Dissertation Cover pages v1.0 EDITS 122815

139
Characterizing the Mechanism of Tumor Suppression by PBRM1 in Clear Cell Renal Cell Carcinoma David Schoenfeld Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy under the Executive Committee of the Graduate School of Arts and Sciences COLUMBIA UNIVERSITY 2016

Transcript of Dissertation Cover pages v1.0 EDITS 122815

Page 1: Dissertation Cover pages v1.0 EDITS 122815

Characterizing the Mechanism of Tumor Suppression by PBRM1 in Clear Cell Renal Cell Carcinoma

David Schoenfeld

Submitted in partial fulfillment of the

requirements for the degree of

Doctor of Philosophy

under the Executive Committee

of the Graduate School of Arts and Sciences

COLUMBIA UNIVERSITY

2016

Page 2: Dissertation Cover pages v1.0 EDITS 122815

© 2015

David Schoenfeld

All rights reserved

Page 3: Dissertation Cover pages v1.0 EDITS 122815

ABSTRACT

Characterizing the Mechanism of Tumor Suppression by PBRM1 in Clear Cell Renal Cell Carcinoma

David Schoenfeld

In this study, we investigated the mechanisms by which PBRM1 functions as a tumor suppressor

in clear cell renal cell carcinoma. PBRM1, also known as BAF180 or Polybromo, is a member of the

PBAF SWI/SNF chromatin remodeling complex. Cancer sequencing studies have revealed that SWI/SNF

components are widely mutated in cancer. PBRM1 is recurrently mutated in various human

malignancies, but it has a particularly high mutation rate in clear cell renal cell carcinoma: ~40% of clear

cell renal cell carcinomas have a PBRM1 mutation, making it the second most highly mutated gene in

clear cell renal cell carcinoma behind VHL. Although many recent studies have looked at how other

SWI/SNF components function in cancer control, relatively little is known about the tumor suppressive

mechanisms of PBRM1 in clear cell renal cell carcinoma.

To investigate PBRM1 function, we manipulated its expression in clear cell renal cell carcinoma

cell lines. In cell lines with intact PBRM1, we stably knocked down its expression using shRNA. In a cell

line with mutant PBRM1, we stably restored expression of the wild-type protein. We found that PBRM1

deficiency significantly enhanced the growth properties of cells, but only when the cells were grown under

stressful conditions, such as reduced serum or a 3-D culture environment. To investigate genes and

pathways influenced by PBRM1 that may confer this growth advantage, we compared gene expression

differences in the clear cell renal cell carcinoma cell lines and murine embryonic fibroblasts with or without

PBRM1. We found that PBRM1 regulated numerous cancer-related genes and pathways.

One gene, ALDH1A1, was consistently upregulated with PBRM1 deficiency across our cell lines.

Further expression analysis using two different clear cell renal cell carcinoma primary tumor datasets

revealed that PBRM1 mutation in primary tumors was also associated with higher ALDH1A1 levels.

ALDH1A1, or aldehyde dehydrogenase 1, is part of the retinoic acid metabolic pathway and irreversibly

converts retinaldehyde to retinoic acid. It functions in hematopoietic stem cell development, white versus

brown fat programming, and insulin signaling. Numerous studies have also identified ALDH1A1 as a

marker of tumor-initiating cells, also known as cancer stem cells. Not much is known about the regulation

of ALDH1A1 expression in cancer, and it has not previously been linked to PBRM1 or SWI/SNF. We

Page 4: Dissertation Cover pages v1.0 EDITS 122815

confirmed that stable knockdown of PBRM1 in clear cell renal cell carcinoma cell lines resulted in higher

ALDH1A1 mRNA and protein expression, and also higher ALDH1-class enzyme activity. Alternatively, re-

expression of wild-type PBRM1, but not cancer-associated mutant PBRM1, lowered ALDH1A1

expression and activity in the PBRM1-mutant line. Additionally, inhibiting ALDH1A1 or knocking it down

in the context of PBRM1 deficiency reduced anchorage-independent growth, while over-expressing

ALDH1A1 in the PBRM1-normal setting increased tumorsphere-forming capacity. These results suggest

that ALDH1A1 is not only a marker of tumor-initiating cells, but can also increase the tumorigenic

potential of cells.

Based on our gene expression analysis, we additionally explored PBRM1 regulation of the EGFR

and IFN pathways. PBRM1 decreased total EGFR protein levels and dampened downstream signaling.

These changes had functional consequences, as PBRM1 deficiency led to faster growth in response to

EGF stimulation. However, it did not create a setting of oncogenic addiction, as PBRM1 deficient cells

were also more resistant to EGFR inhibition. Alternatively, PBRM1 deficiency reduced basal and IFNα-

induced levels of IFI27, a pro-apoptotic interferon response gene, and made cells more resistant to

growth inhibition by IFNα. PBRM1 mutations in cancer would thus be expected to have wide-ranging

effects on a cell, and the targeting of any one specific downstream pathway might have limited efficacy.

Finally, we investigated the molecular mechanisms of how PBRM1 deficiency could alter

transcription, keeping in mind that PBRM1 is one subunit of the larger PBAF complex. In our clear cell

renal cell carcinoma cell lines, we found that mRNA and protein levels of another PBAF-specific subunit,

ARID2, increased with PBRM1 deficiency. PBRM1 mutation in primary tumors was also associated with

significantly higher ARID2 expression. Immunoprecipitation and glycerol gradient fractionation

experiments suggested that more ARID2 may associate with the SWI/SNF components BRG1 and SNF5

after PBRM1 knockdown. ARID2 ChIP-seq analysis revealed that this remnant PBAF-like complex was

bound to fewer locations in the genome, and its binding locations were broadly redistributed. Both gained

and lost ARID2 binding were associated with differential gene expression, of both upregulated and

downregulated genes, indicating that the genomic context influences whether PBAF-binding is activating

or repressive. Interestingly, we also found that ARID2 was required for some of the pro-tumorigenic

Page 5: Dissertation Cover pages v1.0 EDITS 122815

changes associated with PBRM1 deficiency, such as upregulation of ALDH1A1 and EGFR levels, but not

others, such as decreased IFI27 levels, implying alternative modes of transcriptional regulation.

In total, this study implicates PBRM1 in the regulation of numerous cancer-related genes and

pathways in clear cell renal cell carcinoma. PBRM1 mutation would alter the genomic binding of a

residual PBAF-like complex containing ARID2, leading to transcriptional changes that promote tumor

formation and growth. A better understanding of this oncogenic mechanism may reveal novel therapeutic

opportunities.

Page 6: Dissertation Cover pages v1.0 EDITS 122815

i

Table of Contents

List of Figures ………………………………………………………………………………………………….........iv

Acknowledgements………………………………………………………………………………………………….vi

Dedication………………………………………………………………………………………………………….. viii

CHAPTER 1: Introduction ………………………………………………………………………………………….1

Chromatin ………………………………………………………………………………………………………2

SWI/SNF complexes ………………………………………………………………………………………….2

SWI/SNF and cancer ………………………………………………………………………………………….5

PBRM1 background …………………………………………………………………………………………..9

Known roles of PBRM1 in tumor suppression ……………………………………………………………..9

Tumor suppression by other SWI/SNF subunits …………………………………………………………12

Clear Cell Renal Cell Carcinoma …………………………………………………………………………..14

The importance of understanding mechanisms of tumor suppression by PBRM1……………………16

CHAPTER 2: Characterizing the effects of PBRM1 deficiency on growth and gene expression …………17

BACKGROUND AND SIGNIFICANCE……………………………………………………………………...18

RESULTS ……………………………………………………………………………………………………...19

PBRM1 protein levels can be stably knocked-down with lentiviral shRNA vectors ……………….19

Stable expression of wild-type PBRM1 or cancer-associated mutants in a null setting ………….20

PBRM1 deficiency results in faster 2-D growth in reduced serum conditions ……………………..21

PBRM1 knockdown increases the 3-D growth capacity of cells …………………………………….24

Murine embryonic fibroblasts with conditional loss of Pbrm1 grow slower than matched

controls……………………………………………………………………………………………………..25

RNA-seq and microarray analysis reveal cancer-related pathways perturbed with PBRM1

deficiency ………………………………………………………………………………………………….26

RNA-seq analysis of 786-O and A-704 cell lines ……………………………………………26

Top-changed genes and pathways with PBRM1 alteration ………………………………..29

Microarray analysis of Pbrm1 conditional MEFs …………………………………………….34

Overlap of genes with significant expression changes in 786-O, A-704, and MEFs ……37

Analysis of primary tumor datasets reveals that ALDH1A1 is overexpressed in PBRM1

mutant tumors …………………………………………………………………………………..37

Page 7: Dissertation Cover pages v1.0 EDITS 122815

ii

DISCUSSION ………………………………………………………………………………………………….38

CHAPTER 3: PBRM1 suppresses tumorigenic potential through regulation of ALDH1A1 expression, and

the EGFR and IFN pathways ……………………………………………………………………………………..42

BACKGROUND AND SIGNIFICANCE……………………………………………………………………...43

RESULTS ……………………………………………………………………………………………………...44

PBRM1 status affects ALDH1A1 mRNA, protein, and activity levels ………………………………44

PBRM1 knockdown in 786-O increases tumorsphere-forming capacity …………………………...46

Expression of the T232P PBRM1 cancer mutant increases tumorsphere formation in A-704 cells

………………………………………………………………………………………………………………46

Anchorage-independent growth in 786-O requires ALDH1-class enzyme activity ………………..48

