient clinically diagnosed with OPD1 syndrome with an X;1 translocation and unusual pattern of XCI. We discuss the patient’s phenotypic findings in relation to a molecular and cytogenetic analysis of the Xq28 region including the FLNA gene.

6.3: Materials and Methods

6.3.1: Subjects and Consent

Informed consent was obtained for the patient and her parents with IRB approval granted by Columbus Children’s Research Institute.

6.3.2: Tissue Sources, Cell Lines, DNA and RNA Preparations

Peripheral blood lymphocytes (PBL) and buccal swabs were obtained from the patient and parents. PBL were used to create immortalized lymphoblastoid cell lines

(LCL) by Epstein-Barr Virus (EBV) transformation using the Marmoset cell line

GM07404E (Coriell, Camden, NJ). Somatic cell hybrid (SCH) lines were established by polyethylene glycol fusion of -sensitive Chinese Hamster Ovary (CHO) cells with patient lymphocytes(created by Andrew Lidral).239 Separate hybrid cell lines containing a normal X chromosome, a der(X) chromosome, a normal chromosome 1 and a derivative chromosome 1 were isolated. DNA and RNA were isolated from PBL, cell lines, and buccal swabs according to standard procedures (Qiagen, Valencia, Ca; Gentra,

Minneapolis, MN).

108 6.3.3: Molecular and Cytogenetic Breakpoint Mapping

BAC clones from the RPCI-11 panel (Invitrogen, Carlsbad, CA) were used to

map the X-chromosome breakpoint region by fluorescence in-situ hybridization (FISH).

Information on mapping and sequence data was obtained from the Ensembl Genome

Browser (http://www.ensembl.org), the National Center for Biotechnology Information,

(http://www.ncbi.nlm.nih.gov), and the University of California Santa Cruz

(http://genome.ucsc.edu). To create FISH probes, DNA was first isolated from BAC

clones (Qiagen, Valencia, Ca), then biotin-labeled by nick translation (Invitrogen,

Carlsbad, CA). The BAC probes were hybridized to metaphase chromosome spreads

prepared from the patient’s EBV-LCL. Signal was detected using Texas Red-labeled

avidin, and nuclei were counter-stained with DAPI. Concurrent with the effort to map

the breakpoint by FISH, SCH lines were analyzed for the presence or absence of markers

in Xq28 and 1q21 by locus-specific PCR (performed by Andrew Lidral).

6.3.4: X-Inactivation Analysis

To determine XCI pattern, a CAG triplet repeat in the first exon of the androgen

receptor (AR) gene (Xq11.2-Xq12) was analyzed for methylation status using bisulfite

treated DNA.240 Methylation-specific PCR (MSP) was performed using fluorescently-

labeled primers as described by Kubota,241 which were designed to amplify either the unmethylated (active) or methylated (inactive) X chromosome using modified DNA

(Table 6.1). Additionally, unmodified DNA was amplified in order to determine allele size of the AR triplet repeat. PCR products were electrophoresed on an AB 310 Genetic

Analyzer to determine genotype using Genescan software (Applied Biosystems, Foster

City, CA).

109 6.3.5: Expression - RT-PCR

Reverse-transcription PCR (RT-PCR) was used to study the expression of genes near the translocation breakpoints (partly performed by Andrew Lidral). Total RNA was extracted from EBV-LCL and SCH lines (RNeasy Mini Kit, Qiagen, Valencia, CA) and was used for first strand cDNA synthesis (Superscript II RT, Invitrogen, Carlsbad, CA).

Approximately 100ng of cDNA served as a template in PCR reactions. Standard PCR conditions were utilized with a 55° annealing temperature (Table 6.1).

6.3.6: Sequencing

The breakpoint was mapped by walking across chromosomes X and 1 with pairs

of primers to determine which derivative chromosome had positive PCR results

(performed by Andrew Lidral). Once the breakpoint was mapped to less than 1 Kb,

primer pairs were designed to amplify across breakpoint from each derivative

chromosome and the product sequenced. Sequencing was extended around both

breakpoints by the use of overlapping primers to amplify DNA from derivative and

normal chromosome. All sequencing was performed in both directions. BLAST was used to determine the location of the sequence inserted into the derivative X breakpoint region.242 Transfac Matrix Database (v7.0), Biobase was used to screen for transcription factor binding sites and sequence conservation was evaluated using the Vertebrate Multiz

Alignment & PhastCons Conservation (28 Species) track on the UCSC Genome

Bioinformatics website (performed by Andrew Lidral).243-245

Primers designed to flank the coding exons and intron-exon junctions of the

FLNA gene were used for PCR amplification of patient DNA and subsequent sequence analysis (Table 6.1). Additionally, the 3’untranslated region (UTR) of the FLNA gene

110 was sequenced from the patient, parents, and SCH lines using PCR products amplified from DNA and cDNA. Big Dye version 1.1 dye terminator chemistry was used in all sequencing reactions and the products were run by capillary electrophoresis on an AB

3130 genetic analyzer (Applied Biosystems, Foster City, CA).

6.3.7: Northern Blot

RNA isolated from EBV-LCLs was used for northern blot analysis (RNeasy Mini

Kit, Qiagen, Valencia, CA). Approximately 5.5ug of RNA was run on a denaturing formaldehyde gel (1%) and following electrophoresis, RNA integrity was ensured by visualization with ethidium bromide. Prior to transfer, the blot was denatured in 0.05N

NaOH-0.15M NaCl for 20 minutes, and then neutralized in 0.1M Tris-HCl-0.15M NaCl for 30 minutes. RNA was transferred to a nylon membrane (Duralon-UV, Stratagene, La

Jolla, CA), using a pressure blotting system (PosiBlot, Stratagene, La Jolla, CA), followed by UV crosslinking (Spectronics, Westbury, NY). A partial FLNA cDNA clone including the 3’UTR, (Genecopoeia, Inc., Germantown, MD ) was biotin labeled (Pierce,

Rockford, IL) and used to probe for FLNA transcripts during hybridization.

Chemiluminescence was used for signal detection (Pierce, Rockford, IL). After stripping, the blot was re-probed using β-Actin cDNA as a loading control (Clontech,

Mountain View, CA). The relative ratio of FLNA as compared to β-actin was quantified using Labworks software (UVP, Inc., Upland, CA).

6.3.8: Expression Array Analysis

Patient and parental RNAs (isolated from PBL) were converted to cDNA for use in microarray analysis (NuGEN Inc, San Carlos, CA). For each array, 2.2 µg of cDNA was hybridized to Human 133 Plus 2.0 GeneChips (Affymetrix Inc., Santa Clara, CA),

111 containing probe sets that measure 54,676 transcripts from Human RNA. The probe set signals were generated using the RMA algorithm in ArrayAssist 3.4 (Stratagene, La Jolla,

CA) and were used to determine differential gene expression by pair-wise comparisons.

Genes in the patient that were altered by a two-fold increase or decrease (as compared to parental DNA), as well as genes in proximity to the translocation breakpoint regions were sorted for further interpretation.

6.3.9: Comparative Genomic Hybridization

Array comparative genomic hybridization (aCGH) was used to rule out additional chromosomal abnormalities in the patient. For aCGH, EBV-LCL DNA derived from the patient was hybridized to a Spectral chip 2600 platform (PerkinElmer, Waltham, MA) containing 2600 BAC clones. Clones were spaced at approximately1 Mb intervals across the entire genome to ensure adequate coverage for detection of genomic imbalance. Both sex matched and sex mis-matched DNA were used as a normal hybridization controls during separate experiments.

6.3.10: Western Blot

Protein was extracted from EBV-LCLs of the patient and parents. Cells were combined with M-PER (mammalian protein extraction reagent) and Halt protease inhibitor cocktail (Pierce, Rockford, IL) before undergoing multiple freeze thaw cycles to disrupt the cell membrane. Following centrifugation to remove the cellular debris, a protein extract containing both cytoplasmic and nuclear proteins was obtained.

Protein (20ug) was separated by electrophoresis on a 4-20% Tris-glycine gel

(Invitrogen, Carlsbad, CA). Following electrophoresis, proteins were transferred to a nitrocellulose membrane. The membrane was blocked for 1 hour in 5% dry milk with

112 TBST at room temperature. A primary antibody against filamin A (ABP-280, 1:250

dilution) was used to detect protein (BD Biosciences, San Jose, CA). The membrane was

hybridized overnight at 4°C to reduce background. Following a wash procedure using

TBST, the membrane was hybridized in the presence of a secondary antibody labeled with peroxidase (Anti-mouse IgG, 1:10,000) for 1 hour at room temperature (Vector

Laboratories, Burlingame, CA). Enhanced chemiluminescence was used for detection, and the image was captured using the UVP EC imaging system (UVP, Upland, CA).

Filamin A protein was quantitated relative to a total protein stain at the appropriate molecular weight.

6.4: Results

6.4.1: Clinical Report

The patient was born to a 30 year old, gravida 2, para 2 female. The pregnancy

was complicated by the detection of a cleft lip in the fetus on ultrasound. Subsequent amniocentesis revealed an apparently balanced X;1 translocation

[46,X,t(X;1)(q28;q21)dn] (Figure 6.1). A neonatal examination was significant for a unilateral cleft lip, midline cleft palate, hypertelorism, a broad and flat nasal bridge, small low-set ears, preauricular pits, proximally positioned thumbs, short distal phalanges, fifth digit clinodactyly and camptodactyly, and a significant hemangioma over the sacrum. A brain MRI revealed an abnormal and hypoplastic corpus collosum.

The patient is followed on a regular basis by a clinical geneticist (Dr. Annmarie

Sommer). Despite receiving growth hormone therapy for several years, she continues to

be extremely small in size with her height and weight proportionate (Figure 6.2). At the

113 age of 8 years she was 115.5 cm tall (below the 3rd centile), weighed 22.1 kg (at the 3rd centile), and head circumference measured 50.5 cm (10th centile).

At the most recent physical examination, facial features included persistent hypertelorism and a very broad and depressed nasal bridge with a prominent tip. A scar from the cleft lip repair is present on the philtrum and her cleft palate was repaired. The patient has small, cup-shaped auricles. Her neck was supple and the thorax, (including heart and lung examination) was normal. The abdominal exam did not reveal any abnormalities. The extremities were symmetrical, with all digits tapering distally, ulnar deviation of both index fingers, and 5th finger clinodactyly. All small joints are

hypermobile.

Radiological findings included a delayed bone age, bilateral coxa valga and a

steep left acetabular angle. An MRI of the brain revealed a very small corpus callosum,

with almost no splenuim or posterior elements; the myelination pattern was normal.

The patient has some developmental delay. At age 8 years, she was in 3rd grade in a

special needs school and doing well. Her speech was improving. However, she was still

not toilet trained. The family history is negative for birth defects, mental retardation, or genetic disease.

6.4.2: Breakpoint Characterization

Cytogenetic and molecular techniques were used to define the patient’s

translocation breakpoint. Multiple BAC clones in the Xq28 region were used to narrow

the critical region by FISH (Table 6.2), and BAC RP11-115M6 was found to span the

translocation breakpoint in the patient (Figure 6.3). Mapping of the SCH lines by PCR

identified the X-chromosome breakpoint to basepair 153,644,715 within the first intron

114 of the DKC1 gene on Xq28 (Figure 6.4). The breakpoint at 1q21 is located at base pair

153,386,489 within the 35 Kb intergenic span between the DPM3 and KRTCAP2 genes.

Sequencing across the breakpoints of both derivitive chromosomes revealed the deletion of 16bp of chromosome X involving bps 153,644,716 to 153,644,731 (Mar 2006, NCBI

Bld 36.1) and 36 bp from chromosome 1 involving bps 153,386,474-153,386,509 (Mar

2006, NCBI Bld 36.1). In addition, a duplicate copy of 93bp from 153,384,859-

153,384951 (Mar 2006, NCBI Bld 36.1) of chromosome 1 was found inserted into the breakpoint on the derivative X. None of these, nor the surrounding sequences, are within highly conserved regions, except the nearby DKC1 exons. The chromosome 1 breakpoint is within a simple (TA)n repeat bounded by two SINE alu repeats, and the chromosome X

breakpoint region has a significant regulatory potential.246

6.4.3: X-Chromosome Inactivation Status

XCI analysis was performed on patient DNA from buccal cells, PBL, EBV-LCL and SCH lines (Figure 6.5). Parents were analyzed using PBL DNA and EBV-LCL

DNA. The patient displayed a completely non-random XCI pattern with near 100% skewing in all sources (within a 5% limit of detection). Analysis of SCH lines established that the patient’s normal X was active and maternally inherited, while the der(X) was inactive and of paternal origin. The patient’s mother was found to have a random XCI pattern in DNA from PBL.

6.4.4: RT-PCR Analysis

RT-PCR was used to determine the expression of critical genes near the breakpoint regions (Table 6.3). Examination of the normal X SCH line indicated it was the active X-chromosome, as transcripts were detected for all X-chromosome genes

115 studied. Analysis of the der(X) SCH revealed a pattern largely representative of an

inactive X chromosome, with the majority of genes not actively producing transcripts.

Gene expression distal to each breakpoint (1q21 and Xq28) was studied in the

derivative chromosome SCH lines to determine if partial monsomy or functional disomy was occurring in these regions. The spread of XCI was not detected in RT-PCR analysis of chromosome 1 genes, as genes distal to the 1q21 breakpoint were expressed in the isolated der(X) hybrid. In Xq28, RT-PCR results suggested that genes distal to the breakpoint were expressed from the translocated chromosome X material on the der(1).

Functional disomy of this region is a concern, as it is likely that the normal X

chromosome is also expressing these genes.