ALDH1A1 overexpression increases tumorsphere-forming capacity ………………………………49

PBRM1 deficiency increases sensitivity to EGF stimulation ………………………………………...50

PBRM1 status affects total EGFR levels and downstream signaling ………………….….50

PBRM1 status affects growth responsiveness to EGF stimulation ………………………..51

PBRM1 knockdown in 786-O renders cells more resistant to EGFR inhibition ………….52

Decreased sensitivity to IFNα with PBRM1 deficiency ……………………………………………….52

PBRM1 deficiency reduces basal and IFNα-induced IFI27 mRNA levels ………………..52

IFI27 mRNA levels are lower in PBRM1 mutant tumors ……………………………………54

A-704 cells expressing PBRM1 are more growth inhibited by IFNα ………………………54

DISCUSSION ………………………………………………………………………………………………….56

CHAPTER 4: With PBRM1 deficiency, ARID2 still binds in a SWI/SNF complex, but with altered genomic

localization associated with differential gene expression ……………………………………………………...59

BACKGROUND AND SIGNIFICANCE……………………………………………………………………...60

RESULTS ……………………………………………………………………………………………………...60

PBRM1 deficiency increases ARID2 protein levels …………………………………………………..60

Increased ARID2 levels are at least partially due to increased transcription ………………………61

In the absence of PBRM1, ARID2 still associates with SWI/SNF components …………………...62

Subcellular fractionation experiments reveal that ARID2 increases in both the soluble and

insoluble nuclear fractions ………………………………………………………………………………64

ARID2 ChIP-seq indicates altered genomic binding of the PBAF complex that remains after

PBRM1 knockdown ………………………………………………………………………………………66

After PBRM1 knockdown, ARID2 binds to fewer areas of the genome …………………..66

Page 8: Dissertation Cover pages v1.0 EDITS 122815

iii

k-means clustering of ARID2 binding enrichment reveals broad shifts in binding patterns

with PBRM1 knockdown ……………………………………………………………………….68

ARID2 binding differences correlate with gene expression changes ……………………..71

ARID2 binding patterns at specific loci ……………………………………………………….76

PBRM1 knockdown results in widespread increases in H3K4 trimethylation ……………………..79

ARID2 is required for some of the pro-tumorigenic effects of PBRM1 deficiency ………………...81

ALDH1A1 upregulation with PBRM1 deficiency requires ARID2 ………………………….81

ARID2 is required for tumorsphere formation but not cell viability ………………………...86

SNF5, BRG1, and BRM appear dispensable for ALDH1A1 upregulation with PBRM1

deficiency ………………………………………………………………………………………..87

ARID2 is also required for increased EGFR protein levels, but not for decreased IFI27

mRNA levels, seen with PBRM1 knockdown ………………………………………………..89

DISCUSSION ………………………………………………………………………………………………….92

CHAPTER 5: Conclusions ………………………………………………………………………………………..96

MATERIALS AND METHODS: ………………………………………………………………………………...101

REFERENCES: …………………………………………………………………………………………………..116

Page 9: Dissertation Cover pages v1.0 EDITS 122815

iv

List of Figures

Figure 1.1 – Subunit composition of mSWI/SNF complexes ……………………………………………...……3

Figure 1.2 – PBRM1 is widely mutated in cancer …………………………………………………………….....8

Figure 1.3 – The structure of PBRM1 ……………………………………………………………………………..9

Figure 2.1 – PBRM1 knockdown in 786-O ccRCC cell line ……………………………………………...……19

Figure 2.2 – PBRM1 knockdown in ACHN RCC cell line ……………………………………………...………20

Figure 2.3 – Stable expression of wild-type PBRM1 or cancer-associated mutants in A-704 …………….22

Figure 2.4 – 2-D Growth Curves of RCC cell lines at normal and reduced serum conditions …………….23

Figure 2.5 – 2-D Growth Curves of all A-704 lines at normal and reduced serum conditions …………….24

Figure 2.6 – PBRM1 knockdown in 786-O and ACHN cells increases colony formation in soft agar ……25

Figure 2.7 – PBRM1 knockdown in 786-O cells significantly increases tumor xenograft growth ………....26

Figure 2.8 – MEFs with conditional loss of Pbrm1 grow slower than matched control cells ………………27

Figure 2.9 – Histograms of cumulative RPKM value for different sets of samples …………………………30

Figure 2.10 – Histograms of log2 fold-change in RPKM after filtering low-expressed genes ……………...31

Figure 2.11 – Heatmaps of the top changed genes with PBRM1 alteration at each serum condition ……32

Figure 2.12 – Gene set enrichment analysis of RNA-seq meta-analysis identifies pathways significantly

altered by PBRM1…………………………………………….……………………………………………...…….33

Figure 2.13 – Gene ontology analysis of RNA-seq meta-analysis identifies biological processes

significantly altered by PBRM1……………………………………………...…………………………………….35

Figure 2.14 – Significantly altered genes and pathways in Pbrm1 NULL MEFs ……………………………36

Figure 2.15 - Overlap of genes with significant expression changes in 786-O, A-704, and MEFs ……….37

Figure 2.16 – ALDH1A1 is upregulated in PBRM1 mutant tumors …………………………………………..39

Figure 3.1 – PBRM1 status affects ALDH1A1 mRNA, protein, and activity levels …………………………45

Figure 3.2 – PBRM1 knockdown in 786-O increases tumorsphere-forming capacity ……………………...47

Figure 3.3 – PBRM1 mutant T232P increases tumorsphere-forming capacity in A-704 cells …………….48

Figure 3.4 – Anchorage-independent growth in 786-O requires ALDH1-class enzyme activity …............49

Figure 3.5 – ALDH1A1 overexpression increases tumorsphere-forming capacity …………………………50

Figure 3.6 – PBRM1 status affects total EGFR levels and downstream signaling ………………………...51

Figure 3.7 – PBRM1 status affects growth responsiveness to EGF stimulation ……………………………52

Figure 3.8 – PBRM1 knockdown in 786-O renders cells more resistant to EGFR inhibition ….................53

Page 10: Dissertation Cover pages v1.0 EDITS 122815

v

Figure 3.9 – PBRM1 deficiency reduces basal and IFNα-induced IFI27 mRNA levels ……………………54

Figure 3.10 – IFI27 mRNA levels are lower in PBRM1 mutant tumors ………………………………………55

Figure 3.11 – A-704 cells expressing PBRM1 are more growth inhibited by IFNα …………………………55

Figure 4.1 - PBRM1 deficiency increases ARID2 protein levels ……………………………………………...61

Figure 4.2 - Increased ARID2 levels are at least partially due to increased transcription …………………62

Figure 4.3 – In the absence of PBRM1, ARID2 still associates with SWI/SNF components ……………...63

Figure 4.4 – Glycerol gradient fractionation of 786-O cell lines ………………………………………………64

Figure 4.5 – Subcellular fractionation of the 786-O lines ……………………………………………………...65

Figure 4.6 – Genomic annotation of ARID2 binding in 786-O lines …………………………………………..67

Figure 4.7 – ARID2 binding enrichment profiles for ARID2 peaks gained or lost with PBRM1 knockdown

………………………………………………………………………………………………………………………..68

Figure 4.8 – k-means clustering of ARID2 binding enrichment in 786-O control cells ……………………..69

Figure 4.9 – k-means clustering of ARID2 binding enrichment in 786-O PBRM1 shRNA lines …………..71

Figure 4.10 – ARID2 and input DNA average binding profiles for the top upregulated, top downregulated,

and unexpressed genes ....………………………………………………………………………………………..72

Figure 4.11 – BETA analysis of ARID2 binding changes and differential expression ……………………...74

Figure 4.12 – BETA-identified direct gene targets that lose ARID2 binding and have increased gene

expression with PBRM1 knockdown …………………………………………………………………………….75

Figure 4.13 - ARID2 binding patterns at specific loci ………………………………………………………77-79

Figure 4.14 – BETA analysis of H3K4Me3 and H3K4Me1 changes and differential expression …………82

Figure 4.15 – BETA analysis of H3K27Ac, H3K27Me3, and H3K9Ac changes and differential expression

………………………………………………………………………………………………………………………..83

Figure 4.16 – BETA-identified direct gene targets with histone mark changes in H3K4Me3 and H3K4Me1

and differential gene expression …………………………………………………………………………………84

Figure 4.17 – H3K4Me3 binding patterns at specific loci ……………………………………………………..85

Figure 4.18 - ALDH1A1 upregulation with PBRM1 deficiency requires ARID2 …………………………….86

Figure 4.19 – ARID2 is required for tumorsphere formation ………………………………………………….87

Figure 4.20 – ARID2 knockdown has only minor effects on cell growth and viability …………………….. 88

Figure 4.21 - SNF5, BRG1, and BRM appear dispensable for ALDH1A1 upregulation with PBRM1

deficiency …………………………………………………………………………………………………………...90

Figure 4.22 – BRG1 is not required for tumorsphere formation ………………………………………………91

Figure 4.23 - ARID2 is also required for increased EGFR protein levels, but not for decreased IFI27

mRNA levels, seen with PBRM1 knockdown …………………………………………………………………...91

Figure 5.1 – Proposed models of how PBRM1 deficiency leads to transcriptional changes ……………...99

Page 11: Dissertation Cover pages v1.0 EDITS 122815

vi

Acknowledgements

I would first like to thank my research mentor, Dr. Ramon Parsons. He has provided patient and

thoughtful guidance over the years, and manages to strike the right balance between offering advice and

suggesting ideas while still allowing you to grow and explore as a scientist. His continued belief in and

enthusiasm for the scientific enterprise serves as an inspiration to everyone around him. More

importantly, he is also a kind and generous person.

I would also like to thank all the members of the Parsons lab, past and present. You have

generously offered help and advice, and made the lab a great place to work. I genuinely looked forward

to all our lab outings (baseball games, the beach, beer gardens), and will miss them. And even though

most of you work on PTEN-related projects, we still got along. I would like to thank all the research

assistants who’ve worked in the lab (Meaghan Dendy, Zach Rapp, Ari Pollock, and Sarah Pegno) for

keeping things running smoothly. I would also like to thank Rhaisili Rosario for all her assistance and

good cheer – whenever you had a question or needed to get something done, Rhaisili was the person to

go to. Thank you to Zach Rapp, for helping with some mouse work and the cool reverse high-fives.

Thank you to Deepti Mathur for helping with the microarray analysis all those years ago. A big thank you

to Nicole Steinbach for all the immense help and discussion about ChIP-seq – without you blazing the

way in the lab, my ChIP-seq experiments would have been impossible. I would also like to thank Dr.

Doug Barrows for being a good friend to me and my wife Sarah, and his appreciation of how Eli Manning

is a first-ballot hall-of-famer.

I would also like to thank and acknowledge all the people who have assisted with this work. I

would first like to thank Sakellarios Zairis in the lab of Dr. Raul Rabadan for performing a lot of the RNA-

seq and ChIP-seq analysis, and for introducing me to coding (or at least trying to). I would also like to

thank Dr. Rabadan for his advice on data analysis and presentation. Thank you to William Su for

performing the mutagenesis reactions, running a lot of growth curves, running qPRC – generally, being

up for anything. I would like to thank Dr. Ravi Sachidanandam and the members of his lab for performing

the ChIP-seq sequencing and offering advice and guidance along the way. Additionally, thank you to Dr.

Dan Hasson in the lab of Dr. Emily Bernstein for helping teach our lab about native ChIP-seq and lending

us your expertise. I would also like to acknowledge the Transgenic Mouse Core at Columbia University

Page 12: Dissertation Cover pages v1.0 EDITS 122815

vii

for helping make the Pbrm1 fl/fl mouse, and the Genomics Core Facility at the Icahn Institute and

Department of Genetics and Genomic Sciences for performing the RNA-seq library preparation and

sequencing.

I would like to thank my thesis committee, Drs. Jan Kitajewski, Adolfo Ferrando, Wei Gu, and

Ramon Parsons. I enjoyed our committee meetings because I always felt that we were having a

discussion that produced helpful critiques, good ideas, and new directions. Thank you for your time and

assistance. I would also like to thank Dr. Emily Bernstein for agreeing to serve on my defense committee

– I look forward to your comments.

I would also like to thank the Columbia University Integrated Program in Cellular, Molecular and

Biomedical Studies. Thank you to Zaia Sivo for all your help and patience over the years, and to Ron

Liem and Lori Sussel for running a great program. I would also like to thank the Columbia University

MD/PhD program, although they’re not through with me yet. Thank you to Jeffrey Brandt for your help

over the years, and to Drs. Shelanski and Reiner for also running a great program. I would also like to

thank Dr. Patrice Spitalnik for always being available to chat and assure you that everything will be

alright.

Lastly, I would like to thank my family for all their love and support. I would like to thank my

brothers Jon and Adam for all the fun times through the years, for both being an inspiration, and for all the

encouragement. And to my parents, I can’t even begin to thank you – for all your encouragement and

guidance through the years, your support and generosity – none of this would have been possible without

you.

Page 13: Dissertation Cover pages v1.0 EDITS 122815

viii

Dedication

This thesis is dedicated to my wonderful wife, Sarah, and my daughter, Rachel. Sarah, you were

a great labmate and friend before you became more. Coming to New York for school would have been

worth it just to meet you. You’ve made the past few years amazing, and I’ll always treasure our walks

home through Central Park, our cookouts, and even our time in the lab together. Thank you for all your

love and support, and for our spectacular daughter Rachel.

Page 14: Dissertation Cover pages v1.0 EDITS 122815

1

CHAPTER 1: Introduction

Page 15: Dissertation Cover pages v1.0 EDITS 122815

2

Chromatin

In eukaryotic organisms, DNA is packaged with both histone and non-histone proteins to form

chromatin, the basic organizing principle of the genome. The basic unit of chromatin is the nucleosome,

which consists of ~146 base-pairs of DNA wrapped around a histone octamer (two copies each of H2A,

H2B, H3, and H4) (Luger et al., 2012). Nucleosomes are connected by small fragments of “linker” DNA,

and are organized into higher order chromatin structures. The chromatin state of a cell, however, is

highly dynamic, and changes in conjunction with such processes as DNA replication, repair, and

transcription. Chromatin state is governed by a variety of mechanisms, including DNA methylation,

histone variant incorporation, histone post-translational modification, and ATP-dependent chromatin

remodeling (Chen and Dent, 2014). Among the ATP-dependent chromatin remodelers, there are four

major families of complexes: SWI/SNF, ISWI, chromodomain-helicase DNA-binding protein (CHD), and

INO80 complexes.

SWI/SNF complexes

SWI/SNF complexes are multi-subunit chromatin remodeling complexes that alter the connection

between DNA and the histone octamer. Members of the SWI/SNF complex were originally identified in

Saccharomyces cerevisiae in two independent screens for mutants affecting mating type switching

(SWItching Defective, SWI) or growth on sucrose (Sucrose NonFermenting, SNF) (Stern et al., 1984)

(Breeden and Nasmyth, 1987) (Neigeborn and Carlson, 1984). Several of the identified genes were later

found to encode proteins that associate in a large multisubunit complex that is capable of altering

nucleosome structure in an ATP-dependent manner (Cairns et al., 1994) (Cote et al., 1994). The

Kornberg lab subsequently identified a similar complex in S. cerevisiae that shared some of the same

subunits as the original complex and could also remodel chromatin in an ATP-dependent manner (Cairns

et al., 1996). This complex, which they termed RSC (for Remodels the Structure of Chromatin), was

unique, however, in its high abundance and absolute requirement for viability. Similar complexes to

SWI/SNF and RSC were later identified in Drosophila melanogaster (bap and pbap) based on homology

of subunits and similar biochemical activity (Tamkun et al., 1992) (Papoulas et al., 1998). The Crabtree

group purified the mammalian SWI/SNF complex using similar methods (Wang et al., 1996).

Page 16: Dissertation Cover pages v1.0 EDITS 122815

3

Mammalian SWI/SNF complexes consist of 12-16 subunits (Wang et al., 1996) (Kadoch et al.,

2013), which can be divided into a core complex that is capable of in vitro nucleosome remodeling, and

accessory subunits that are thought to confer functional specificity to the complexes in vivo (Phelan,

1999). The core complex consists of one of two mutually exclusive ATPase domains: either BRM (for

brahma homologue; also known as SMARCA2) or BRG1 (for BRM-related gene 1; also known as

SMARCA4); and SNF5 (also known as SMARCAB1, INI1, or BAF47), BAF155, and BAF170. BRG1-

containing complexes can be further divided based on their accessory subunits: ARID1A or ARID1B (also

known as BAF250A or B) define the BAF complex (for Brg1-Associated Factors; also known as SWI/SNF-

A), while PBRM1 (also known as BAF180, Polybromo-1, or PB1), ARID2 (also known as BAF200), and

BRD7 are found exclusively in the PBAF complex (for Polybromo Brg1-Associated Factors; also known

as SWI/SNF-B) (Wilson and Roberts, 2011). The mammalian BAF complex is thought to be more like the

yeast SWI/SNF complex, while PBAF is thought to more resemble the RSC complex. Figure 1.1 shows

the subunit composition of the different mammalian SWI/SNF complexes.

BRG1/BRM

BAF170

BAF155

SNF5

ARID1 (A or B)

B-actin

BAF57

BAF45 (A, B, C or D)

BAF53 (A or B)

BAF60 (A, B, or C)

SS18

BRG1

BAF170

BAF155

SNF5

BRD7

ARID2

PBRM1

B-actin

BAF57

BAF45 (A, B, C or D)

BAF53 (A or B)

BAF60 (A, B, or C)

SS18

PBAF BAF

Figure 1.1 - Subunit composition of mSWI/SNF complexes. BAF complexes contain one of two mutually

exclusive ATP-dependent DNA-helicases (either BRM or BRG1), whereas the PBAF complex only contains

BRG1. The complexes also contain accessory subunits that are thought to confer functional specificity in

vivo, by altering where the complexes bind in the genome and what co-factors they bind. PBRM1, along

with ARID2 and BRD7, are the defining subunits of the PBAF complex, while ARID1A and ARID1B are

specific to the BAF complex.

Page 17: Dissertation Cover pages v1.0 EDITS 122815

4

As chromatin remodelers, SWI/SNF complexes alter the association of DNA with histones in an

ATP-dependent manner. SWI/SNF nucleosome binding is thought to disrupt DNA-histone contacts and

induce DNA translocation and loop formation (Wilson and Roberts, 2011). Loop propagation then results

in either nucleosome sliding or ejection of an adjacent nucleosome. In either case, the end result is a

more exposed portion of DNA that is now capable of binding to additional cofactors. In yeast, SWI/SNF

action is exclusively activating, i.e., nearby genes are transcribed more (Carlson and Laurent, 1994).