RT-PCR findings of significance include the expression of DKC1, from only the

normal X-chromosome, apparently due to the disruption of the gene on the der(X) by the

translocation breakpoint. Additionally, FLNA expression was noted in both the normal X

and the der(X) SCH lines using multiple primer pairs. FLNA is typically thought to

undergo XCI as previously demonstrated by Carrel.153 While this finding was suggestive

of functional disomy, it should be noted that the level of expression from the der(X) was

less (though not quantified) than the expression level seen in the normal X-chromosome

(Figure 6.6).

6.4.5: Molecular Analysis of FLNA

The FLNA gene was extensively analyzed in our patient at the levels of DNA,

RNA, and protein. At the DNA level, whole exon deletions (≥ 1exon) of the FLNA gene were excluded by PCR using SCH DNA and EBV-LCL DNA (data not shown). DNA sequence analysis of FLNA did not detect evidence of a mutation in the coding regions or

116 exon-intron boundaries of the gene, although a polymorphism (*88C>T) was discovered

in the patient’s 3’UTR (Figure 6.7). To our knowledge, this base change has not been

reported in a database or literature source, though it was found by sequence analysis in

3/102 chromosomes from normal female controls sequenced by the laboratory (frequency

~3%). Analysis of parental DNA revealed that the patient inherited the polymorphism from her heterozygous mother.

At the level of transcription, the *88C>T polymorphism in the FLNA 3’UTR

served as a marker for allelic expression. Sequence analysis using FLNA RT-PCR

products determined that only the variant (T) allele was expressed in the patient (cDNA

derived from PBL and EBV-LCL). Analysis of maternal cDNA (derived from PBL)

established bi-allelic expression of the FLNA gene. Results from SCH cDNA indicated

that the isolated normal X expressed the variant (T) allele, while the der(X) expressed the

wildtype (C) allele. To determine if the variant allele altered mRNA folding, secondary

structure was predicted using Mfold analysis software247. Localized structure analysis,

(which included the last exon and 3’UTR of the FLNA gene) suggested that the variant

(T) may alter mRNA folding (Figure 6.8).

Additional RNA studies were performed by northern blot analysis using a FLNA cDNA probe (exon 37-3’UTR). No evidence of alternate splicing was detected in the patient within the limits of detection of the assay. Expression levels were quantified relative to ACTB, with FLNA expression found to be equivalent between our patient and a normal female control (Figure 6.9).

At a translational level, filamin A protein of the appropriate molecular weight was detected in our patient at levels equivalent to parental controls (Figure 6.10).

117 6.4.6: Expression Array Analysis

An Affymetrix U133 chip was used to assess genome wide expression in our

patient compared to parental controls. Gene expression that was increased or decreased

by two-fold as compared to parental samples was further analyzed in an attempt to

explain the patient’s phenotype. Additionally, genes near the translocation breakpoint

regions were examined for possible monosomic or disomic expression. Notably, FLNA and DKC1 gene expression were found to be normal in the patient. Monosomic

expression of genes distal to the 1q21 breakpoint was not observed in the patient.

Evidence of functional disomy in Xq28 is apparent from the microarray results, as genes distal to the breakpoint region demonstrate increased levels of expression. A comparison of expression array data and RT-PCR results are presented in Table 6.4.

6.4.7: Array Comparative Genomic Hybridization

No additional chromosomal abnormalities were detected in the patient by aCGH at a 1 Mb level.

6.5: Discussion

The X;1 translocation patient described in this study displays an atypical pattern

of XCI and clinical features suggestive of OPD1 syndrome. Clefts of the primary palate

are rarely reported in female patients with OPD.194, 196 The occurrence in this patient may be the result of other gene(s) being affected by the translocation. Alternatively, her cleft

palate may suggest that FLNA is involved in the development of both the primary and

secondary palates. The latter is supported by observing FLNA expression during primary

palatogenesis in mice (Andrew Lidral unpublished data).

118 Despite extensive cytogenetic and molecular analysis of the patient, a definitive

cause for her symptoms was not established. Possible explanations for her phenotype

include: a position effect mutation due to the translocation breakpoint, an undetected

FLNA-related mutation, or altered gene dosage due to consequences of atypical XCI.

One possible cause of OPD1 syndrome and additional anomalies in our patient is

a position effect mutation caused by the translocation breakpoint in Xq28. Position effect mutations have been reported to cause a variety of genetic disorders including campomelic dysplasia, cleft lip and palate, aniridia, holoprosencephaly, X-linked deafness, and preaxial polydactyly among others.173, 178, 248, 249 In our patient, the Xq28 translocation breakpoint in DKC1 may potentially interrupt a long range cis-acting FLNA regulatory element and/or distort the chromatin structure thus altering FLNA expression and resulting in her OPD1 phenotype. The translocation breakpoint in DKC1 lies distal to the FLNA gene by approximately 400 Kb, a distance well within the reported range of up to 1Mb for position effect mutations to occur.173, 178, 248 There is not strong evidence for a chromosome 1 gene being involved in our patient since all of the tested chromosome 1 genes are expressed from both the normal and derivative 1 chromosomes.

Furthermore, the chromosome 1 breakpoint does not interrupt any genes or highly conserved regions.

The patient’s X-inactivation status is a confounding variable in the argument for a position effect mutation. In buccal cells and lymphocytes, the patient displays a completely skewed XCI pattern, with her normal X remaining active. This is unusual for a balanced X;autosome translocation, as the der(X) typically remains active in order to prevent the silencing of autosomal genes caused by the spread of XCI. The most

119 probable cause of the patient’s skewed XCI pattern is due to the interruption of the DKC1 gene by the translocation breakpoint. DKC1 encodes the 58kDa dyskerin protein that participates in rRNA processing and functions in telomere maintanence.250 Mutations in

DKC1 are known to cause dyskeratosis congenita (DKC) [OMIM 30500], a disorder characterized by nail dystophy, hyperpigmentation of the skin, and leukoplakia associated with aplastic anemia. Female carriers of a DKC1 mutation are reported to have a completely skewed X-chromosome lineage, with the X-chromosome bearing the mutant allele inactivated.251-253 One of the hallmarks of DKC is a defect in cell proliferation due

to bone marrow dysfunction. As a result, affected cells have a survival disadvantage over

normal cells, especially in rapidly dividing tissues. This post-inactivation cell selection

mechanism is the likely cause of our patient’s non-random XCI pattern. While skewing

was observed in two types of tissue, (PBL and buccal cells) it is important to note that

this pattern may not be representative of all tissues in the patient.

An undetected FLNA-related mutation is another possible explanation for the

phenotype of the patient, with the translocation not having any direct attributory effect.

No mutations were detected by sequence analysis of FLNA coding regions and exon-

intron boundaries in the patient. Additionally, large deletions were ruled out by

screening the isolated normal X and der(X) chromosomes in the SCHs. Two recent

studies screening for mutations within the coding regions of FLNA have been reported in

the literature. In an OPD2 cohort, 70% of patients had a detectable FLNA mutation,

while only 57% of patients with FMD had a detectable mutation.208, 213 In the latter

report, mutations were not always detected even in classic FMD patients. These data

imply that mutations within the coding region are not the only cause of FLNA-related

120 disease. The breakpoint in this patient suggests that a FLNA regulatory element may exist within the DKC1 genomic sequence and this is supported by the significant predicted regulatory potential. We suggest that the pathogenesis of disease may be more fully elucidated by screening both FLNA regulatory regions and FLNA binding partners.

Due to the size and complexity of the FLNA gene, and the ever increasing number of reported binding partners, this will be an arduous process. Furthermore, it is reasonable to speculate that OPSD may be multigenic in nature with mutations in a number of genes for pathways interacting directly or indirectly with FLNA.

The discovery of a *88C>T variant in the 3’ UTR region of FLNA presents a

challenge in characterizing this patient. XCI analysis and RT-PCR sequencing data suggest that only the FLNA allele carrying the variant (T) is expressed in PBL and EBV-

LCLs of the patient. On the contrary, results from RT-PCR data, and SCH sequence

analysis suggest that the FLNA gene may be expressed from both the patient’s der(X) and

normal X-chromosomes. One possible explanation is that the patient expresses very low

level FLNA transcripts from her der(X) chromosome which prove to be undetectable in

total RNA. The consequences of possible low level FLNA functional disomy are

unknown, but may be compatible with the theory that OPSD are the result of FLNA gain-

of-function mutations.

The data regarding OPSD-like phenotypes and Xq28 ploidy correlations are

ambiguous. Duplication of Xq28 including FLNA was found in a male with PH, mental

retardation, shortened fingers, clinodactyly and syndactyly which contrasts with PH being

associated with loss of function mutations.215 It is possible that the duplication reduced

FLNA levels; however, gene expression was not characterized in this patient. Two

121 additional patients with Xq28 duplications including FLNA were described as having

digital and palatal features consistent with OPSD, although the PH status was not characterized.254 That no PH was observed in two mouse models lacking filamin A

protein expression contradicts the expectation that loss of function is associated with

PH.255, 256 Hence, there isn’t a clear correlation between gene dose and phenotype to infer whether our patient’s phenotype is caused by FLNA loss of function or misregulation.

The lack of FLNA mutations in many patients with OPSD suggests that noncoding

mutations may cause altered regulation of FLNA expression and have the same functional

effect as missense mutations.

Mfold analysis was used to predict the effect of the *88 C>T polymorphism on

FLNA mRNA folding. Based on optimal structure predictions, the analysis suggests that

this base change is capable of altering FLNA secondary structure. An alteration in

mRNA stability is a possible consequence when considering the effect of a 3’UTR

variant.

Finally, it is possible that the patient’s phenotypic abnormalities arose due to

altered gene dosage caused by an atypical pattern of XCI. Consequences of the patient’s

XCI pattern include functional monosomy of 1q21 and functional disomy of Xq28. The

effects of XCI must be considered relative to the X-inactivation center of the

chromosome, specifically the XIST gene (Xq13.2). Because XIST is functional on the

patient’s der(X) chromosome, it has the potential to spread into translocated 1q material

resulting in monosomic gene expression. The spread of XCI is known to occur in a

discontinuous manner with the inactivation signal capable of crossing over 100Mb of

DNA.151 Although the spread of XCI into our patient’s autosomal material was not

122 observed by RT-PCR, only a limited number of genes were studied. Analysis of array

data suggests that monosomic gene expression on 1q was not occurring in this patient.

Genes distal to the Xq28 breakpoint were tested for functional disomy in our patient. Findings were compared to published XCI data from human-mouse SCH lines in order to confirm inactivation status.153 Functional disomy was presumed to occur in

Xq28 genes distal to the breakpoint on the basis of RT-PCR and expression array

analyses. In the patient, expression is likely to occur from both the active normal X-

chromosome as well as translocated Xq28 material on the der(1), as it is no longer subject

to XCI. Of the genes demonstrating functional disomy, the extent to which they would

have an impact on the patient’s phenotype is not known. Microarray analysis allowed for

a better quantification of gene expression, and revealed that a majority of genes distal to

the breakpoint demonstrated increased gene expression in comparison to parental

controls. Functional disomy in the Xq28 region has been reported in a number of other

patients, with the phenotypic findings recently compiled by Sanlaville.257 Common features among 19 patients included growth retardation, microcephaly, hyptonia, feeding difficulties, developmental delay, abnormal palate, hypoplastic genitalia, and increased susceptibility to infections.

While no previously reported FLNA binding partners were present in the list of disomic genes, a number of receptors, ion channels, and signaling molecules were included, such as CLIC2, (an intracellular chloride channel), and RAB39B (Ras-related

gene). These are interesting candidates as filamin A has previously been shown to

interact with another chloride channel protein, CFTR, as well as several RAS –related

proteins.234, 258

123 While the cause of OPD1 in the patient has not been fully elucidated, the most likely explanations were presented in this dissertation. Potential mechanisms for OPD1 pathogenesis in relation to FLNA include a position effect caused by the translocation breakpoint, an undetected FLNA-related mutation, or altered gene dosage due to atypical

XCI. Our hypothesis is based on the patient’s clinical findings, the close proximity of the

Xq28 breakpoint to the FLNA gene, and the relatively high percentage of FLNA mutation-negative patients within the OPSD. It is our hope that the findings presented in this dissertation may help to clarify the possible cause of FLNA-related disease in future patients.

124

Figure 6.1: X;1 Translocation Breakpoint A cytogenetic characterization of the patient’s balanced translocation, 46,X,t(X;1)(q28;q21)dn

125 Figure 6.1

126

Figure 6.2: Clinical Features of the Patient (A). The X;1 translocation patient with phenotypic features of OPD1. Prominent facial features include widely spaced eyes, small low-set ears, a broad nasal bridge, and a cleft lip and palate. (B). The patient at the age of 5 years. (C). A close up view of the patient’s hand. Notable features include, distally tapering digits, ulnar deviation of the index finger and 5th finger clinodactyly.

127 Figure 6.2

128

Figure 6.3 Fluorescence In-Situ Hybridization Analysis BAC RP11-115M6 (containing Xq28 sequence) spanned the translocation breakpoint, as the probe hybridized to the patient’s normal X, der(X) and der(1) chromosomes.

129 Figure 6.3

130

Figure 6.4: Map of the Xq28 Breakpoint Region The patient’s translocation breakpoint occurs within intron 1 of the DKC1 gene, as indicated by the solid black arrow. The FLNA gene is located proximal to the breakpoint at a distance of approximately 400kB. Gray arrows beneath FLNA and DKC1 indicate the direction of transcription.

131 Figure 6.4

132

Figure 6.5: X-Chromosome Inactivation Analysis At the AR locus, the patient inherited maternal allele 2 (M2) and the paternal allele (P)as shown by unmodified DNA. Methylation specific PCR using bisulfite modified DNA revealed complete skewing in the patient. Analysis of SCH DNA revealed that inactivation was biased toward the paternally derived der(X) chromosome (P), with the normal X remaining active (M2).

133 Figure 6.5

134

Figure 6.6: Disomic Expression of the FLNA Gene RT-PCR products are shown on an agarose gel. Expression was demonstrated from the patient, the normal X and der (X) SCH lines, and normal controls. Expression from both SCH lines is indicative of functional disomy of the FLNA gene in the patient.

135 Figure 6.6

136

Figure 6.7: Sequence Analysis of a 3’UTR FLNA Polymorphism The patient’s variant (T) allele was maternally inherited. Expression studies suggested that only the variant (T) was actively transcribed in the patient, unlike her mother who demonstrated expression from both alleles. Analysis of SCH cDNA revealed that the active normal X chromosome carried the (T) variant, while the inactive der(X) carried wildtype sequence.

137 Figure 6.7

138

Figure 6.8: Mfold Predictions of Optimal Secondary Structure Mfold software was used to predict the optimal secondary structure of the FLNA gene using both wildtype sequence (A) and the 3’ UTR polymorphism found in the patient (B). The results suggested that the *88C>T base change may influence RNA folding in FLNA transcripts.

139 Figure 6.8

140

Figure 6.9: Northern Blot Analysis of FLNA transcripts No evidence of alternate splicing was detected in patient RNA derived from her EBV- LCL. FLNA expression levels in the patient were equivalent those seen in the normal female control as compared to ACTB.

141 Figure 6.9

142

Figure 6.10 Western Blot Analysis of Filamin A Detection of the filamin A protein by western blotting was used to assess relative protein expression in our patient. The western blot revealed that the patient produces an appropriately sized filamin A protein in levels comparable to controls.

143 Figure 6.10

144

Table 6.1: Primer Sequences Primers used for PCR, sequencing, RT-PCR and methylation-specific PCR are listed in the table. Additional reaction conditions, including annealing temperature and MgCl2 , are listed.

145 Table 6.1

Primers used for PCR and Sequencing Annealing

Primer Name Forward Primer Sequence Reverse Primer Sequence Temp (°C) MgCl2 FLN1 2.1b CGCGCCTCAAAATGAGTAG GAAGGGGGTGGTTTGGAG 60 1.5mM FLN1 3b GGTGTCTACCTCCTCCTTGG AGGGAGGCTGTGAGTCTGG 60 1.5mM FLN1 4b CGCAGAGGCAGGAGAGACT TGTCACAGGCAGAAAACAGG 60 1.5mM FLN1 5b CTGTAGGGGACCGGATCG GCCACGTTTAGATGGGACAC 55 1.5mM FLN1 6b GACACCAGGAGGAGGTAGGG CACAGTAACCTGTCCCCAGAA 55 1.5mM FLN1 7b GGAATGTGCCTGACTTAGGG TGAGCCTTTGCTAAGAGCAG 55 1.5mM FLN1 8b GGGGGAGGCTTGTGACCT GAGGCACCTGCTCAGCTC 60 1.5mM FLN1 9b GATCTTTACGGCAGGTGAGG GCATGTGCCCAGACAGTAGA 55 1.5mM FLN1 10b ACAGTTCCTGGGGTCACAGT CTCTCCCTCTGCCAAGACAA 55 1.5mM FLN1 11b CCCAGTAAGTTGGCCTGGAG ATCAGGTGGGGAGGCAGA 60 1.5mM FLN1 12 CCACATGGGCTCTTCCTGCC TGGGCCGTCCTTGCCATCGTCT 60 1.5mM FLN1 13 AGACGATGGCAAGGACGGCCCA AGCAGGGCGAGACTTAGGCCAT 60 1.5mM FLN1 14A TGCCTGGGTTGTAGCTCTG GTGGCCTGCTGGTCAGTG 55 1.5mM FLN1 15b ACTTCGGGTCCAAGTCCAG GTGTGCCACAACCACTTGAA 55 1.5mM FLN1 16A GCTGAGGGAGCTTCTGAGTC GAGACCTCGCAGGGACAC 55 1.5mM FLN1 17A GGTGTCCCTGCGAGGTCT GGGAGGAGAAGGCCTTAGAG 55 1.5mM FLN1 18b TCCTCTTTGCTGACCAGGTG GAGGAACACACAGGGACCAT 55 1.5mM FLN1 19 CATCAAGGGTAGGAGGGCTT GCTGCTGCATGAGGAGGCTG 60 1.5mM FLN1 20 CAGCCTCCTCATGCAGCAGC CCTGGATGTGACAAAGGCCT 60 1.5mM FLN1 21 AGGCCTTTGTCACATCCAGG CACAGTGGGTTCTACCCTTA 55 1.5mM FLN1 22.2b CCGGGGACTACAACATCAAC GGCAGAGGGGAATGTAGGTA 55 1.5mM FLN1 23 TGCCGTGGGGTATGTGACGG CCGTCTGCCAGCCTGTGGGA 60 1.5mM FLN1 24b TGGAGTACACGCCTTACGAG AAACAGACAGCCGGTCATTC 55 1.5mM FLN1 25A CTGGGAACCCCAGGAATG TGGGGACGAGCAGACAGT 55 1.5mM FLN1 26 CTGGGACTGTCTGCTCGTCC CCTCACAGGACACTGCCCT 60 1.5mM FLN1 27b TCGGGTGGATGAGGATAAAG GAGTAGGCAGCACCTTGACC 55 1.5mM FLN1 28 TGCTGAGAACCTGTCTGACT CCGCAGCCCACACTCCAGCC 60 1.5mM FLN1 29 GCTACTCGGGCTTGGGCCAA AGCGAGCCCTTGCACACAGG 60 1.5mM FLN1 30 CCTGTGTGCAAGGGCTCGCT GGTCATGCTCAGCTCCGGAA 55 1.5mM FLN1 31 AACCCGAGCTCACACGCTGG CAGACACCCCTGCTGACCTA 55 1.5mM FLN1 32/33 ACAACAGACTCTCCAGCAGC TCCCGAGCTCCTTCCCAAGT 55 1.5mM FLN1 34 AGACTGTGCCAATGAGCTGC TTGTGGAATGGCAGCCTCGT 55 1.5mM FLN1 35 ACCTCACAGAGAGATGGGGC CAGGAGTATCTCCTGAGTCC 55 1.5mM FLN1 36A CCTCAGTGCAGGCCAACT CCTGCCACCCGTTTCTGT 55 1.5mM FLN1 37b CACTGGGAGCAGTGACAGAA CGAATGAAATCCCTGGACAC 55 1.5mM FLN1 38A CAGGGGGTTCAGAAAGGAG AGGGGATGGATACCCCTGAG 55 1.5mM FLN1 39A GCATAGCACCGAGGCTCA GGCCCTGGTGTAGTGAGG 55 1.5mM FLN1 40b AGCACGTGCCTGGTGAGT GGCTCCACCCCTCCTCTT 55 1.5mM FLN1 41 TCAAGCAGCCCCAAGAGGAG TGGCCAACGCAGGAGAGCGA 55 1.5mM FLN1 42 GGCATGTCTACCTTGGCTAT TGGGTCCAATACCCACACTC 55 1.5mM FLN1 43A AGGCTGTTCGTGGGAGGT CGGTGGCTCTGGTCTGTC 55 1.5mM FLN1 44A CCTTTCCGTCCTCCCTTG GGCTGCTTACAGAAGCGGTA 55 1.5mM FLN1 45 CATCTGCTGGTTTGAGGAGG CTCCTGTTGTCACCAAGAGC 55 1.5mM FLN1 46b ACAACAGGAGGCACCTGGA AGAGTGGGTGGGGCTAAGAG 55 1.5mM FLN1 47 GCACCACAGCCACCTCTTAG CCATCCTGTGATTTCTGGCC 55 1.5mM FLN1 48b GTCACCCCAGAACTGGCTTA CAGACTCAGGGCACCACAAC 55 1.5mM

Continued

146 Table 6.1 continued

Primers used for RT-PCR Annealing

Primer Name Forward Primer Sequence Reverse Primer Sequence Temp (°C) MgCl2 SLC9A6 CTCCGCTGACAACAACACTC TGTGCAAATTTTACAAATCACTAAG 55 1.5mM L1CAM GATGAGACCTTCGGCGAGTA GCATCTCCTGTCCTGGACTC 55 1.5mM TKTL ATCGTCTCCAGTGCAAAAGC TCCGAACTAGGGATGAATGAA 55 1.5mM FLN GGGCTGAGCAAGGCCTAC CGGGGTTGAGGGGAAGAG 55 1.5mM EMD ACTTATGGGGAGCCCGAGTC CCTAGCTCTGAAGCCCAGAC 55 1.5mM RPL10 ACTGTGGCCAGGGTTCAC GGGGCAGCACATTGGAAG 55 1.5mM DNase1L1 ATGGGGAGGACACCACAGT GCAGCAACAGAACAGTGAGG 55 1.5mM TAZ CTACTTCCCCCGCTTTGG TCTAAAAGCCACGTTTGAGC 55 1.5mM ATP61P1 CCCAGCATCTACTCCTTCCA GAAAGCAACAACACGGACAC 55 1.5mM GDI GCCCAGGGCAAGTACATAG TCGAGGAGAACAGACACAGG 55 1.5mM XAP5 GTGCTGAGGAGCTGGTACG GCACACTGAGGGAGCTGA 55 1.5mM PLXA3 ACATGGATGCCTACCTGGTG AGGACGGCACAGGACTGA 55 1.5mM ITBA2 GGTCCCTGGCACCAGATG CAGGAAAGTAGCAACTGTGG 55 1.5mM UBL4 CCCTGGAGAAGGTGCTACTA CTCCCCATGCTCCGAGA 55 1.5mM P3 GGCTGGCCCCACTCCAC AACAGGCTGCTGTAGATGAAG 55 1.5mM FAM3A GCCGGAGATGTCAACGAC TGAGCCTCAGCCTCTGTC 55 1.5mM NEMO CTGGCCGAGAAGAAGGAG TCAACAGCTGAAGCGTAAGG 55 1.5mM GAB3 CTGAGCAGTGGTGCCCTTAC GTTCTGGCCAAGACCTTGAA 55 1.5mM DKC1 AAGTGACGAGACTCCTCCAG AGTTTCTCCTCCAGCTTCAA 55 1.5mM CLIC2 AAACACCCCACTTCTGGATG AGCCTTTTCTCCTGTAAGAGC 55 1.5mM VBP1 CCAGATTCTTGCTGGCAGAT TCAACCTGTAAGGATAAAGGGGTA 55 1.5mM ARNT TGAGCAACATGTTCAACAACC AGGGGAACAGCCAGAAGG 55 1.5mM RAB13 AGCATGGAATCCGATTTTTC CATCTACCTATGTGACCCTCCA 55 1.5mM NESCA ACCTCCCGAGATGCCTTC GTTCACTGGCCATTCCATTC 55 1.5mM FLNA Univ F GTTGTGGTGCCCTGAGTCTG 55 1.5mM FLNA 3'UTR 2R CAAGTGAAAGCCGAGAGGTC 55 1.5mM FLNA 3'UTR 3R CCTCCCAGAACCAAAGAAGA 55 1.5mM

Primers Used for Methylation-Specific PCR Annealing

Primer Name Forward Primer Sequence Reverse Primer Sequence Temp (°C) MgCl2 AR Unmod. Tet- TCCAGAATCTGTTCCAGAGCGTGC CTCTACGATGGGCTTGGGGAGAAC 65 2.5mM AR-M Methyl. 6 Fam- GCGAGCGTAGTATTTTTCGGC AACCAAATAACCTATAAAACCTCTACG 60 2.0mM AR-U Unmethyl. Hex- GTTGTGAGTGTAGTATTTTTTGGT CAAATAACCTATAAAACCTCTACA 58 2.0mM

147

Table 6.2: Refinement of the X;1 Translocation Breakpoint by FISH Mapping BAC clones were used as probes in FISH experiments in order to narrow the breakpoint region in the patient. BAC clone RP11-115M6 was found to span the translocation breakpoint.

148 Table 6.2:

Breakpoint Position BAC Probe Location (Mb) (Xq28) RP11-723E19 149.2 Proximal RP11-314B03 152.6 Proximal RP11-115M6 153.6 Spans Breakpoint RP11-810M4 154.2 Distal RP11-218L14 154.3 Distal

149

Table 6.3: RT-PCR Analysis RT-PCR analysis was used to determine gene expression near the breakpoint regions. Expression was determined from patient and SCH line cDNA in addition to controls. X- chromosome genes proximal to the breakpoint (including DKC1) were expressed from the normal X SCH line, while those distal were expressed from the der (1) SCH line. Several genes demonstrated expression from both the normal X, and der (X) SCH lines. The most notable is the FLNA gene which is thought to undergo XCI. This RT-PCR analysis suggests that the patient demonstrates disomic expression of the FLNA gene. Chromosome 1 genes proximal to the breakpoint were found to be expressed in the der (1) SCH line, while those distal were expressed from the der(X). While, there is a potential risk for monosomic expression of chromosome 1 genes due to the spread of XCI, it was not observed in this analysis.