However, in higher organisms, SWI/SNF activity can result in the activation or repression of nearby

genes, depending on the gene and cellular context (Chi et al., 2002) (Isakoff et al., 2005).

Specificity of action of SWI/SNF complexes is achieved through the complex combinatorial

assembly of its subunits, many of which exist as multiple isoforms (Wu et al., 2009). These different

complexes can be mutually exclusive depending on the cell type and state of differentiation. For

example, the esBAF complex identified by the Crabtree lab is only found in embryonic stem (ES) cells

and is essential for maintaining stem cell pluripotency and self-renewal capacity (Ho et al., 2009).

Alternatively, other SWI/SNF complexes are necessary for the terminal differentiation of certain cell types,

such as neurons or myocytes (Lessard et al., 2007) (Albini and Puri, 2010).

For complexes that co-exist in a particular cell, specificity of action is thought to be achieved

through the docking of specific transcription factors and/or complex interactions with chromatin (e.g.,

reading the “histone code”). SWI/SNF subunits are known to associate with a variety of other factors

involved in transcriptional control, including but not limited to histone modifying enzymes (Sif et al., 2001)

(Kia et al., 2008) (Wilson et al., 2010), cell cycle control proteins such as RB (Dunaief et al., 1994), p53

(Burrows et al., 2010), MYC (Romero et al., 2012), and smads (Xi et al., 2008), and nuclear hormone

receptors (Lemon et al., 2001). Through these interactions, SWI/SNF complexes are capable of altering

particular transcriptional programs. Additionally, multiple SWI/SNF subunits contain “reader” domains

that can bind post-translational histone modifications, such as bromodomains that bind acetylated lysine

residues and chromo-related domains that bind methylated histones. PBRM1 contains six tandem

bromodomains, BRG1 and BRM each have one bromodomain, and BAF155 and BAF170 each have one

chromo-related domain (Wu et al., 2009). These subunits may help target SWI/SNF complexes to

specific genomic loci based on histone modification status.

Page 18: Dissertation Cover pages v1.0 EDITS 122815

5

Chromatin immunoprecipitation-sequencing (ChIP-seq) employs next generation sequencing to

determine where a protein is binding in the genome at high resolution, and has previously been employed

to study some SWI/SNF subunits. The Crabtree group performed ChIP-seq on BRG1 in ES cells to see

where the esBAF complex was binding (Ho et al., 2009). They found that the complex was most highly

enriched at transcriptional start sites (TSS), and was playing roles in both gene activation and repression.

Interestingly, they concluded that esBAF played more of a repressive role to fine-tune the levels of key

ES-specific genes and prevent their overexpression, which would lead to perturbation of the pluripotency

circuitry and eventual differentiation. A 2011 ChIP-seq study investigating where the core SWI/SNF

constituents (BRG1, BAF155, BAF170, and SNF5) bind in HeLa cells supported some of the same

conclusions: SWI/SNF binds to TSS and mediates both gene activation and repression (Euskirchen et al.,

2011). This study also found high enrichment at enhancer sites, suggesting SWI/SNF involvement in

long-range gene regulation, and that some SWI/SNF subunits may assemble in preliminary configurations

and wait poised on gene regulatory elements to receive the rest of the complex. This intriguing

conclusion is supported by another study in D. melanogaster that found that the SWI/SNF complexes bap

and pbap function as holoenzymes, in which the core complex can assemble on its own on the genome

but cannot participate in transcriptional control in vivo without the complex-specific subunits, such as

polybromo (the fly orthologue of PBRM1) (Moshkin et al., 2007).

SWI/SNF and cancer

It is now recognized that SWI/SNF subunits are widely mutated in human malignancies. Indeed,

surveys of recent cancer exome sequencing studies indicate that SWI/SNF components are mutated in

~20% of all cancers (Kadoch et al., 2013) (Shain and Pollack, 2013). SNF5 was the first SWI/SNF

subunit implicated as a tumor suppressor. Nearly all malignant rhabdoid tumors, a very aggressive

pediatric cancer associated with tumors in the brain, kidney, and soft tissue, are associated with biallelic

loss of SNF5 (Versteege et al., 1998). It is thought that these tumors have very few cancer-associated

mutations, and in some cases, SNF5 mutation is the only detectable somatic mutation (McKenna et al.,

2008) (Lee et al., 2012). Conditional knockout of SNF5 in mouse models leads to fully penetrant

Page 19: Dissertation Cover pages v1.0 EDITS 122815

6

lymphomas and rhabdoid tumors with a median onset of 11 weeks, an incredibly rapid timeframe

(Roberts et al., 2002).

Other SWI/SNF subunits are mutated across a variety of other malignancies (Helming et al.,

2014a). For example, BRG1 expression is absent in 15-50% of primary human non-small cell lung

cancer samples, and mutations are frequently seen in a variety of lung cancer cell lines (Reisman et al.,

2003) (Medina et al., 2008). BRG1 is also mutated in medulloblastoma (Parsons et al., 2011), 11% of

esophageal adenocarcinomas (Dulak et al., 2013), and to a lesser extent in melanoma and pancreatic

ductal adenocarcinoma (PDA) (Shain and Pollack, 2013). Additionally, it was recently discovered that

BRG1 is mutated and protein expression is lost in practically all cases of small cell carcinoma of the

ovary, hypercalcemic type, a rare, very aggressive form of ovarian cancer primarily diagnosed in young

women (Witkowski et al., 2014) (Jelinic et al., 2014) (Ramos et al., 2014).

The “targeting” subunits of the complex are also highly involved in cancer. The BAF-specific

subunit ARID1A is mutated in 50% of ovarian clear cell carcinomas, a highly lethal subtype (Jones et al.,

2010). It is also mutated in 30% of endometrioid tumors (Wiegand et al., 2010), and to a lesser extent in

multiple other cancers, including 14% of intrahepatic cholangiocarcinomas (Jiao et al., 2013), ~10% of

esophageal adenocarcinomas (Dulak et al., 2013), ~8% of gastric adenocarcinomas (Zang et al., 2012),

and a small percentage of PDA (Biankin et al., 2012). Meanwhile, PBAF-specific ARID2 is mutated in

~18% of hepatitis C-associated hepatocellular carcinomas (Li et al., 2011), ~9% of melanomas (Hodis et

al., 2012), and a small percentage of PDA (Biankin et al., 2012).

The other PBAF-specific subunit, PBRM1, is also mutated in human malignancies. In a 2008

study, the Parsons lab used representational difference analysis to identify candidate tumor suppressor

genes by looking for chromosomal regions that are homozygously deleted in the breast cancer cell line

HCC1143 (Xia et al., 2008). This approach revealed a 70 kilobase deletion at chromosomal location

3p21 that encodes exons 12-22 of PBRM1. Screening of breast cancer cell lines and primary tumors

identified three other PBRM1 truncation mutations (~4% primary tumors and cell lines were mutated). In

a 2011 study, exome sequencing of seven cases of clear cell renal cell carcinoma (ccRCC) identified four

homozygous truncating mutations in the PBRM1 gene (Varela et al., 2011). Confirmatory analysis of 227

primary ccRCC samples revealed PBRM1 mutations in 41% of the cases. Moreover, in this same study,

Page 20: Dissertation Cover pages v1.0 EDITS 122815

7

analysis of data from a forward genetic screen using a conditional Sleeping Beauty transposon system

looking for drivers of PDA in a KrasG12D mouse model revealed significant enrichment of insertion sites in

PBRM1, across multiple stages of pancreatic cancer. Another study identified structural mutations

converging on SWI/SNF subunits, including PBRM1, in a PDA panel using high-resolution genomic

profiling and mutational data (Shain et al., 2012). Overall, aberrant SWI/SNF subunits were present in

34% of samples tested, suggesting a significant role for SWI/SNF activity in pancreatic tumorigenesis.

Additionally, the same study that identified recurrent ARID1A mutations in intrahepatic

cholangiocarcinomas also detected PBRM1 mutations in 13% of cases and 25% of gallbladder

carcinomas (Jiao et al., 2013). Studies from the Cancer Genome Atlas (TCGA) have found similar

PBRM1 mutations rates in these cancers, along with identifying new malignancies with recurrent PBRM1

mutations, such as bladder urothelial carcinoma (9%) and stomach adenocarcinoma (7%) (Cancer

Genome Atlas Research, 2014b) (Cancer Genome Atlas Research, 2014a). The mutation frequency of

PBRM1 across various cancers is presented in Figure 1.2, which summarizes data from 105 studies as

presented by the cBioPortal for Cancer Genomics (Gao et al., 2013) (Cerami et al., 2012).

Page 21: Dissertation Cover pages v1.0 EDITS 122815

8

ccR

CC

Ch

ola

ngi

oca

rcin

om

a

Bla

dd

er

Sto

mac

h

Fig

ure

1.2

– P

BR

M1

is

wid

ely

mu

tate

d i

n c

an

ce

r.

Mu

tation

s o

f P

BR

M1

are

fo

un

d in

~4

0%

of cle

ar

cell

ren

al cell

ca

rcin

om

as (

ccR

CC

), m

akin

g it

the

se

con

d m

ost

hig

hly

mu

tate

d g

ene

in c

cR

CC

(b

eh

ind

VH

L).

P

BR

M1 is m

uta

ted

to

a lesse

r d

eg

ree

in

oth

er

typ

es o

f can

ce

r, in

clu

din

g

ch

ola

ngio

ca

rcin

om

a, b

ladd

er,

an

d s

tom

ach

can

ce

r.