150 Table 6.3 Coriell Location normal Coriell Ensembl (Mb) Cytogenetic Normal CHO male chm 1 Gene Name 1-11-07 Band Patient Der (1) Der (X) X DNA (GM01989) (GM13139) X Chromosome SLC9A6 134.9 Xq26.3 + 0 0 0 + FLNA 153.23 Xq28 + 0 + + 0 + EMD 153.26 Xq28 + 0 0 + 0 + RPL10 153.28 Xq28 + 0 0 0 + DNASE1L1 153.28 Xq28 + 0 +/- + 0 + TAZ 153.29 Xq28 + 0 0 0 + ATP6AP1 153.31 Xq28 + 0 0 0 + GDI1 153.32 Xq28 + 0 0 0 + FAM50A 153.33 Xq28 + 0 0 0 + PLXNA3 153.34 Xq28 + 0 0 0 + LAGE3 153.36 Xq28 + 0 0 0 + 151 UBL4 153.37 Xq28 + 0 0 0 + FAM3A 153.39 Xq28 + 0 0 0 + IKBKG 153.42 Xq28 + 0 + + 0 + GAB3 153.56 Xq28 + 0 0 + 0 + DKC1 (Breakpoint) 153.64 Xq28 + 0 0 + 0 + MPP1 153.66 Xq28 + + 0 0 + VBP1 154.1 Xq28 + + 0 0 + CLIC2 154.16 Xq28 + ? 0 0 + Chromosome 1 ARNT 149.05 1q21.2 + + 0 0 + RAB13 152.22 1q21.3 + + 0 0 + (Genes distal to bkpt) RUSC1 153.56 1q22 + 0 + 0 + LMNA 154.32 1q22 + 0 + 0 0 ++ NUF2 (CDCA1) 161.56 1q23.3 + 0 + 0 0 ++ SERPINC1 172.14 1q25.1 + 0 + 0 0 +/- + IRF6 208.02 1q32.2 + 0 + 0 +

Table 6.4 Combined RT-PCR and Array Results for Xq28 Genes The table represents the combined expression data for Xq28 genes generated from array expression and RT-PCR analyses. XCI data from Willard and Carrel153 is provided in the table as a reference to the inactivation status of a given gene. The XCI ratio represents the number of times biallelic expression was observed out of 9 independent SCHs. In the analysis of array expression data, the patient was compared to a reference level generated from each parent. Data from the array suggest that genes distal to the Xq28 breakpoint are expressed at higher levels in the patient in comparison to proximally located genes. This finding, in addition to RT-PCR data is suggestive of functional disomy for Xq28 genes distal to the breakpoint region with the presumption that the normal X is active. Notably, the expression array results do not corroborate RT-PCR results for the disomic expression of the FLNA gene in the patient.

152 Table 6.4

Location (in Mb) Patient Patient/ Patient/ [Ensembl release Total SCH SCH SCH Paternal Gene Maternal Gene Gene Symbol 45 June 2007] Undergoes XCI1 RNA Normal X der(X) der(1) Expression Expression FLNA 153.23 Yes +++0 0.89 0.86 EMD 153.26 Yes ++00 0.99 0.99 RPL10 153.28 Yes + 00 1.02 1.01 DNASE1L1 153.28 Yes +++/-0 1.08 1.09 TAZ 153.29 Yes + 00 1.00 1.03 GDI1 153.32 Yes + 00 0.96 1.00 UBL4 153.37 Yes + 00 1.19 1.19 IKBKG 153.42 9/9 Xi expressed +++0 1.02 1.02 GAB3 153.56 8/9 Xi expressed ++00 0.98 0.89 153 DKC1 153.64 Yes ++00 0.99 0.97 Genes Distal to Breakpoint MPP1 153.64 Yes + 0 + 1.67 1.67 F8 153.72 Yes 1.45 1.46 F8A1 153.77 Yes 1.59 1.67 FUNDC2 153.91 Yes 1.64 1.7 MTCP1 153.94 Yes 1.43 1.54 BRCC3 153.95 3/9 Xi expressed 1.96 1.85 VBP1 154.1 1/9 Xi expressed + 0 + 1.33 1.38 RAB39B 154.14 Yes 1.60 1.64 CLIC2 154.16 3/9 Xi expressed + 0? 2.67 1.54 TMLHE 154.37 Yes 2.18 2.09 1 XCI expresssion data from human-mouse somatic cell hybrids (SCH) [Carrel and Willard 2005] Xi = inactive X chromosome (from human-mouse SCH lines);

APPENDIX

Copyright Permission Granted from Applied Biosystems

Dear Ms. Cottrell:

Applera Corporation, through its Applied Biosystems Group (“Applied Biosystems”), is pleased to grant you limited, non-exclusive permission to reproduce the SNPlex Chemistry Overview figure from the AB SNPlex user guide displaying the SNPlex chemistry overview ("Image").

Applied Biosystems grants you such permission solely for the purpose of allowing you to include the Image in your dissertation titled "The Association of Antioxidant-Related Gene Polymorphisms and Second Primary Malignancy in a Pediatric Hodgkin’s Lymphoma Population."

Please include the following acknowledgement close to the Image: “Courtesy of Applied Biosystems”.

Finally, we thank you for your continued support of Applied Biosystems. Please feel free to call me if you have any questions.

Very truly yours, Chris Gigliotti Senior Trademark Administrator Applied Biosystems 850 Lincoln Centre Drive Foster City, CA 94404 Phone: 650-554-3590 Fax: 650-638-6677 [email protected]

154

REFERENCES

1. Ries LAG MD, Krapcho M, Mariotto A, Miller BA, Feuer EJ, Clegg L, Horner MJ, Howlander N, Eisner MP, Reichman M, Edwards BK (eds.). SEER Cancer Statistics Review, 1975-2004. National Cancer Institute. Bethesda, MD. 2007.

2. Horning SJ, Hoppe RT, Breslin S, Bartlett NL, Brown BW, Rosenberg SA. Stanford V and radiotherapy for locally extensive and advanced Hodgkin's disease: mature results of a prospective clinical trial. J Clin Oncol. Feb 1 2002;20(3):630-637.

3. Hunger SP, Link MP, Donaldson SS. ABVD/MOPP and low-dose involved-field radiotherapy in pediatric Hodgkin's disease: the Stanford experience. J Clin Oncol. Oct 1994;12(10):2160-2166.

4. Franke S, Wlodarska I, Maes B, Vandenberghe P, Delabie J, Hagemeijer A, De Wolf-Peeters C. Lymphocyte predominance Hodgkin disease is characterized by recurrent genomic imbalances. Blood. Mar 15 2001;97(6):1845-1853.

5. Weiss LM CJ, MacLennon K, and Warnke RA. Pathology of classical Hodgkin's Disease. Philadelphia: Lippencott Williams & Wilkins; 1999.

6. Marafioti T, Hummel M, Anagnostopoulos I, Foss HD, Falini B, Delsol G, Isaacson PG, Pileri S, Stein H. Origin of nodular lymphocyte-predominant Hodgkin's disease from a clonal expansion of highly mutated germinal-center B cells. N Engl J Med. Aug 14 1997;337(7):453-458.

7. Kanzler H, Kuppers R, Hansmann ML, Rajewsky K. Hodgkin and Reed- Sternberg cells in Hodgkin's disease represent the outgrowth of a dominant tumor clone derived from (crippled) germinal center B cells. J Exp Med. Oct 1 1996;184(4):1495-1505.

155 8. Kuppers R, Rajewsky K, Zhao M, Simons G, Laumann R, Fischer R, Hansmann ML. Hodgkin disease: Hodgkin and Reed-Sternberg cells picked from histological sections show clonal immunoglobulin gene rearrangements and appear to be derived from B cells at various stages of development. Proc Natl Acad Sci U S A. Nov 8 1994;91(23):10962-10966.

9. Cossman J, Messineo C, Bagg A. Reed-Sternberg cell: survival in a hostile sea. Lab Invest. Mar 1998;78(3):229-235.

10. Cartwright RA, Watkins G. Epidemiology of Hodgkin's disease: a review. Hematol Oncol. Mar 2004;22(1):11-26.

11. Jaffe ES, Banks PM, Nathwani B, Said J, Swerdlow SH. Recommendations for the reporting of lymphoid neoplasms: A report from the Association of Directors of Anatomic and Surgical Pathology. Mod Pathol. Jan 2004;17(1):131-135.

12. Schwartz CL. The management of Hodgkin disease in the young child. Curr Opin Pediatr. Feb 2003;15(1):10-16.

13. Kelly KM, Hutchinson RJ, Sposto R, Weiner MA, Lones MA, Perkins SL, Massey V. Feasibility of upfront dose-intensive chemotherapy in children with advanced-stage Hodgkin's lymphoma: preliminary results from the Children's Cancer Group Study CCG-59704. Ann Oncol. 2002;13 Suppl 1:107-111.

14. Nachman JB, Sposto R, Herzog P, Gilchrist GS, Wolden SL, Thomson J, Kadin ME, Pattengale P, Davis PC, Hutchinson RJ, White K. Randomized comparison of low-dose involved-field radiotherapy and no radiotherapy for children with Hodgkin's disease who achieve a complete response to chemotherapy. J Clin Oncol. Sep 15 2002;20(18):3765-3771.

15. Yung L, Linch D. Hodgkin's lymphoma. Lancet. Mar 15 2003;361(9361):943- 951.

16. Alexander FE, Daniel CP, Armstrong AA, Clark DA, Onions DE, Cartwright RA, Jarrett RF. Case clustering, Epstein-Barr virus Reed-Sternberg cell status and herpes virus serology in Hodgkin's disease: results of a case-control study. Eur J Cancer. 1995;31A(9):1479-1486.

156 17. Grasser FA, Murray PG, Kremmer E, Klein K, Remberger K, Feiden W, Reynolds G, Niedobitek G, Young LS, Mueller-Lantzsch N. Monoclonal antibodies directed against the Epstein-Barr virus-encoded nuclear antigen 1 (EBNA1): immunohistologic detection of EBNA1 in the malignant cells of Hodgkin's disease. Blood. Dec 1 1994;84(11):3792-3798.

18. Wang D, Liebowitz D, Kieff E. An EBV membrane protein expressed in immortalized lymphocytes transforms established rodent cells. Cell. Dec 1985;43(3 Pt 2):831-840.

19. Mack TM, Cozen W, Shibata DK, Weiss LM, Nathwani BN, Hernandez AM, Taylor CR, Hamilton AS, Deapen DM, Rappaport EB. Concordance for Hodgkin's disease in identical twins suggesting genetic susceptibility to the young-adult form of the disease. N Engl J Med. Feb 16 1995;332(7):413-418.

20. Kadan-Lottick NS, Kawashima T, Tomlinson G, Friedman DL, Yasui Y, Mertens AC, Robison LL, Strong LC. The risk of cancer in twins: a report from the childhood cancer survivor study. Pediatr Blood Cancer. Apr 2006;46(4):476-481.

21. Reed DM. On the pathological changes in Hodgkin's disease, with special reference to its relation to tuberculosis. Johns Hopkins Hosp Rep. 1902;10:133- 196.

22. Mauch PM, Hoppe RT, Armitage JO, Diehl V, Weiss LM. Hodgkin Lymphoma. New York City: Lippencott William & Wilkens; 2007.

23. Neglia JP, Friedman DL, Yasui Y, Mertens AC, Hammond S, Stovall M, Donaldson SS, Meadows AT, Robison LL. Second malignant neoplasms in five- year survivors of childhood cancer: childhood cancer survivor study. J Natl Cancer Inst. Apr 18 2001;93(8):618-629.

24. Mertens AC, Yasui Y, Neglia JP, Potter JD, Nesbit ME, Jr., Ruccione K, Smithson WA, Robison LL. Late mortality experience in five-year survivors of childhood and adolescent cancer: the Childhood Cancer Survivor Study. J Clin Oncol. Jul 1 2001;19(13):3163-3172.

25. Metayer C, Lynch CF, Clarke EA, Glimelius B, Storm H, Pukkala E, Joensuu T, van Leeuwen FE, van't Veer MB, Curtis RE, Holowaty EJ, Andersson M, Wiklund T, Gospodarowicz M, Travis LB. Second cancers among long-term

157 survivors of Hodgkin's disease diagnosed in childhood and adolescence. J Clin Oncol. Jun 2000;18(12):2435-2443.

26. van Leeuwen FE, Klokman WJ, Veer MB, Hagenbeek A, Krol AD, Vetter UA, Schaapveld M, van Heerde P, Burgers JM, Somers R, Aleman BM. Long-term risk of second malignancy in survivors of Hodgkin's disease treated during adolescence or young adulthood. J Clin Oncol. Feb 2000;18(3):487-497.

27. Bhatia S, Robison LL, Oberlin O, Greenberg M, Bunin G, Fossati-Bellani F, Meadows AT. Breast cancer and other second neoplasms after childhood Hodgkin's disease. N Engl J Med. Mar 21 1996;334(12):745-751.

28. Green DM, Hyland A, Barcos MP, Reynolds JA, Lee RJ, Hall BC, Zevon MA. Second malignant neoplasms after treatment for Hodgkin's disease in childhood or adolescence. J Clin Oncol. Apr 2000;18(7):1492-1499.

29. Wolden SL, Lamborn KR, Cleary SF, Tate DJ, Donaldson SS. Second cancers following pediatric Hodgkin's disease. J Clin Oncol. Feb 1998;16(2):536-544.

30. Swerdlow AJ, Barber JA, Hudson GV, Cunningham D, Gupta RK, Hancock BW, Horwich A, Lister TA, Linch DC. Risk of second malignancy after Hodgkin's disease in a collaborative British cohort: the relation to age at treatment. J Clin Oncol. Feb 2000;18(3):498-509.

31. Sankila R, Garwicz S, Olsen JH, Dollner H, Hertz H, Kreuger A, Langmark F, Lanning M, Moller T, Tulinius H. Risk of subsequent malignant neoplasms among 1,641 Hodgkin's disease patients diagnosed in childhood and adolescence: a population-based cohort study in the five Nordic countries. Association of the Nordic Cancer Registries and the Nordic Society of Pediatric Hematology and Oncology. J Clin Oncol. May 1996;14(5):1442-1446.

32. Foss Abrahamsen A, Andersen A, Nome O, Jacobsen AB, Holte H, Foss Abrahamsen J, Kvaloy S. Long-term risk of second malignancy after treatment of Hodgkin's disease: the influence of treatment, age, and follow-up time. Ann Oncol. 2002;13:1786-1791.

33. Thomson AB, Wallace WH. Treatment of paediatric Hodgkin's disease. a balance of risks. Eur J Cancer. Mar 2002;38(4):468-477.

158 34. Haukvik UK, Dieset I, Bjoro T, Holte H, Fossa SD. Treatment-related premature ovarian failure as a long-term complication after Hodgkin's lymphoma. Ann Oncol. Sep 2006;17(9):1428-1433.