Th

e f

requ

en

cie

s o

f P

BR

M1

mu

tatio

n in v

ari

ou

s h

um

an

ma

ligna

ncie

s a

re s

ho

wn

, a

s c

om

pile

d

by t

he

cB

ioP

ort

al fo

r C

ance

r G

en

om

ics in N

ove

mbe

r 2

01

5.

Fro

m c

Bio

Po

rtal

fo

r C

ance

r G

eno

mic

s, N

ove

mb

er

201

5

Page 22: Dissertation Cover pages v1.0 EDITS 122815

9

PBRM1 background

PBRM1 is a 180 kilodalton protein encoded by the PBRM1 gene located at chromosomal position

3p21 in Homo sapiens. As shown in Figure 1.3, it consists of six tandem bromodomains, two tandem

bromo-adjacent homology (BAH) domains, a DNA-binding high mobility group (HMG) domain, and two

LXXLL hormone recognition motifs. The bromodomains are capable of binding to acetylated lysine

residues, such as those found on histone tails (Chandrasekaran and Thompson, 2007). It is the only

protein found in humans with this many bromodomains. Although the exact function of the BAH domains

is unknown, histone acetylation at H4K16 and tri-methylation at H3K79 have been implicated as key

regulators of BAH-domain containing protein activity (Armache et al., 2011). Based on its functional

domain composition, PBRM1 is thought to represent the combination of three individual components of

the yeast RSC complex: Rsc1, Rsc2, and Rsc4 (Xue et al., 2000). Human PBRM1 shares 50% amino

acid similarity with Drosophila melanogaster and 92% similarity with Mus musculus. It was identified by

the Crabtree lab in a screen for the mammalian homologues of yeast SWI/SNF, and is one of the defining

subunits of the PBAF complex (Wang et al., 1996).

Known roles of PBRM1 in tumor suppression

Studies of PBRM1 function in lower organisms may provide valuable insights into PBRM1 tumor

suppression in mammals. In yeast, the RSC complex is involved in transcriptional regulation, DNA repair

(Kent et al., 2007) (Shim et al., 2007), chromatid cohesion (Gartenberg, 2009), and kinetochore formation

(Desai et al., 2009). In D. melanogaster, the pbap complex has been found to antagonize the EGFR

pathway. In one study, the authors looked at pbap-specific functions by focusing on bap170 (the fly

PBRM1 Isoform 2 (AAG48933.1), form most expressed in breast/renal epithelia

Figure 1.3 - The structure of PBRM1. PBRM1 consists of 6 tandem bromodomains, which bind to

acetylated lysine residues, 2 BAH domains, and a DNA-binding HMG domain. It also contains the two

indicated LXXLL hormone recognition motifs. Selected cancer missense mutations are indicated in red.

Page 23: Dissertation Cover pages v1.0 EDITS 122815

10

orthologue of ARID2) (Rendina et al., 2010). They found a genetic interaction between bap170 and

signaling components of the EGFR pathway, and concluded that bap170 negatively regulated the EGFR

pathway through the repression of rhomboid and activation of argos. The authors did not explore whether

these findings also translated to polybromo (the fly orthologue of PBRM1). In another study, the authors

used a temperature-sensitive snr1-mutant (the fly orthologue of human SNF5) that produced wing

patterning phenotypes to conduct an unbiased genetic modifier screen to look for SWI/SNF target genes

and cofactors (Curtis et al., 2011). Components of the EGFR, Notch, and DPP pathways were identified

as gene targets, and epistasis experiments revealed a connection between pbap (but not bap) and the

histone demethylase lsd1 as co-repressors of these pathways. Other studies have linked pbap to

regulation of genes involved in tissue remodeling and immune function (Carrera et al., 2008), and

polybromo has also been found to be required for histone H3.3 replacement at chromatin boundaries, a

key event in cellular memory (Nakayama et al., 2012).

In mice, the effects of global loss of Pbrm1 have been examined (Wang et al., 2004). Pbrm1

knockout mice displayed severe hypoplastic ventricle development and trophoblast placental defects, and

died between embryonic days 12.5-15.5. Mice heterozygous for the loss of Pbrm1 were viable and

appeared normal, although it is unclear whether they were followed long-term for tumor development.

The authors noted that the defects resembled those seen for loss of RXRα and PPARγ, and they

subsequently identified certain retinoic acid-induced genes (RARβ2 and CRABPII) that are important for

heart development and that are aberrantly expressed with Pbrm1 loss. This finding further supports the

notion that the PBAF complex is involved in nuclear hormone receptor transcriptional programs (Lemon et

al., 2001).

Another study in mice has linked PBRM1 to the repression of IL-10 in Th2 cells (Wurster et al.,

2012). The authors used a conditional knock-out mouse for Pbrm1 in T cells, and noted no defects in T

cell development. However, in the Th2 cell population, the immunoregulatory cytokine IL-10 was

upregulated. The authors found that Pbrm1 bound to regulatory elements of the IL-10 gene, and that

Pbrm1 loss precipitated a switch to Arid1a binding, increased acetylation, and increased CBP

recruitment.

Page 24: Dissertation Cover pages v1.0 EDITS 122815

11

Previous studies have also provided some clues as to how PBRM1 may function as a tumor

suppressor. The studies from the Parsons lab first identifying PBRM1 as a tumor suppressor indicated

that it exerts cell cycle control through induction of p21 (Xia et al., 2008). Restoration of PBRM1

expression in HCC1143 cells resulted in G1 arrest induced by PBRM1 binding to the p21 promoter.

Further, PBRM1 was critical for p21 upregulation and G1 arrest in response to TGFβ treatment or γ-

irradiation in the non-transformed breast line MCF10A. PBRM1, along with BRD7, has also been

identified as a vital component of p53-associated replicative senescence (Burrows et al., 2010). In the

2011 ccRCC exome sequencing study, knockdown of PBRM1 in ccRCC cell lines with wild-type PBRM1

led to increased proliferation, growth in soft agar, and migration (Varela et al., 2011). Gene set

enrichment analysis of microarray data from these knockdown experiments supported PBRM1’s role in

cell cycle control, and also associated it with chromosomal stability. This is in agreement with findings

from the initial study identifying the PBAF complex – the authors reported that the PBAF complex

localized to kinetochores during mitosis (Xue et al., 2000).

Two more recent studies have further supported PBRM1’s role in maintaining genome integrity.

In the first study, the authors found that PBRM1 was important for sister chromatid centromeric cohesion

in mouse embryonic stem cells, human fibroblasts, and an osteosarcoma cell line, and that siRNA

knockdown of PBRM1 or expression of PBRM1 cancer-associated missense mutations led to

chromosomal instability (Brownlee et al., 2014). The same group also reported that PBRM1 is required

for transcriptional silencing and subsequent DNA repair at early time points at a subset of DNA double-

strand breaks (Kakarougkas et al., 2014). Interestingly, the authors suggest that this process is

dependent on ATM phosphorylation of PBRM1, and that PBAF-chromatin remodeling activity may act

downstream of ATM to help promote H2AK119 monoubiquitination and double-strand break repair.

The structure of PBRM1 provides additional clues regarding its function. As mentioned above, its

six tandem bromodomains can bind acetylated lysine residues, and thus may help target the PBAF

complex to specific genomic locations based on histone modifications (Chandrasekaran and Thompson,

2007). A study of BAH domain activity suggests the two BAH domains may be playing a similar role

(Armache et al., 2011). Although ChIP-seq analysis has been performed on other SWI/SNF subunits,

there are no published reports of PBRM1 ChIP-seq showing where it binds in the genome. PBRM1 also

Page 25: Dissertation Cover pages v1.0 EDITS 122815

12

contains two hormone recognition motifs, and the PBAF complex has been found to associate with

multiple nuclear hormone receptors and to mediate hormone-induced transcription in vitro (Lemon et al.,

2001). No “hotspot” mutations of PBRM1 have been identified in human cancers. Instead, known

PBRM1 mutations are spread across functional domains and consist of truncating nonsense mutations,

missense mutations, and in-frame deletions/insertions (Xia et al., 2008) (Varela et al., 2011) (Shain et al.,

2012).

The PBAF complex has also been linked to interferon signaling (Yan et al., 2005). This study

initially identified ARID2 as a PBAF complex member. Looking in HeLa cells, the authors found that

ARID2 was required for PBAF complex assembly – without it, the complex fell apart and PBRM1 was

degraded. However, the reverse was not true – PBRM1 knockdown did not cause a reciprocal decline in

ARID2 levels. In HCC1143 cells, where PBRM1 is not expressed, ARID2 was expressed and was still

capable of associating in a complex with other SWI/SNF components. Additionally, ARID2, but not

PBRM1, was required for the transcription of IFITM1, an interferon-responsive gene, in response to

interferon-α (IFNα) stimulation. These findings suggest that a PBAF-like complex remains even after

PBRM1 loss, and that this complex is still capable of altering transcriptional programs.

As part of their large-scale genetic and molecular profiling of ccRCC tumors, the Cancer Genome

Atlas consortium identified enriched gene classes in sets of differentially expressed genes between

tumors with mutations in different ccRCC tumor suppressors (VHL, PBRM1, SETD2, and BAP1) (Cancer

Genome Atlas Research, 2013). Tumors with PBRM1 mutations enriched for the following gene sets:

glycoproteins; signaling; secreted; and extracellular region (q-values: 2.8E-33; 1.6E-33; 1.5E-26; 9.5E-23,

respectively). The authors also performed unsupervised clustering of the tumors based on mRNA

expression, and found a higher PBRM1 mutation rate in a subtype associated with better survival.