35. Clark ST, Radford JA, Crowther D, Swindell R, Shalet SM. Gonadal function following chemotherapy for Hodgkin's disease: a comparative study of MVPP and a seven-drug hybrid regimen. J Clin Oncol. Jan 1995;13(1):134-139.

36. Kelly KM, Perentesis JP. Polymorphisms of drug metabolizing enzymes and markers of genotoxicity to identify patients with Hodgkin's lymphoma at risk of treatment-related complications. Ann Oncol. 2002;13 Suppl 1:34-39.

37. Schimpff SC, Diggs CH, Wiswell JG, Salvatore PC, Wiernik PH. Radiation- related thyroid dysfunction: implications for the treatment of Hodgkin's disease. Ann Intern Med. Jan 1980;92(1):91-98.

38. Shalet SM, Gibson B, Swindell R, Pearson D. Effect of spinal irradiation on growth. Arch Dis Child. May 1987;62(5):461-464.

39. Kenney LB, Yasui Y, Inskip PD, Hammond S, Neglia JP, Mertens AC, Meadows AT, Friedman D, Robison LL, Diller L. Breast cancer after childhood cancer: a report from the Childhood Cancer Survivor Study. Ann Intern Med. Oct 19 2004;141(8):590-597.

40. van Leeuwen FE, Klokman WJ, Stovall M, Dahler EC, van't Veer MB, Noordijk EM, Crommelin MA, Aleman BM, Broeks A, Gospodarowicz M, Travis LB, Russell NS. Roles of radiation dose, chemotherapy, and hormonal factors in breast cancer following Hodgkin's disease. J Natl Cancer Inst. Jul 2 2003;95(13):971-980.

41. Hodgson DC, Hudson MM, Constine LS. Pediatric hodgkin lymphoma: maximizing efficacy and minimizing toxicity. Semin Radiat Oncol. Jul 2007;17(3):230-242.

42. Robison LL, Mertens AC, Boice JD, Breslow NE, Donaldson SS, Green DM, Li FP, Meadows AT, Mulvihill JJ, Neglia JP, Nesbit ME, Packer RJ, Potter JD, Sklar CA, Smith MA, Stovall M, Strong LC, Yasui Y, Zeltzer LK. Study design and cohort characteristics of the Childhood Cancer Survivor Study: a multi- institutional collaborative project. Med Pediatr Oncol. Apr 2002;38(4):229-239.

159 43. Nichols KE, Levitz S, Shannon KE, Wahrer DC, Bell DW, Chang G, Hegde S, Neuberg D, Shafman T, Tarbell NJ, Mauch P, Ishioka C, Haber DA, Diller L. Heterozygous germline ATM mutations do not contribute to radiation-associated malignancies after Hodgkin's disease. J Clin Oncol. Apr 1999;17(4):1259.

44. Mertens AC, Mitby PA, Radloff G, Jones IM, Perentesis J, Kiffmeyer WR, Neglia JP, Meadows A, Potter JD, Friedman D, Yasui Y, Robison LL, Davies SM. XRCC1 and glutathione-S-transferase gene polymorphisms and susceptibility to radiotherapy-related malignancies in survivors of Hodgkin disease. Cancer. Sep 15 2004;101(6):1463-1472.

45. Seedhouse C, Russell N. Advances in the understanding of susceptibility to treatment-related acute myeloid leukaemia. Br J Haematol. Jun 2007;137(6):513- 529.

46. Thompson LH, West MG. XRCC1 keeps DNA from getting stranded. Mutat Res. Feb 16 2000;459(1):1-18.

47. Seedhouse C, Bainton R, Lewis M, Harding A, Russell N, Das-Gupta E. The genotype distribution of the XRCC1 gene indicates a role for base excision repair in the development of therapy-related acute myeloblastic leukemia. Blood. Nov 15 2002;100(10):3761-3766.

48. Lunn RM, Langlois RG, Hsieh LL, Thompson CL, Bell DA. XRCC1 polymorphisms: effects on aflatoxin B1-DNA adducts and glycophorin A variant frequency. Cancer Res. Jun 1 1999;59(11):2557-2561.

49. Nelson HH, Kelsey KT, Mott LA, Karagas MR. The XRCC1 Arg399Gln polymorphism, sunburn, and non-melanoma skin cancer: evidence of gene- environment interaction. Cancer Res. Jan 1 2002;62(1):152-155.

50. Hu JJ, Smith TR, Miller MS, Mohrenweiser HW, Golden A, Case LD. Amino acid substitution variants of APE1 and XRCC1 genes associated with ionizing radiation sensitivity. Carcinogenesis. Jun 2001;22(6):917-922.

51. Pemble S, Schroeder KR, Spencer SR, Meyer DJ, Hallier E, Bolt HM, Ketterer B, Taylor JB. Human glutathione S-transferase theta (GSTT1): cDNA cloning and the characterization of a genetic polymorphism. Biochem J. May 15 1994;300 (Pt 1):271-276.

160 52. Seidegard J, Vorachek WR, Pero RW, Pearson WR. Hereditary differences in the expression of the human glutathione transferase active on trans-stilbene oxide are due to a gene deletion. Proc Natl Acad Sci U S A. Oct 1988;85(19):7293-7297.

53. Jawad M, Seedhouse CH, Russell N, Plumb M. Polymorphisms in human homeobox HLX1 and DNA repair RAD51 genes increase the risk of therapy- related acute myeloid leukemia. Blood. Dec 1 2006;108(12):3916-3918.

54. Bolufer P, Collado M, Barragan E, Calasanz MJ, Colomer D, Tormo M, Gonzalez M, Brunet S, Batlle M, Cervera J, Sanz MA. Profile of polymorphisms of drug- metabolising enzymes and the risk of therapy-related leukaemia. Br J Haematol. Feb 2007;136(4):590-596.

55. Larson RA, Wang Y, Banerjee M, Wiemels J, Hartford C, Le Beau MM, Smith MT. Prevalence of the inactivating 609C-->T polymorphism in the NAD(P)H:quinone oxidoreductase (NQO1) gene in patients with primary and therapy-related myeloid leukemia. Blood. Jul 15 1999;94(2):803-807.

56. Felix CA, Walker AH, Lange BJ, Williams TM, Winick NJ, Cheung NK, Lovett BD, Nowell PC, Blair IA, Rebbeck TR. Association of CYP3A4 genotype with treatment-related leukemia. Proc Natl Acad Sci U S A. Oct 27 1998;95(22):13176-13181.

57. Blanco JG, Edick MJ, Hancock ML, Winick NJ, Dervieux T, Amylon MD, Bash RO, Behm FG, Camitta BM, Pui CH, Raimondi SC, Relling MV. Genetic polymorphisms in CYP3A5, CYP3A4 and NQO1 in children who developed therapy-related myeloid malignancies. Pharmacogenetics. Nov 2002;12(8):605- 611.

58. Jazbec J, Kitanovski L, Aplenc R, Debeljak M, Dolzan V. No evidence of association of methylenetetrahydrofolate reductase polymorphism with occurrence of second neoplasms after treatment of childhood leukemia. Leuk Lymphoma. Jun 2005;46(6):893-897.

59. Brewster AM, Alberg AJ, Strickland PT, Hoffman SC, Helzlsouer K. XPD polymorphism and risk of subsequent cancer in individuals with nonmelanoma skin cancer. Cancer Epidemiol Biomarkers Prev. Aug 2004;13(8):1271-1275.

161 60. Wiseman H, Halliwell B. Damage to DNA by reactive oxygen and nitrogen species: role in inflammatory disease and progression to cancer. Biochem J. Jan 1 1996;313 (Pt 1):17-29.

61. Valko M, Rhodes CJ, Moncol J, Izakovic M, Mazur M. Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chem Biol Interact. Mar 10 2006;160(1):1-40.

62. Richter C, Gogvadze V, Laffranchi R, Schlapbach R, Schweizer M, Suter M, Walter P, Yaffee M. Oxidants in mitochondria: from physiology to diseases. Biochim Biophys Acta. May 24 1995;1271(1):67-74.

63. Loschen G, Flohe L, Chance B. Respiratory chain linked H(2)O(2) production in pigeon heart mitochondria. FEBS Lett. Nov 1 1971;18(2):261-264.

64. Hug H, Strand S, Grambihler A, Galle J, Hack V, Stremmel W, Krammer PH, Galle PR. Reactive oxygen intermediates are involved in the induction of CD95 ligand mRNA expression by cytostatic drugs in hepatoma cells. J Biol Chem. Nov 7 1997;272(45):28191-28193.

65. Pelicano H, Carney D, Huang P. ROS stress in cancer cells and therapeutic implications. Drug Resist Updat. Apr 2004;7(2):97-110.

66. Serrano J, Palmeira CM, Kuehl DW, Wallace KB. Cardioselective and cumulative oxidation of mitochondrial DNA following subchronic doxorubicin administration. Biochim Biophys Acta. Apr 21 1999;1411(1):201-205.

67. Tsang WP, Chau SP, Kong SK, Fung KP, Kwok TT. Reactive oxygen species mediate doxorubicin induced p53-independent apoptosis. Life Sci. Sep 5 2003;73(16):2047-2058.

68. Juchau MR, Fantel AG, Harris C, Beyer BK. The potential role of redox cycling as a mechanism for chemical teratogenesis. Environ Health Perspect. Dec 1986;70:131-136.

69. Ravi D, Das KC. Redox-cycling of anthracyclines by thioredoxin system: increased superoxide generation and DNA damage. Cancer Chemother Pharmacol. Nov 2004;54(5):449-458.

162 70. Halliwell B. Oxidative stress and cancer: have we moved forward? Biochem J. Jan 1 2007;401(1):1-11.

71. Mates JM, Perez-Gomez C, Nunez de Castro I. Antioxidant enzymes and human diseases. Clin Biochem. Nov 1999;32(8):595-603.

72. Toyokuni S, Okamoto K, Yodoi J, Hiai H. Persistent oxidative stress in cancer. FEBS Lett. Jan 16 1995;358(1):1-3.

73. Okamoto K, Toyokuni S, Uchida K, Ogawa O, Takenewa J, Kakehi Y, Kinoshita H, Hattori-Nakakuki Y, Hiai H, Yoshida O. Formation of 8-hydroxy-2'- deoxyguanosine and 4-hydroxy-2-nonenal-modified proteins in human renal-cell carcinoma. Int J Cancer. Sep 15 1994;58(6):825-829.

74. Jaruga P, Zastawny TH, Skokowski J, Dizdaroglu M, Olinski R. Oxidative DNA base damage and antioxidant enzyme activities in human lung cancer. FEBS Lett. Mar 14 1994;341(1):59-64.

75. Enstrom JE, Kanim LE, Klein MA. Vitamin C intake and mortality among a sample of the United States population. Epidemiology. May 1992;3(3):194-202.

76. Kelm M. Nitric oxide metabolism and breakdown. Biochim Biophys Acta. May 5 1999;1411(2-3):273-289.

77. Alderton WK, Cooper CE, Knowles RG. Nitric oxide synthases: structure, function and inhibition. Biochem J. Aug 1 2001;357(Pt 3):593-615.

78. Bergendi L, Benes L, Durackova Z, Ferencik M. Chemistry, physiology and pathology of free radicals. Life Sci. 1999;65(18-19):1865-1874.

79. Carr AC, McCall MR, Frei B. Oxidation of LDL by myeloperoxidase and reactive nitrogen species: reaction pathways and antioxidant protection. Arterioscler Thromb Vasc Biol. Jul 2000;20(7):1716-1723.

80. Klatt P, Lamas S. Regulation of protein function by S-glutathiolation in response to oxidative and nitrosative stress. Eur J Biochem. Aug 2000;267(16):4928-4944.

163 81. Fridovich I. Superoxide dismutases. Adv Enzymol Relat Areas Mol Biol. 1986;58:61-97.

82. Ho YS, Crapo JD. Isolation and characterization of complementary encoding human manganese-containing superoxide dismutase. FEBS Lett. Mar 14 1988;229(2):256-260.

83. Hjalmarsson K, Marklund SL, Engstrom A, Edlund T. Isolation and sequence of complementary DNA encoding human extracellular superoxide dismutase. Proc Natl Acad Sci U S A. Sep 1987;84(18):6340-6344.

84. Chance B, Sies H, Boveris A. Hydroperoxide metabolism in mammalian organs. Physiol Rev. Jul 1979;59(3):527-605.

85. Guidot DM, McCord JM, Wright RM, Repine JE. Absence of electron transport (Rho 0 state) restores growth of a manganese-superoxide dismutase-deficient Saccharomyces cerevisiae in . Evidence for electron transport as a major source of superoxide generation in vivo. J Biol Chem. Dec 15 1993;268(35):26699-26703.

86. Brown KM, Arthur JR. Selenium, selenoproteins and human health: a review. Public Health Nutr. Apr 2001;4(2B):593-599.

87. Villette S, Kyle JA, Brown KM, Pickard K, Milne JS, Nicol F, Arthur JR, Hesketh JE. A novel single nucleotide polymorphism in the 3' untranslated region of human glutathione peroxidase 4 influences lipoxygenase metabolism. Blood Cells Mol Dis. Sep-Oct 2002;29(2):174-178.

88. Gromer S, Eubel JK, Lee BL, Jacob J. Human selenoproteins at a glance. Cell Mol Life Sci. Nov 2005;62(21):2414-2437.

89. Wu G, Fang YZ, Yang S, Lupton JR, Turner ND. Glutathione metabolism and its implications for health. J Nutr. Mar 2004;134(3):489-492.

90. Stryer L. Biochemistry. (3rd ed). New York: W.H. Freeman and Company; 1988.

91. Townsend DM, Tew KD, Tapiero H. The importance of glutathione in human disease. Biomed Pharmacother. May-Jun 2003;57(3-4):145-155. 164 92. Brigelius-Flohe R. Tissue-specific functions of individual glutathione peroxidases. Free Radic Biol Med. Nov 1999;27(9-10):951-965.

93. Chu FF, Doroshow JH, Esworthy RS. Expression, characterization, and tissue distribution of a new cellular selenium-dependent glutathione peroxidase, GSHPx-GI. J Biol Chem. Feb 5 1993;268(4):2571-2576.

94. Yoshimura S, Watanabe K, Suemizu H, Onozawa T, Mizoguchi J, Tsuda K, Hatta H, Moriuchi T. Tissue specific expression of the plasma glutathione peroxidase gene in rat kidney. J Biochem (Tokyo). Jun 1991;109(6):918-923.

95. Forsberg L, de Faire U, Morgenstern R. Low yield of polymorphisms from EST blast searching: analysis of genes related to oxidative stress and verification of the P197L polymorphism in GPX1. Hum Mutat. 1999;13(4):294-300.

96. Maiorino M, Thomas JP, Girotti AW, Ursini F. Reactivity of phospholipid hydroperoxide glutathione peroxidase with membrane and lipoprotein lipid hydroperoxides. Free Radic Res Commun. 1991;12-13 Pt 1:131-135.

97. Thomas JP, Maiorino M, Ursini F, Girotti AW. Protective action of phospholipid hydroperoxide glutathione peroxidase against membrane-damaging lipid peroxidation. In situ reduction of phospholipid and cholesterol hydroperoxides. J Biol Chem. Jan 5 1990;265(1):454-461.

98. Ursini F, Maiorino M, Gregolin C. The selenoenzyme phospholipid hydroperoxide glutathione peroxidase. Biochim Biophys Acta. Mar 29 1985;839(1):62-70.

99. Arai M, Imai H, Sumi D, Imanaka T, Takano T, Chiba N, Nakagawa Y. Import into mitochondria of phospholipid hydroperoxide glutathione peroxidase requires a leader sequence. Biochem Biophys Res Commun. Oct 14 1996;227(2):433-439.

100. Pushpa-Rekha TR, Burdsall AL, Oleksa LM, Chisolm GM, Driscoll DM. Rat phospholipid-hydroperoxide glutathione peroxidase. cDNA cloning and identification of multiple transcription and translation start sites. J Biol Chem. Nov 10 1995;270(45):26993-26999.

101. Arai M, Imai H, Koumura T, Yoshida M, Emoto K, Umeda M, Chiba N, Nakagawa Y. Mitochondrial phospholipid hydroperoxide glutathione peroxidase 165 plays a major role in preventing oxidative injury to cells. J Biol Chem. Feb 19 1999;274(8):4924-4933.

102. Armitage P, Doll R. The age distribution of cancer and a multi-stage theory of carcinogenesis. 1954. Int J Epidemiol. Dec 2004;33(6):1174-1179.

103. Armitage P. Multistage models of carcinogenesis. Environ Health Perspect. Nov 1985;63:195-201.

104. Xie J, Fan R, Meng Z. Protein oxidation and DNA-protein crosslink induced by sulfur dioxide in lungs, livers, and hearts from mice. Inhal Toxicol. Jul 2007;19(9):759-765.

105. Malins DC, Haimanot R. Major alterations in the nucleotide structure of DNA in cancer of the female breast. Cancer Res. Oct 1 1991;51(19):5430-5432.

106. Loft S, Poulsen HE. Cancer risk and oxidative DNA damage in man. J Mol Med. Jun 1996;74(6):297-312.

107. Davies KJ. The broad spectrum of responses to oxidants in proliferating cells: a new paradigm for oxidative stress. IUBMB Life. Jul 1999;48(1):41-47.

108. Thews O, Lambert C, Kelleher DK, Biesalski HK, Vaupel P, Frank J. Impact of therapeutically induced reactive oxygen species and radical scavenging by alpha- tocopherol on tumor cell adhesion. Oncol Rep. Oct 2007;18(4):965-971.

109. Kim KH, Cho YS, Park JM, Yoon SO, Kim KW, Chung AS. Pro-MMP-2 activation by the PPARgamma agonist, ciglitazone, induces cell invasion through the generation of ROS and the activation of ERK. FEBS Lett. Jul 10 2007;581(17):3303-3310.

110. Ushio-Fukai M. VEGF signaling through NADPH oxidase-derived ROS. Antioxid Redox Signal. Jun 2007;9(6):731-739.

111. Huang SS, Zheng RL. Biphasic regulation of angiogenesis by reactive oxygen species. Pharmazie. Mar 2006;61(3):223-229.

166 112. Oberley LW, Buettner GR. Role of superoxide dismutase in cancer: a review. Cancer Res. Apr 1979;39(4):1141-1149.

113. Sun Y. Free radicals, antioxidant enzymes, and carcinogenesis. Free Radic Biol Med. 1990;8(6):583-599.

114. Oberley TD, Oberley LW. Antioxidant enzyme levels in cancer. Histol Histopathol. Apr 1997;12(2):525-535.

115. Elchuri S, Oberley TD, Qi W, Eisenstein RS, Jackson Roberts L, Van Remmen H, Epstein CJ, Huang TT. CuZnSOD deficiency leads to persistent and widespread oxidative damage and hepatocarcinogenesis later in life. Oncogene. Jan 13 2005;24(3):367-380.

116. Li Y, Huang TT, Carlson EJ, Melov S, Ursell PC, Olson JL, Noble LJ, Yoshimura MP, Berger C, Chan PH, Wallace DC, Epstein CJ. Dilated cardiomyopathy and neonatal lethality in mutant mice lacking manganese superoxide dismutase. Nat Genet. Dec 1995;11(4):376-381.

117. Van Remmen H, Ikeno Y, Hamilton M, Pahlavani M, Wolf N, Thorpe SR, Alderson NL, Baynes JW, Epstein CJ, Huang TT, Nelson J, Strong R, Richardson A. Life-long reduction in MnSOD activity results in increased DNA damage and higher incidence of cancer but does not accelerate aging. Physiol Genomics. Dec 16 2003;16(1):29-37.

118. Sato K, Ito K, Kohara H, Yamaguchi Y, Adachi K, Endo H. Negative regulation of catalase gene expression in hepatoma cells. Mol Cell Biol. Jun 1992;12(6):2525-2533.

119. Ishii K, Zhen LX, Wang DH, Funamori Y, Ogawa K, Taketa K. Prevention of mammary tumorigenesis in acatalasemic mice by vitamin E supplementation. Jpn J Cancer Res. Jul 1996;87(7):680-684.

120. Chu FF, Esworthy RS, Chu PG, Longmate JA, Huycke MM, Wilczynski S, Doroshow JH. Bacteria-induced intestinal cancer in mice with disrupted Gpx1 and Gpx2 genes. Cancer Res. Feb 1 2004;64(3):962-968.

121. Society AC. Cancer Facts and Figures 2007. American Cancer Society. Available from: 167 http://www.cancer.org/docroot/STT/content/STT_1x_Cancer_Facts__Figures_20 07.asp. accessed September 19, 2007, 2007.

122. Robison LL, Green DM, Hudson M, Meadows AT, Mertens AC, Packer RJ, Sklar CA, Strong LC, Yasui Y, Zeltzer LK. Long-term outcomes of adult survivors of childhood cancer. Cancer. Dec 1 2005;104(11 Suppl):2557-2564.

123. Davies SM. Subsequent malignant neoplasms in survivors of childhood cancer: Childhood Cancer Survivor Study (CCSS) studies. Pediatr Blood Cancer. Jun 15 2007;48(7):727-730.

124. Lum A, Le Marchand L. A simple mouthwash method for obtaining genomic DNA in molecular epidemiological studies. Cancer Epidemiol Biomarkers Prev. Aug 1998;7(8):719-724.

125. Barrett JC, Fry B, Maller J, Daly MJ. Haploview: analysis and visualization of LD and haplotype maps. Bioinformatics. Jan 15 2005;21(2):263-265.

126. Dudbridge F, Gusnanto A, Koeleman BP. Detecting multiple associations in genome-wide studies. Hum Genomics. Mar 2006;2(5):310-317.

127. Crichton N. Cox proportional model. J Clin Nurs. Nov 2002;11(6):723.

128. Hsu FC, Liang KY, Beaty TH. Multipoint linkage disequilibrium mapping approach: incorporating evidence of linkage and linkage disequilibrium from unlinked region. Genet Epidemiol. Jul 2003;25(1):1-13.

129. Primer on type I and type II errors. Eff Clin Pract. Nov-Dec 2001;4(6):284-285.

130. Benjamini Y, Drai D, Elmer G, Kafkafi N, Golani I. Controlling the false discovery rate in behavior genetics research. Behav Brain Res. Nov 1 2001;125(1- 2):279-284.

131. Storey JD, Tibshirani R. Statistical significance for genomewide studies. Proc Natl Acad Sci U S A. Aug 5 2003;100(16):9440-9445.

168 132. Perneger TV. What's wrong with Bonferroni adjustments. Bmj. Apr 18 1998;316(7139):1236-1238.

133. Esposito LA, Kokoszka JE, Waymire KG, Cottrell B, MacGregor GR, Wallace DC. Mitochondrial oxidative stress in mice lacking the glutathione peroxidase-1 gene. Free Radic Biol Med. Mar 1 2000;28(5):754-766.

134. Yant LJ, Ran Q, Rao L, Van Remmen H, Shibatani T, Belter JG, Motta L, Richardson A, Prolla TA. The selenoprotein GPX4 is essential for mouse development and protects from radiation and oxidative damage insults. Free Radic Biol Med. Feb 15 2003;34(4):496-502.

135. Knopp EA, Arndt TL, Eng KL, Caldwell M, LeBoeuf RC, Deeb SS, O'Brien KD. Murine phospholipid hydroperoxide glutathione peroxidase: cDNA sequence, tissue expression, and mapping. Mamm Genome. Jun 1999;10(6):601-605.

136. Brigelius-Flohe R, Maurer S, Lotzer K, Bol G, Kallionpaa H, Lehtolainen P, Viita H, Yla-Herttuala S. Overexpression of PHGPx inhibits hydroperoxide-induced oxidation, NFkappaB activation and apoptosis and affects oxLDL-mediated proliferation of rabbit aortic smooth muscle cells. Atherosclerosis. Oct 2000;152(2):307-316.

137. Hurst R, Korytowski W, Kriska T, Esworthy RS, Chu FF, Girotti AW. Hyperresistance to cholesterol hydroperoxide-induced peroxidative injury and apoptotic death in a tumor cell line that overexpresses glutathione peroxidase isotype-4. Free Radic Biol Med. Nov 1 2001;31(9):1051-1065.

138. Nomura K, Imai H, Koumura T, Kobayashi T, Nakagawa Y. Mitochondrial phospholipid hydroperoxide glutathione peroxidase inhibits the release of cytochrome c from mitochondria by suppressing the peroxidation of cardiolipin in hypoglycaemia-induced apoptosis. Biochem J. Oct 1 2000;351(Pt 1):183-193.

139. Lefevre G, Beljean-Leymarie M, Beyerle F, Bonnefont-Rousselot D, Cristol JP, Therond P, Torreilles J. [Evaluation of lipid peroxidation by measuring thiobarbituric acid reactive substances]. Ann Biol Clin (Paris). May-Jun 1998;56(3):305-319.

140. Lyon MF. Gene action in the X-chromosome of the mouse (Mus musculus L.). Nature. Apr 22 1961;190:372-373.

169 141. Lyon MF. Sex chromatin and gene action in the mammalian X-chromosome. Am J Hum Genet. Jun 1962;14:135-148.

142. Monteiro J, Derom C, Vlietinck R, Kohn N, Lesser M, Gregersen PK. Commitment to X inactivation precedes the twinning event in monochorionic MZ twins. Am J Hum Genet. Aug 1998;63(2):339-346.

143. Puck JM, Stewart CC, Nussbaum RL. Maximum-likelihood analysis of human T- cell X chromosome inactivation patterns: normal women versus carriers of X- linked severe combined immunodeficiency. Am J Hum Genet. Apr 1992;50(4):742-748.

144. Amos-Landgraf JM, Cottle A, Plenge RM, Friez M, Schwartz CE, Longshore J, Willard HF. X chromosome-inactivation patterns of 1,005 phenotypically unaffected females. Am J Hum Genet. Sep 2006;79(3):493-499.

145. Brown CJ, Robinson WP. The causes and consequences of random and non- random X chromosome inactivation in humans. Clin Genet. Nov 2000;58(5):353- 363.

146. Migeon BR. Non-random X chromosome inactivation in mammalian cells. Cytogenet Cell Genet. 1998;80(1-4):142-148.

147. Brown CJ, Lafreniere RG, Powers VE, Sebastio G, Ballabio A, Pettigrew AL, Ledbetter DH, Levy E, Craig IW, Willard HF. Localization of the X inactivation centre on the human X chromosome in Xq13. Nature. Jan 3 1991;349(6304):82- 84.

148. Brown CJ, Ballabio A, Rupert JL, Lafreniere RG, Grompe M, Tonlorenzi R, Willard HF. A gene from the region of the human X inactivation centre is expressed exclusively from the inactive X chromosome. Nature. Jan 3 1991;349(6304):38-44.

149. Russell LB. Mammalian X-chromosome action: inactivation limited in spread and region of origin. Science. May 31 1963;140:976-978.

150. Keitges EA, Palmer CG. Analysis of spreading of inactivation in eight X autosome translocations utilizing the high resolution RBG technique. Hum Genet. Mar 1986;72(3):231-236. 170 151. White WM, Willard HF, Van Dyke DL, Wolff DJ. The spreading of X inactivation into autosomal material of an x;autosome translocation: evidence for a difference between autosomal and X-chromosomal DNA. Am J Hum Genet. Jul 1998;63(1):20-28.