Tumor suppression by other SWI/SNF subunits

The mechanisms of tumor suppression by other SWI/SNF subunits may also prove illuminating

for PBRM1 action. Restoration of SNF5 to a malignant rhabdoid tumor tissue culture model led to re-

expression of p15 and p16 by SNF5 recruitment, along with BRG1, to the promoter regions of these

genes. This recruitment was accompanied by the removal of PRC1 (polycomb repressive complex 1) and

Page 26: Dissertation Cover pages v1.0 EDITS 122815

13

PRC2 and subsequent loss of the H3K27me3 repressive mark (Kia et al., 2008). The authors additionally

saw recruitment of MLL1, a histone methyltransferase, and an increase in the H3K4Me3 active mark.

This study confirmed earlier work in D. melanogaster which showed that SWI/SNF proteins function as

trithorax group proteins and oppose the action of polycomb group proteins (Tamkun et al., 1992) (Kal et

al., 2000). More recent work has extended this principle to a mouse model where SNF5 is conditionally

deleted in peripheral T cells, leading to higher levels of EZH2, the methytransferase in the PRC2

complex, higher H3K27me3 at repressed targets, and ultimately transformation (Wilson et al., 2010).

Strikingly, double conditional loss of SNF5 and EZH2 completely ablated tumor formation. The

antagonism between EZH2 and SWI/SNF extends to other cancer types and SWI/SNF subunits. For

example, non-small cell lung cancers with BRG1 mutations display enhanced sensitivity to topoisomerase

II inhibitors in combination with EZH2 inhibition (Fillmore et al., 2015). The authors also found that EGFR

gain-of-function mutant tumors displayed the same sensitivity due to genetic antagonism between EGFR

and BRG1. Another report demonstrated a synthetic lethal relationship between EZH2 inhibition and

ARID1A mutations in ovarian cancer cells (Bitler et al., 2015).

Previous studies have also demonstrated that one must consider what remains of the SWI/SNF

complex when investigating mutations in individual subunits. For example, SNF5 loss is only

transforming when BRG1 is present, implying that aberrant SWI/SNF activity, but not the total lack of

activity, is oncogenic (Wang et al., 2009). A recently identified SWI/SNF complex member, SS18, also

dramatically alters complex formation when mutated in cancer (Kadoch and Crabtree, 2013). SS18 is

part of a chromosomal translocation found in practically all cases of synovial sarcoma (the SS18-SSX

fusion). This oncogenic fusion protein competes for binding in the complex with wild-type SS18, and

displaces SNF5 from the complex. The cancer-associated complex is then capable of binding to the

SOX2 locus, resulting in decreased H3K27Me3 levels and increased SOX2 transcription.

Numerous recent reports have also demonstrated a synthetic lethal relationship between

homologous SWI/SNF subunits. For example, two independent studies identified BRM as an essential

gene in BRG1 mutant cancers (Hoffman et al., 2014) (Wilson et al., 2014). In the BRG1 mutant setting,

BRM is upregulated and is incorporated more into residual SWI/SNF complexes. When BRM is then

depleted, the cells undergo cell cycle arrest and senescence. ARID1A and ARID1B, mutually exclusive

Page 27: Dissertation Cover pages v1.0 EDITS 122815

14

subunits in the BAF complex, exhibit a similar relationship (Helming et al., 2014b). Although co-mutations

of these genes occur, suggesting some cancer-promoting cooperativity, at least one functional allele of

ARID1B remains in the ARID1A mutant setting. These studies indicate that some level of SWI/SNF

activity may be essential for growth in cells, and suggest specific cancer vulnerabilities that could be

targeted therapeutically.

As is the case for PBRM1, other SWI/SNF subunits are involved in maintaining genomic stability.

A 2009 study found BRG1, but not BRM or SNF5, was responsible for maintaining the heterochromatic

state of pericentromeric regions, and that BRG1 deficiency led to the appearance of micronuclei and

aberrant mitoses (Bourgo et al., 2009). BRG1 activity is also required for the decatenation of sister

chromatids after replication through the action of topoisomerase IIalpha (TOP2A) (Dykhuizen et al.,

2013). With BRG1 deficiency or the expression of cancer-associated BRG1 mutants, resulting in less

accessible chromatin, TOP2A has a decreased ability to bind to the genome, resulting in anaphase bridge

formation and G2/M-phase block, eventually leading to an increased percentage of cells with >4n DNA

content.

Clear Cell Renal Cell Carcinoma

It is estimated that ~62,000 new patients in the United States will be diagnosed with kidney

(renal) cancer in 2015 (Society, 2015). Nearly all of these cancers will be renal cell carcinoma, which can

be further classified based on histological appearance. A large majority of renal cell carcinomas (~75%)

will be of the clear cell subtype (ccRCC), which is defined by the appearance of “clear” cytoplasm in cells

typically arranged in nests with interspersed blood vessels (Larkin et al., 2012). Nearly two-thirds of

ccRCC cases are diagnosed when the cancer is still local (Society, 2015). These patients are primarily

treated with surgical resection or nephrectomy, which is generally curative: the 5-year relative survival is

92%. However, in patients with metastatic or recurrent disease, treatment options are limited and

disease-free survival rates are poor. ccRCC is largely resistant to traditional chemotherapy and radiation.

For metastatic cases, approved therapies include cytokine therapy (IFNα and IL-2), anti-mTOR drugs

(temsirolimus and everolimus), and anti-VEGF therapy, including tyrosine kinase inhibitors that target

VEGF-receptors in addition to other kinases (bevacizumab, pazopanib, sorafenib, sunitinib, and axitinib)

Page 28: Dissertation Cover pages v1.0 EDITS 122815

15

(Institute, 2015). While these therapies delay disease progression and some extend overall survival rates

by a few months, very rarely are they curative.

The development of ccRCC has long been associated with inactivating mutations of the Von-

Hippel Lindau (VHL) gene, which normally negatively regulates the activity of Hypoxia-Inducible Factors

(HIF-1α and HIF-2α) (Latif et al., 1993) (Iliopoulos et al., 1995) (Iliopoulos et al., 1996). In normoxic

conditions, VHL, which is an E3-ubiquitin ligase, targets HIFα subunits for degradation (Maxwell et al.,

1999) (Ohh et al., 2000). In hypoxic conditions, or in the absence of VHL, HIFα subunits are stabilized,

and induce a pro-angiogenic transcriptional program. Through the efforts of large-scale cancer

sequencing projects, it is now known that VHL activity is lost through a variety of mechanism (e.g.,

mutation with LOH, deletion, promoter hypermethylation) in >90% of ccRCC tumors (Cancer Genome

Atlas Research, 2013) (Sato et al., 2013). Inactivation of VHL is thus considered to be a “truncal” event

seemingly required for the development of ccRCC (Gerlinger et al., 2012) (Gerlinger et al., 2014).

Previous studies have demonstrated, however, that VHL loss alone is not enough to induce

ccRCC (Ma et al., 2003) (Young et al., 2008). Loss of heterozygosity studies implied the existence of a

“gatekeeper” gene or genes at chromosomal location 3p21 – the genomic location of PBRM1 (van den

Berg et al., 1996) (Clifford et al., 1998). Recent sequencing studies of ccRCC have also implicated

KDM6A, KDM5C, SETD2, and BAP1, all histone modifying enzymes, as being mutated in ~10-15% of

ccRCC, stressing the important role of chromatin modification in this lineage of cancer (Dalgliesh et al.,

2010) (Pena-Llopis et al., 2012). Strikingly, SETD2 and BAP1 are also located at chromosome position

3p21. In ccRCC, one arm of 3p is lost in nearly all cases (91%), leaving cells vulnerable to mutation of

the other allele of these tumor suppressors (VHL is also located nearby at 3p25) (Cancer Genome Atlas

Research, 2013). However, while PBRM1 and SETD2 are often mutated concurrently, BAP1 and

PBRM1 mutations are anticorrelated (Pena-Llopis et al., 2012). Gene expression profiling of BAP1

versus PBRM1 mutant ccRCCs suggests that they regulate different gene expression programs.

Additionally, compared to PBRM1 mutant tumors, BAP1 mutant tumors are of higher grade and worse

survival (Kapur et al., 2013) (Joseph et al., 2015). From these studies, the authors concluded the BAP1

and PBRM1 mutant tumors may be distinct molecular genetic subtypes.

Page 29: Dissertation Cover pages v1.0 EDITS 122815

16

Large-scale genetic profiling studies of ccRCC have also detected mutations in other SWI/SNF

subunits, albeit at much lower rates than PBRM1 mutation (Cancer Genome Atlas Research, 2013) (Sato

et al., 2013). ARID1A and ARID1B were hemizygously deleted in 10% and 20% of tumors, respectively,

in one study (Sato et al., 2013), while both studies detected mutations in SMARCA4 and ARID1A in a low

percentage of tumors. These studies also implicated other pathways as being frequently altered in

ccRCC, including the PI3K-AKT pathway.