152. Carrel L, Willard HF. Heterogeneous gene expression from the inactive X chromosome: an X-linked gene that escapes X inactivation in some human cell lines but is inactivated in others. Proc Natl Acad Sci U S A. Jun 22 1999;96(13):7364-7369.

153. Carrel L, Willard HF. X-inactivation profile reveals extensive variability in X- linked gene expression in females. Nature. Mar 17 2005;434(7031):400-404.

154. Chow JC, Yen Z, Ziesche SM, Brown CJ. Silencing of the mammalian X chromosome. Annu Rev Genomics Hum Genet. 2005;6:69-92.

155. Disteche CM, Filippova GN, Tsuchiya KD. Escape from X inactivation. Cytogenet Genome Res. 2002;99(1-4):36-43.

156. Salido EC, Yen PH, Koprivnikar K, Yu LC, Shapiro LJ. The human enamel protein gene amelogenin is expressed from both the X and the Y chromosomes. Am J Hum Genet. Feb 1992;50(2):303-316.

157. Watanabe M, Zinn AR, Page DC, Nishimoto T. Functional equivalence of human X- and Y-encoded isoforms of ribosomal protein S4 consistent with a role in Turner syndrome. Nat Genet. Jul 1993;4(3):268-271.

158. Craig IW, Mill J, Craig GM, Loat C, Schalkwyk LC. Application of microarrays to the analysis of the inactivation status of human X-linked genes expressed in lymphocytes. Eur J Hum Genet. Aug 2004;12(8):639-646.

159. Sudbrak R, Wieczorek G, Nuber UA, Mann W, Kirchner R, Erdogan F, Brown CJ, Wohrle D, Sterk P, Kalscheuer VM, Berger W, Lehrach H, Ropers HH. X chromosome-specific cDNA arrays: identification of genes that escape from X- inactivation and other applications. Hum Mol Genet. Jan 1 2001;10(1):77-83.

160. Talebizadeh Z, Simon SD, Butler MG. X chromosome gene expression in human tissues: male and female comparisons. Genomics. Dec 2006;88(6):675-681.

171 161. Gersen SL, Keagle MB, eds. The Principles of Clinical Cytogenetics. (Second ed). Totowa: Humana Press; 2005.

162. Gardner RJ, Sutherland GR. Chromosome Abnormalities and Genetic Counseling. (Third ed). New York: Oxford University Press; 2004.

163. Waters JJ, Campbell PL, Crocker AJ, Campbell CM. Phenotypic effects of balanced X-autosome translocations in females: a retrospective survey of 104 cases reported from UK laboratories. Hum Genet. Apr 2001;108(4):318-327.

164. Sarto GE, Therman E, Patau K. X inactivation in man: a woman with t(Xq-- ;12q+). Am J Hum Genet. May 1973;25(3):262-270.

165. Madan K. Balanced structural changes involving the human X: effect on sexual phenotype. Hum Genet. 1983;63(3):216-221.

166. Therman E, Laxova R, Susman B. The critical region on the human Xq. Hum Genet. Oct 1990;85(5):455-461.

167. Ray PN, Belfall B, Duff C, Logan C, Kean V, Thompson MW, Sylvester JE, Gorski JL, Schmickel RD, Worton RG. Cloning of the breakpoint of an X;21 translocation associated with Duchenne muscular dystrophy. Nature. Dec 19-1986 Jan 1 1985;318(6047):672-675.

168. Cockburn DJ, Munro EA, Craig IW, Boyd Y. Mapping of X chromosome translocation breakpoints in females with Duchenne muscular dystrophy with respect to exons of the dystrophin gene. Hum Genet. Dec 1992;90(4):407-412.

169. Boyd Y, Munro E, Ray P, Worton R, Monaco T, Kunkel L, Craig I. Molecular heterogeneity of translocations associated with muscular dystrophy. Clin Genet. Apr 1987;31(4):265-272.

170. Matsumoto N, Pilz DT, Fantes JA, Kittikamron K, Ledbetter DH. Isolation of BAC clones spanning the Xq22.3 translocation breakpoint in a lissencephaly patient with a de novo X;2 translocation. J Med Genet. Oct 1998;35(10):829-832.

171. Zemni R, Bienvenu T, Vinet MC, Sefiani A, Carrie A, Billuart P, McDonell N, Couvert P, Francis F, Chafey P, Fauchereau F, Friocourt G, des Portes V, 172 Cardona A, Frints S, Meindl A, Brandau O, Ronce N, Moraine C, van Bokhoven H, Ropers HH, Sudbrak R, Kahn A, Fryns JP, Beldjord C, Chelly J. A new gene involved in X-linked mental retardation identified by analysis of an X;2 balanced translocation. Nat Genet. Feb 2000;24(2):167-170.

172. Bedell MA, Jenkins NA, Copeland NG. Good genes in bad neighbourhoods. Nat Genet. Mar 1996;12(3):229-232.

173. Kleinjan DA, van Heyningen V. Long-range control of gene expression: emerging mechanisms and disruption in disease. Am J Hum Genet. Jan 2005;76(1):8-32.

174. Kleinjan DJ, van Heyningen V. Position effect in human genetic disease. Hum Mol Genet. 1998;7(10):1611-1618.

175. Fantes J, Redeker B, Breen M, Boyle S, Brown J, Fletcher J, Jones S, Bickmore W, Fukushima Y, Mannens M, et al. Aniridia-associated cytogenetic rearrangements suggest that a position effect may cause the mutant phenotype. Hum Mol Genet. Mar 1995;4(3):415-422.

176. Pfeifer D, Kist R, Dewar K, Devon K, Lander ES, Birren B, Korniszewski L, Back E, Scherer G. Campomelic dysplasia translocation breakpoints are scattered over 1 Mb proximal to SOX9: evidence for an extended control region. Am J Hum Genet. Jul 1999;65(1):111-124.

177. de Kok YJ, Merkx GF, van der Maarel SM, Huber I, Malcolm S, Ropers HH, Cremers FP. A duplication/paracentric inversion associated with familial X-linked deafness (DFN3) suggests the presence of a regulatory element more than 400 kb upstream of the POU3F4 gene. Hum Mol Genet. Nov 1995;4(11):2145-2150.

178. de Kok YJ, Vossenaar ER, Cremers CW, Dahl N, Laporte J, Hu LJ, Lacombe D, Fischel-Ghodsian N, Friedman RA, Parnes LS, Thorpe P, Bitner-Glindzicz M, Pander HJ, Heilbronner H, Graveline J, den Dunnen JT, Brunner HG, Ropers HH, Cremers FP. Identification of a hot spot for microdeletions in patients with X- linked deafness type 3 (DFN3) 900 kb proximal to the DFN3 gene POU3F4. Hum Mol Genet. Sep 1996;5(9):1229-1235.

179. Belloni E, Muenke M, Roessler E, Traverso G, Siegel-Bartelt J, Frumkin A, Mitchell HF, Donis-Keller H, Helms C, Hing AV, Heng HH, Koop B, Martindale D, Rommens JM, Tsui LC, Scherer SW. Identification of Sonic hedgehog as a

173 candidate gene responsible for holoprosencephaly. Nat Genet. Nov 1996;14(3):353-356.

180. Roessler E, Ward DE, Gaudenz K, Belloni E, Scherer SW, Donnai D, Siegel- Bartelt J, Tsui LC, Muenke M. Cytogenetic rearrangements involving the loss of the Sonic Hedgehog gene at 7q36 cause holoprosencephaly. Hum Genet. Aug 1997;100(2):172-181.

181. McElreavey K, Vilain E, Barbaux S, Fuqua JS, Fechner PY, Souleyreau N, Doco- Fenzy M, Gabriel R, Quereux C, Fellous M, Berkovitz GD. Loss of sequences 3' to the testis-determining gene, SRY, including the Y pseudoautosomal boundary associated with partial testicular determination. Proc Natl Acad Sci U S A. Aug 6 1996;93(16):8590-8594.

182. Mattei MG, Mattei JF, Ayme S, Giraud F. X-autosome translocations: cytogenetic characteristics and their consequences. Hum Genet. 1982;61(4):295-309.

183. Schmidt M, Du Sart D. Functional disomies of the X chromosome influence the cell selection and hence the X inactivation pattern in females with balanced X- autosome translocations: a review of 122 cases. Am J Med Genet. Jan 15 1992;42(2):161-169.

184. Wolff DJ, Schwartz S, Montgomery T, Zackowski JL. Random X inactivation in a girl with a balanced t(X;9) and an abnormal phenotype. Am J Med Genet. Jun 5 1998;77(5):401-404.

185. Sands ME. Mental retardation in association with a balanced X-autosome translocation and random inactivation of the X chromosomes. Clin Genet. May 1980;17(5):309-316.

186. Hagemeijer A, Hoovers J, Smit EM, Bootsma D. Replication pattern of the X chromosomes in three X/autosomal translocations. Cytogenet Cell Genet. 1977;18(6):333-348.

187. Mattei MG, Mattei JF, Ayme S, Malpuech G, Giraud F. A dynamic study in two new cases of X chromosome translocations. Hum Genet. Apr 24 1978;41(3):251- 257.

174 188. Mattei JF, Mattei MG, Giraud F. DNA replication patterns and consequences of translocations between X chromosomes and autosomes. In: Sandberg AA, ed. Cytogenetics of the Mammalian X-chromosome. New York: Alan R. Liss; 1983:449-459.

189. Popova BC, Tada T, Takagi N, Brockdorff N, Nesterova TB. Attenuated spread of X-inactivation in an X;autosome translocation. Proc Natl Acad Sci U S A. May 16 2006;103(20):7706-7711.

190. Sharp AJ, Spotswood HT, Robinson DO, Turner BM, Jacobs PA. Molecular and cytogenetic analysis of the spreading of X inactivation in X;autosome translocations. Hum Mol Genet. Dec 1 2002;11(25):3145-3156.

191. Kaiser-Rogers K, Rao K. Structural Chromosome Rearrangements. In: Gersen SL, Keagle MB, eds. The Principles of Clinical Cytogenetics. (Second ed). Totowa: Humana Press; 2005:165-206.

192. Dudding BA, Gorlin RJ, Langer LO. The oto-palato-digital syndrome. A new symptom-complex consisting of deafness, dwarfism, cleft palate, characteristic facies, and a generalized bone dysplasia. Am J Dis Child. Feb 1967;113(2):214- 221.

193. Taybi H. Generalized skeletal dysplasia with multiple anomalies. A note on Pyle's disease. Am J Roentgenol Radium Ther Nucl Med. Sep 1962;88:450-457.

194. Robertson SP. Otopalatodigital syndrome spectrum disorders: otopalatodigital syndrome types 1 and 2, frontometaphyseal dysplasia and Melnick-Needles syndrome. Eur J Hum Genet. Jan 2007;15(1):3-9.

195. Gorlin RJ. Minneapolis; 1993.

196. Hidalgo-Bravo A, Pompa-Mera EN, Kofman-Alfaro S, Gonzalez-Bonilla CR, Zenteno JC. A novel filamin A D203Y mutation in a female patient with otopalatodigital type 1 syndrome and extremely skewed X chromosome inactivation. Am J Med Genet A. Jul 15 2005;136(2):190-193.

197. Smith DW. Recognizable patterns of human malformations: genetic, embryologic and clinical aspects. Philadelphia: WB Saunders; 1970.

175 198. Zaytoun GM, Harboyan G, Kabalan W. The oto-palato-digital syndrome: variable clinical expressions. Otolaryngol Head Neck Surg. Feb 2002;126(2):129-140.

199. Gorlin RJ, Poznanski AK, Hendon I. The oto-palato-digital (OPD) syndrome in females. Oral Surg Oral Med Oral Pathol. Feb 1973;35(2):218-224.

200. Pazzaglia UE, Beluffi G. Oto-Palato-Digital syndrome in four generations of a large family. Clin Genet. Oct 1986;30(4):338-344.

201. Hoar DI, Field LL, Beards F, Hoganson G, Rollnick B, Hoo JJ. Tentative assignment of gene for oto-palato-digital syndrome to distal Xq (Xq26-q28). Am J Med Genet. Jan 15 1992;42(2):170-172.

202. Biancalana V, Le Marec B, Odent S, van den Hurk JA, Hanauer A. Oto-palato- digital syndrome type I: further evidence for assignment of the locus to Xq28. Hum Genet. Dec 1991;88(2):228-230.

203. Robertson SP, Walsh S, Oldridge M, Gunn T, Becroft D, Wilkie AO. Linkage of otopalatodigital syndrome type 2 (OPD2) to distal Xq28: evidence for allelism with OPD1. Am J Hum Genet. Jul 2001;69(1):223-227.

204. Fitch N, Jequier S, Gorlin R. The oto-palato-digital syndrome, proposed type II. Am J Med Genet. Aug 1983;15(4):655-664.

205. Superti-Furga A, Gimelli F. Fronto-metaphyseal dysplasia and the oto-palato- digital syndrome. Dysmorph Clin Genet. 1987;1:2-5.

206. Verloes A, Lesenfants S, Barr M, Grange DK, Journel H, Lombet J, Mortier G, Roeder E. Fronto-otopalatodigital osteodysplasia: clinical evidence for a single entity encompassing Melnick-Needles syndrome, otopalatodigital syndrome types 1 and 2, and frontometaphyseal dysplasia. Am J Med Genet. Feb 28 2000;90(5):407-422.

207. Robertson S, Gunn T, Allen B, Chapman C, Becroft D. Are Melnick-Needles syndrome and oto-palato-digital syndrome type II allelic? Observations in a four- generation kindred. Am J Med Genet. Aug 22 1997;71(3):341-347.

176 208. Robertson SP, Jenkins ZA, Morgan T, Ades L, Aftimos S, Boute O, Fiskerstrand T, Garcia-Minaur S, Grix A, Green A, Der Kaloustian V, Lewkonia R, McInnes B, van Haelst MM, Macini G, Illes T, Mortier G, Newbury-Ecob R, Nicholson L, Scott CI, Ochman K, Brozek I, Shears DJ, Superti-Furga A, Suri M, Whiteford M, Wilkie AO, Krakow D. Frontometaphyseal dysplasia: mutations in FLNA and phenotypic diversity. Am J Med Genet A. Aug 15 2006;140(16):1726-1736.

209. Gorlin RJ, Cohen MM, Jr. Frontometaphyseal dysplasia. A new syndrome. Am J Dis Child. Sep 1969;118(3):487-494.

210. Zenker M, Nahrlich L, Sticht H, Reis A, Horn D. Genotype-epigenotype- phenotype correlations in females with frontometaphyseal dysplasia. Am J Med Genet A. May 15 2006;140(10):1069-1073.

211. Melnick JC, Needles CF. An undiagnosed bone dysplasia. A 2 family study of 4 generations and 3 generations. Am J Roentgenol Radium Ther Nucl Med. 1966;97:39-48.

212. Kristiansen M, Knudsen GP, Soyland A, Westvik J, Orstavik KH. Phenotypic variation in Melnick-Needles syndrome is not reflected in X inactivation patterns from blood or buccal smear. Am J Med Genet. Mar 1 2002;108(2):120-127.

213. Robertson SP, Twigg SR, Sutherland-Smith AJ, Biancalana V, Gorlin RJ, Horn D, Kenwrick SJ, Kim CA, Morava E, Newbury-Ecob R, Orstavik KH, Quarrell OW, Schwartz CE, Shears DJ, Suri M, Kendrick-Jones J, Wilkie AO. Localized mutations in the gene encoding the cytoskeletal protein filamin A cause diverse malformations in humans. Nat Genet. Apr 2003;33(4):487-491.

214. Fox JW, Lamperti ED, Eksioglu YZ, Hong SE, Feng Y, Graham DA, Scheffer IE, Dobyns WB, Hirsch BA, Radtke RA, Berkovic SF, Huttenlocher PR, Walsh CA. Mutations in filamin 1 prevent migration of cerebral cortical neurons in human periventricular heterotopia. Neuron. Dec 1998;21(6):1315-1325.

215. Fink JM, Dobyns WB, Guerrini R, Hirsch BA. Identification of a duplication of Xq28 associated with bilateral periventricular nodular heterotopia. Am J Hum Genet. Aug 1997;61(2):379-387.

216. Guerrini R, Mei D, Sisodiya S, Sicca F, Harding B, Takahashi Y, Dorn T, Yoshida A, Campistol J, Kramer G, Moro F, Dobyns WB, Parrini E. Germline

177 and mosaic mutations of FLN1 in men with periventricular heterotopia. Neurology. Jul 13 2004;63(1):51-56.

217. Lu J, Sheen V. Periventricular heterotopia. Epilepsy Behav. Sep 2005;7(2):143- 149.

218. Sheen VL, Dixon PH, Fox JW, Hong SE, Kinton L, Sisodiya SM, Duncan JS, Dubeau F, Scheffer IE, Schachter SC, Wilner A, Henchy R, Crino P, Kamuro K, DiMario F, Berg M, Kuzniecky R, Cole AJ, Bromfield E, Biber M, Schomer D, Wheless J, Silver K, Mochida GH, Berkovic SF, Andermann F, Andermann E, Dobyns WB, Wood NW, Walsh CA. Mutations in the X-linked filamin 1 gene cause periventricular nodular heterotopia in males as well as in females. Hum Mol Genet. Aug 15 2001;10(17):1775-1783.

219. Gorlin JB, Yamin R, Egan S, Stewart M, Stossel TP, Kwiatkowski DJ, Hartwig JH. Human endothelial actin-binding protein (ABP-280, nonmuscle filamin): a molecular leaf spring. J Cell Biol. Sep 1990;111(3):1089-1105.

220. Patrosso MC, Repetto M, Villa A, Milanesi L, Frattini A, Faranda S, Mancini M, Maestrini E, Toniolo D, Vezzoni P. The exon-intron organization of the human X-linked gene (FLN1) encoding actin-binding protein 280. Genomics. May 1 1994;21(1):71-76.

221. Hartwig JH, Stossel TP. Isolation and properties of actin, myosin, and a new actinbinding protein in rabbit alveolar macrophages. J Biol Chem. Jul 25 1975;250(14):5696-5705.

222. Stossel TP, Condeelis J, Cooley L, Hartwig JH, Noegel A, Schleicher M, Shapiro SS. Filamins as integrators of cell mechanics and signalling. Nat Rev Mol Cell Biol. Feb 2001;2(2):138-145.

223. Feng Y, Walsh CA. The many faces of filamin: a versatile molecular scaffold for cell motility and signalling. Nat Cell Biol. Nov 2004;6(11):1034-1038.

224. Popowicz GM, Schleicher M, Noegel AA, Holak TA. Filamins: promiscuous organizers of the cytoskeleton. Trends Biochem Sci. Jul 2006;31(7):411-419.

225. Gimona M, Djinovic-Carugo K, Kranewitter WJ, Winder SJ. Functional plasticity of CH domains. FEBS Lett. Feb 20 2002;513(1):98-106. 178 226. Lebart MC, Mejean C, Casanova D, Audemard E, Derancourt J, Roustan C, Benyamin Y. Characterization of the actin binding site on smooth muscle filamin. J Biol Chem. Feb 11 1994;269(6):4279-4284.

227. Popowicz GM, Muller R, Noegel AA, Schleicher M, Huber R, Holak TA. Molecular structure of the rod domain of dictyostelium filamin. J Mol Biol. Oct 1 2004;342(5):1637-1646.

228. Kwak KB, Kambayashi J, Kang MS, Ha DB, Chung CH. Cell-penetrating inhibitors of calpain block both membrane fusion and filamin cleavage in chick embryonic myoblasts. FEBS Lett. May 24 1993;323(1-2):151-154.

229. Pudas R, Kiema TR, Butler PJ, Stewart M, Ylanne J. Structural basis for vertebrate filamin dimerization. Structure. Jan 2005;13(1):111-119.

230. Chakarova C, Wehnert MS, Uhl K, Sakthivel S, Vosberg HP, van der Ven PF, Furst DO. Genomic structure and fine mapping of the two human filamin gene paralogues FLNB and FLNC and comparative analysis of the filamin gene family. Hum Genet. Dec 2000;107(6):597-611.

231. Glogauer M, Arora P, Chou D, Janmey PA, Downey GP, McCulloch CA. The role of actin-binding protein 280 in integrin-dependent mechanoprotection. J Biol Chem. Jan 16 1998;273(3):1689-1698.

232. Bishop AL, Hall A. Rho GTPases and their effector proteins. Biochem J. Jun 1 2000;348 Pt 2:241-255.

233. Ohta Y, Hartwig JH, Stossel TP. FilGAP, a Rho- and ROCK-regulated GAP for Rac binds filamin A to control actin remodelling. Nat Cell Biol. Aug 2006;8(8):803-814.

234. Ohta Y, Suzuki N, Nakamura S, Hartwig JH, Stossel TP. The small GTPase RalA targets filamin to induce filopodia. Proc Natl Acad Sci U S A. Mar 2 1999;96(5):2122-2128.

235. Bellanger JM, Astier C, Sardet C, Ohta Y, Stossel TP, Debant A. The Rac1- and RhoG-specific GEF domain of Trio targets filamin to remodel cytoskeletal actin. Nat Cell Biol. Dec 2000;2(12):888-892.

179 236. Robertson SP. Filamin A: phenotypic diversity. Curr Opin Genet Dev. Jun 2005;15(3):301-307.

237. Disteche CM, Eicher EM, Latt SA. Late replication in an X-autosome translocation in the mouse: correlation with genetic inactivation and evidence for selective effects during embryogenesis. Proc Natl Acad Sci U S A. Oct 1979;76(10):5234-5238.

238. Young K, Barth CK, Moore C, Weaver DD. Otopalatodigital syndrome type II associated with omphalocele: report of three cases. Am J Med Genet. Feb 15 1993;45(4):481-487.

239. Cirullo RE, Dana S, Wasmuth JJ. Efficient procedure for transferring specific human genes into Chinese hamster cell mutants: interspecific transfer of the human genes encoding leucyl- and asparaginyl-tRNA synthetases. Mol Cell Biol. May 1983;3(5):892-902.

240. Allen RC, Zoghbi HY, Moseley AB, Rosenblatt HM, Belmont JW. Methylation of HpaII and HhaI sites near the polymorphic CAG repeat in the human androgen-receptor gene correlates with X chromosome inactivation. Am J Hum Genet. Dec 1992;51(6):1229-1239.

241. Kubota T, Nonoyama S, Tonoki H, Masuno M, Imaizumi K, Kojima M, Wakui K, Shimadzu M, Fukushima Y. A new assay for the analysis of X-chromosome inactivation based on methylation-specific PCR. Hum Genet. Jan 1999;104(1):49- 55.

242. McGinnis S, Madden TL. BLAST: at the core of a powerful and diverse set of sequence analysis tools. Nucleic Acids Res. Jul 1 2004;32(Web Server issue):W20-25.

243. Matys V, Fricke E, Geffers R, Gossling E, Haubrock M, Hehl R, Hornischer K, Karas D, Kel AE, Kel-Margoulis OV, Kloos DU, Land S, Lewicki-Potapov B, Michael H, Munch R, Reuter I, Rotert S, Saxel H, Scheer M, Thiele S, Wingender E. TRANSFAC: transcriptional regulation, from patterns to profiles. Nucleic Acids Res. Jan 1 2003;31(1):374-378.

244. Siepel A, Bejerano G, Pedersen JS, Hinrichs AS, Hou M, Rosenbloom K, Clawson H, Spieth J, Hillier LW, Richards S, Weinstock GM, Wilson RK, Gibbs RA, Kent WJ, Miller W, Haussler D. Evolutionarily conserved elements in 180 vertebrate, insect, worm, and yeast genomes. Genome Res. Aug 2005;15(8):1034- 1050.

245. Karolchik D, Baertsch R, Diekhans M, Furey TS, Hinrichs A, Lu YT, Roskin KM, Schwartz M, Sugnet CW, Thomas DJ, Weber RJ, Haussler D, Kent WJ. The UCSC Genome Browser Database. Nucleic Acids Res. Jan 1 2003;31(1):51-54.

246. King DC, Taylor J, Elnitski L, Chiaromonte F, Miller W, Hardison RC. Evaluation of regulatory potential and conservation scores for detecting cis- regulatory modules in aligned mammalian genome sequences. Genome Res. Aug 2005;15(8):1051-1060.

247. Zuker M. Mfold web server for nucleic acid folding and hybridization prediction. Nucleic Acids Res. Jul 1 2003;31(13):3406-3415.

248. Wirth J, Wagner T, Meyer J, Pfeiffer RA, Tietze HU, Schempp W, Scherer G. Translocation breakpoints in three patients with campomelic dysplasia and autosomal sex reversal map more than 130 kb from SOX9. Hum Genet. Feb 1996;97(2):186-193.

249. Davies SJ, Wise C, Venkatesh B, Mirza G, Jefferson A, Volpi EV, Ragoussis J. Mapping of three translocation breakpoints associated with orofacial clefting within 6p24 and identification of new transcripts within the region. Cytogenet Genome Res. 2004;105(1):47-53.

250. Marrone A, Walne A, Dokal I. Dyskeratosis congenita: telomerase, telomeres and anticipation. Curr Opin Genet Dev. Jun 2005;15(3):249-257.

251. Devriendt K, Matthijs G, Legius E, Schollen E, Blockmans D, van Geet C, Degreef H, Cassiman JJ, Fryns JP. Skewed X-chromosome inactivation in female carriers of dyskeratosis congenita. Am J Hum Genet. Mar 1997;60(3):581-587.

252. Vulliamy TJ, Knight SW, Dokal I, Mason PJ. Skewed X-inactivation in carriers of X-linked dyskeratosis congenita. Blood. Sep 15 1997;90(6):2213-2216.

253. Ferraris AM, Forni GL, Mangerini R, Gaetani GF. Nonrandom X-chromosome inactivation in hemopoietic cells from carriers of dyskeratosis congenita. Am J Hum Genet. Aug 1997;61(2):458-461.

181 254. Lachlan KL, Collinson MN, Sandford RO, van Zyl B, Jacobs PA, Thomas NS. Functional disomy resulting from duplications of distal Xq in four unrelated patients. Hum Genet. Oct 2004;115(5):399-408.

255. Feng Y, Chen MH, Moskowitz IP, Mendonza AM, Vidali L, Nakamura F, Kwiatkowski DJ, Walsh CA. Filamin A (FLNA) is required for cell-cell contact in vascular development and cardiac morphogenesis. Proc Natl Acad Sci U S A. Dec 26 2006;103(52):19836-19841.

256. Hart AW, Morgan JE, Schneider J, West K, McKie L, Bhattacharya S, Jackson IJ, Cross SH. Cardiac malformations and midline skeletal defects in mice lacking filamin A. Hum Mol Genet. Aug 15 2006;15(16):2457-2467.

257. Sanlaville D, Prieur M, de Blois MC, Genevieve D, Lapierre JM, Ozilou C, Picq M, Gosset P, Morichon-Delvallez N, Munnich A, Cormier-Daire V, Baujat G, Romana S, Vekemans M, Turleau C. Functional disomy of the Xq28 chromosome region. Eur J Hum Genet. May 2005;13(5):579-585.

258. Thelin WR, Chen Y, Gentzsch M, Kreda SM, Sallee JL, Scarlett CO, Borchers CH, Jacobson K, Stutts MJ, Milgram SL. Direct interaction with filamins modulates the stability and plasma membrane expression of CFTR. J Clin Invest. Feb 2007;117(2):364-374.

182