The importance of understanding mechanisms of tumor suppression by PBRM1

Cancer sequencing studies have revealed that numerous subunits of the SWI/SNF chromatin

remodeling complex are widely mutated in human malignancies. PBRM1, a defining subunit of the PBAF

type of SWI/SNF complex, is mutated in a variety of cancers, including in ~40% of clear cell renal cell

carcinomas (ccRCC), making it the second most highly mutated gene in ccRCC (behind VHL). Although

many recent studies have looked at how other SWI/SNF components function in cancer control, relatively

little is known about the tumor suppressive mechanisms of PBRM1 in ccRCC. Additionally, there is a

large unmet need for more effective therapies for metastatic ccRCC. In this original research study, we

will investigate how PBRM1 functions as a tumor suppressor in ccRCC. In doing so, we hope to gain

better insight into the molecular mechanisms underlying oncogenic transformation in ccRCC, and

ultimately assist in the discovery of new treatment paradigms.

Page 30: Dissertation Cover pages v1.0 EDITS 122815

17

CHAPTER 2: Characterizing the effects of PBRM1 deficiency on growth and gene

expression

Page 31: Dissertation Cover pages v1.0 EDITS 122815

18

BACKGROUND AND SIGNIFICANCE

Past studies and cancer sequencing efforts have identified PBRM1 as a tumor suppressor that is

recurrently mutated in various cancers, including ccRCC, cholangiocarcinoma, gastric adenocarcinoma,

PDA, and breast cancer. Many of these studies also explored how PBRM1 functions in cancer control.

For example, the studies from the Parsons lab first identifying PBRM1 as a tumor suppressor indicated

that it exerts cell cycle control through induction of p21 (Xia et al., 2008). Restoration of PBRM1

expression in the breast cancer line HCC1143 resulted in G1 arrest induced by PBRM1 binding to the

p21 promoter. In the 2011 ccRCC exome sequencing study, knockdown of PBRM1 in ccRCC cell lines

with wild-type PBRM1 led to increased proliferation, growth in soft agar, and migration (Varela et al.,

2011). Gene set enrichment analysis of microarray data from these knockdown experiments supported

PBRM1’s role in cell cycle control, and also associated it with chromosomal stability. Other ccRCC

genetic profiling efforts have linked PBRM1-mutant tumors to other pathways, including glycoproteins,

and secreted and signaling pathways (Cancer Genome Atlas Research, 2013) (Pena-Llopis et al., 2012).

While these studies have provided valuable insight into some PBRM1 tumor suppressor actions, we

believe that others have yet to be uncovered, particularly for PBRM1 action in ccRCC.

In this chapter, we manipulate the expression of PBRM1 in ccRCC cell lines to create tools to

study its function. We then use various assays to assess how PBRM1 deficiency affects growth. We

hypothesize that because numerous studies have identified PBRM1 as a tumor suppressor, PBRM1

deficiency will increase the growth properties of cells. We also perform gene expression profiling to

identify genes and pathways influenced by PBRM1 that will be explored in later chapters.

Page 32: Dissertation Cover pages v1.0 EDITS 122815

19

RESULTS

PBRM1 protein levels can be stably knocked-down with lentiviral shRNA vectors

As a tool to study PBRM1 function in ccRCC, we created cell lines in which we stably knocked-

down PBRM1 expression levels. 786-O is a VHL-negative ccRCC cell line commonly used in the study of

this disease (Iliopoulos et al., 1995) (Varela et al., 2011). It also contains wild-type PBRM1. We

transduced 786-O cells with two different lentiviral vectors containing shRNA targeting PBRM1 (termed

PBRM1 shRNA #1 and #2), or with a lentiviral vector containing an off-target shRNA as a control (control

or “C” shRNA). PBRM1 shRNAs #1 and #2 reduced PBRM1 mRNA levels 70% and 85%, respectively

(Figure 2.1A). Even greater levels of knockdown were achieved at the protein level: for shRNA #1,

protein levels were reduced ~90%; and for shRNA #2, protein levels were reduced nearly 100% (Figure

2.1B). These levels of knockdown were stable over time.

We performed the same knockdown procedure in the ACHN renal cell carcinoma cell line. The

ACHN cell line has a heterozygous nonsense mutation of PBRM1, but seems to have near-normal levels

of wild-type PBRM1 by Western blot, and PBRM1 siRNA knockdown causes a significant increase in

proliferation (Varela et al., 2011). It has no noted VHL mutations. Although we only achieved ~50% and

70% reductions in mRNA levels using PBRM1 shRNAs #1 and #2 in this cell line, protein levels of

Figure 2.1 – PBRM1 knockdown in 786-O ccRCC cell line. A.) Quantitative reverse-

transcription polymerase chain reaction (qRT-PCR) analysis of PBRM1 mRNA levels

(relative to GAPDH) in 786-O cells transduced with lentiviruses containing an off-target

shRNA (C), or PBRM1-targeting shRNA (#1 and #2). Error bars represent standard

deviations from n=3. The delta-delta-Ct method was used to analyze the data, with

786-O control shRNA set to 1. B.) Western blot analysis of PBRM1 protein levels in

the 786-O lines described in (A).

Page 33: Dissertation Cover pages v1.0 EDITS 122815

20

PBRM1 were reduced dramatically with both hairpins (Figure 2.2A and B). In subsequent experiments,

we will primarily work with the 786-O lines we created, and will use the ACHN cell lines for confirmatory

analysis in another culture model.

Stable expression of wild-type PBRM1 or cancer-associated mutants in a null setting

The A-704 cell line is another ccRCC line (VHL mutant) that contains a homozygous truncating

mutation of PBRM1 (Varela et al., 2011) (see Table 2.1 for a summary of notable mutations in the RCC

cell lines). We transduced these cells with either an empty-vector containing retrovirus (hereafter referred

to as EV) or a retrovirus containing wild-type PBRM1 with a C-terminal V5 tag (referred to as WT) (Figure

2.3A). After the initial selection period, PBRM1 expression was stable over time in the A-704 WT cells –

no PBRM1 expression was detectable in the EV line. We also introduced cancer-associated mutant

versions of PBRM1 into these cells. Examining the spectrum of PBRM1 mutations, the ccRCC exome

sequencing study identified two in-frame deletions, both in the context of 3p LOH: a six-amino acid

deletion (p.M1209_E1214delMFYKKE) in the second BAH domain (6AAD), and a single amino acid

deletion (Ile57) in the first bromodomain (Varela et al., 2011). Additionally, the authors scored the nine

missense mutations identified for likelihood of functional impact, and found that three mutations (p.T232P,

p.A597D and p.H1204P) were likely to have a deleterious effect. We introduced these mutations into our

Figure 2.2 – PBRM1 knockdown in ACHN RCC cell line. A.) qRT-PCR analysis of

PBRM1 mRNA levels (relative to GAPDH) in ACHN cells transduced with lentiviruses

containing an off-target shRNA (C), or PBRM1-targeting shRNA (#1 and #2). Error

bars represent standard deviations from n=3. The delta-delta-Ct method was used to

analyze the data, with ACHN control shRNA set to 1. B.) Western blot analysis of

PBRM1 protein levels in the ACHN lines described in (A).

Page 34: Dissertation Cover pages v1.0 EDITS 122815

21

PBRM1 expression vector (except for the Ile57 deletion mutation, which we were unable to generate),

and transduced the A-704 cells to generate stable expression lines (Figure 2.3B). We were unable to

detect expression of the A597D mutant, possibly indicating that the mutant protein is unstable and is

degraded. Similarly, the 6AAD mutant was expressed at very low levels compared to WT. On the other

hand, the T232P and H1204P mutants expressed at comparable levels to the WT protein. For this

reason, we decided to use these mutant protein lines in future experiments.

To check whether our exogenously expressed, V5-tagged proteins (WT, H1204P, and T232P)

successfully incorporated into PBAF complexes, we performed V5-immunoprecipitation experiments

using nuclear extracts from each cell line (Figure 2.3C). The WT and mutant proteins all bound to BRG1

and SNF5 (core SWI/SNF subunits), and to ARID2 (PBAF-specific), indicating that the exogenously

expressed proteins were incorporated into PBAF complexes. This result further implies that exclusion of

PBRM1 from the PBAF complex is not a tumor-promoting mechanism for these missense mutations.

PBRM1 deficiency results in faster 2-D growth in reduced serum conditions

As PBRM1 is a tumor suppressor, we expected PBRM1 knockdown in the 786-O and ACHN cell

lines to increase growth rates when the cells were grown on tissue culture plates (2-D growth).

Alternatively, when WT PBRM1 was expressed in A-704 cells, we expected the cells to grow slower.

However, at normal culturing conditions using 10% fetal bovine serum (FBS), the growth rates between

the +PBRM1 and –PBRM1 conditions were not dramatically different (Figure 2.4A-C, left column). For

both the 786-O and ACHN lines, knockdown of PBRM1 with shRNA #1 significantly increased growth,

Cell Line Cell Type

PBRM1 status VHL status HIF1α status Other notable

mutations

786-O ccRCC wild-type homozygous

deletion

homozygous deletion (cBio)

ATM (CCLE), TP53 (CCLE), PTEN (CCLE)

ACHN RCC heterozygous

truncating mutation

wild-type (no noted

mutations) wild-type --

A-704 ccRCC homozygous

truncating mutation

misssense mutation

(I180N) (CCLE)

wild-type (although

large chr14 deletion by CNA, cBio)

RARA (CCLE), SMAD2 (CCLE)

Table 2.1 – Summary of notable mutations in the RCC cell lines. CCLE refers to the Cancer Cell Line

Encyclopedia, and cBio to the cBioPortal for Cancer Genomics.

Page 35: Dissertation Cover pages v1.0 EDITS 122815

22

although the gap with the control cells was not very large. shRNA #2 did not increase growth at all, and

in fact led to slightly slower growth in the 786-O cells. In the A-704 cells, expression of WT PBRM1

slowed growth compared to the EV control, but, again, not very dramatically.

We hypothesized that stressing the cells by growing them in reduced serum conditions could

widen the growth gap. When the 786-O cells were grown in 1% serum, both PBRM1 hairpin lines grew

significantly faster than control cells (Figure 2.4A, right panel). The same was true for the ACHN lines

grown in 6% serum (Figure 2.4B, right panel). Likewise, when the A-704 cells were grown in 6% serum,

the growth difference between the EV and WT cells widened dramatically (Figure 2.4C, right panel). The

Figure 2.3 – Stable expression of wild-type PBRM1 or cancer-associated mutants in

A-704. A.) The PBRM1 null cell line A-704 was transduced with either empty-vector

containing retrovirus (EV), or a retrovirus containing wild-type PBRM1 with a C-terminal V5

tag (WT). After selection, expression of PBRM1 was checked via Western blot analysis

using an antibody against PBRM1. B.) A-704 cells were also transduced with various

retroviruses containing cancer-associated mutant PBRM1. Expression was checked via

Western blot analysis. C.) V5-immunoprecipitation experiments were performed using

nuclear extracts from the indicated A-704 cell lines created in (A) and (B). Western blot

analysis was performed to check for binding to SNF5, ARID2, and BRG1.

Page 36: Dissertation Cover pages v1.0 EDITS 122815

23

growth differences that emerge at reduced serum conditions largely seem to be the result of the cells

containing PBRM1 losing their ability to proliferate.

We also tested the growth effects of expressing the H1204P and T232P mutants in A-704 cells

and saw similar results. At full serum, the growth differences between the WT and cancer-mutant lines

Figure 2.4 – 2-D Growth Curves of RCC cell lines at normal and reduced serum

conditions. A.) 2-D growth curves of the indicated 786-O cell lines grown on 48-well

tissue culture plates at normal (10%) and reduced (1%) FBS. n=6 per timepoint for each

line. B.) Growth curves of the ACHN lines at normal (10%) and reduced (6%) FBS. n=4-6

per timepoint for each line. C.) Growth curves of the A-704 lines at normal (10%) and

reduced (6%) FBS. n=3 per timepoint for each line. Two-way ANOVAs using Tukey’s

multiple comparisons test was performed on each set of growth curves. * P<0.05; **

P<0.01; *** P <0.001; **** P<0.0001.

Page 37: Dissertation Cover pages v1.0 EDITS 122815

24

were minimal – at later timepoints, the T232P mutant line grew slightly slower than the WT line, and all

lines grew slower than the EV control line (Figure 2.5A). At reduced serum conditions (6%), a growth gap

emerged, in which all lines grew significantly faster than the WT line (Figure 2.5B). However, compared

to the EV control line, the cancer-mutant lines still grew significantly slower, suggesting that the cancer

mutants may be behaving in a hypomorphic manner.

PBRM1 knockdown increases the 3-D growth capacity of cells

When the RCC cell lines were placed under more stressful conditions by growing them at

reduced serum, a significant growth advantage conveyed by PBRM1 deficiency emerged. We reasoned

that a similar situation might occur using 3-D growth assays, which more stringently test the transformed

nature of cells. First, we compared the ability of our cell lines to form colonies in soft agar, a measure of

anchorage independent growth and tumorigenic potential. In both 786-O and ACHN cells, PBRM1

knockdown with either hairpin significantly increased colony forming capacity (Figure 2.6). The A-704 cell

lines were unable to form colonies in soft agar.

As a further test of the 3-D growth capacity of PBRM1 knockdown cells, we performed tumor

xenograft experiments with 786-O control shRNA and PBRM1 shRNA #2 cells. One million cells of each

line were subcutaneously injected into the ventral surface of nude mice. After an initial latency period, the

Figure 2.5 – 2-D Growth Curves of all A-704 lines at normal and reduced serum

conditions. 2-D growth curves as in Figure 2.4, except growth data was collected using an

Incucyte Zoom machine as a % confluence of a well on a tissue culture plate. A.) 2-D growth

curves of the indicated A-704 cell lines grown on 48-well tissue culture plates at normal (10%)

and B.) reduced (6%) FBS. n=3 per timepoint for each line. Two-way ANOVAs using Tukey’s

multiple comparisons test was performed on each set of growth curves, and significant

differences with the WT line are shown. * P<0.05; ** P<0.01; *** P <0.001; **** P<0.0001.

Page 38: Dissertation Cover pages v1.0 EDITS 122815

25

PBRM1 knockdown cells grew significantly faster than the control cells (Figure 2.7A), and had formed

significantly larger tumors by the time the mice were euthanized (Figure 2.7B,D). Protein extracted from

the tumors and analyzed by Western blot analysis confirmed PBRM1 knockdown persisted in the PBRM1

shRNA line (Figure 2.7C).

Murine embryonic fibroblasts with conditional loss of Pbrm1 grow slower than matched controls

Murine embryonic fibroblasts (MEFs) are typically used to study the effects of a particular genetic

event, such as gene loss, in a “clean” genetic background compared to tumor cell lines, which often

contain a plethora of additional mutations. To this end, we isolated MEFs from conditional Pbrm1 mice

we had generated in which LoxP sites were engineered to flank exon 2 of the Pbrm1 allele, such that Cre-

mediated recombination would result in a frame-shifting mutation. Using three sets of MEFs from

different embryos, we infected either with adenovirus expressing Cre-recombinase (NULL cells) or GFP

as a control (WT cells). Western blots analysis indicated effective recombination and loss of Pbrm1

expression in the NULL cells (Figure 2.8A). Growth curve analysis, however, revealed that the Pbrm1

NULL MEFs grew significantly slower than the WT controls (Figure 2.8B). This was not totally surprising,

as tumor suppressor loss-induced senescence has been observed for other tumor suppressors in MEFs,

including for VHL (Young et al., 2008).

Figure 2.6 – PBRM1 knockdown in 786-O and ACHN cells increases

colony formation in soft agar. Single cell suspensions of 786-O (A) and

ACHN (B) were plated with soft agar and colony formation was measured

after 23 days. Error bars represent standard errors of the mean (SEM),

n=3/line; statistical testing was performed using unpaired t-tests against the

control shRNA.

Page 39: Dissertation Cover pages v1.0 EDITS 122815

26

RNA-seq and microarray analysis reveal cancer-related pathways perturbed with PBRM1

deficiency

RNA-seq analysis of 786-O and A-704 cell lines

Having created various cell culture systems of PBRM1 alteration, we next asked what genes and

transcriptional programs were influenced by PBRM1. To answer this question using the 786-O and A-704

cell lines, we employed RNA-seq, a technique which uses next-generation sequencing to quantify mRNA

transcript levels with wide coverage of the transcriptome and high dynamic range. Additionally, because

we saw larger growth differences emerge at reduced serum conditions, we decided to quantify mRNA

levels in cells growing at normal and reduced serum conditions. mRNA from the 786-O control shRNA

Figure 2.7 – PBRM1 knockdown in 786-O cells significantly increases tumor

xenograft growth. One million cells of 786-O control shRNA or PBRM1 shRNA #2 cells

were subcutaneously injected into the ventral surface of athymic nude mice (n=4). A.)

Tumors were palpable after 10 days, and tumor growth was then tracked until tumors

exceeded 1cm3, at which time the mice were euthanized. B.) Excised tumor xenografts of

indicated lines after mice euthanasia on day 34 post-injection – excised tumor weights are

shown in (D). C.) Western blot analysis of protein extracted from tumor xenografts. For the

growth curve in (A), two-way ANOVA using Tukey’s multiple comparisons test was

performed. For the tumor weight comparison in (D), an unpaired t-test was performed. *

P<0.05; ** P<0.01; *** P <0.001; **** P<0.0001.

Page 40: Dissertation Cover pages v1.0 EDITS 122815

27

and PBRM1 shRNA #1 and #2 lines and the A-704 EV and WT lines was isolated, RNA-seq libraries

were prepared, and 100 base-pair (bp) single-end sequencing was performed using an Illumina HiSeq

machine. The depth of coverage for each sample is shown in Table 2.2.

Sample Name FBS

%

# of Reads

(x106)

786-O control shRNA 10 45.0

786-O PBRM1 shRNA #1 10 27.5

786-O PBRM1 shRNA #2 10 36.4

786-O control shRNA 1 41.1

786-O PBRM1 shRNA #1 1 41.5

786-O PBRM1 shRNA #2 1 36.0

A-704 EV 10 28.5

A-704 WT 10 39.1

A-704 EV 6 31.6

A-704 WT 6 46.4

Next, we processed the RNA-seq raw data using the Tuxedo suite of tools and aligned our reads

using Cufflinks. Cuffdiff was used for expression comparisons between samples (e.g., A-704 EV vs. WT

at 6% serum), generating RPKM, fold-change, and p-values. Before proceeding to the next stage of

analysis, however, we wanted to filter out genes with very low RPKM values regardless of PBRM1 status,

Figure 2.8 – MEFs with conditional loss of Pbrm1 grow slower than matched control cells.

Three sets of Pbrm1 fl/fl MEFs were infected with a GFP or Cre-expressing adenovirus to

generate Pbrm1 WT and NULL MEFs, respectively. A.) Western blot analysis showing loss of

Pbrm1 expression in the NULL MEFs. B.) Growth curve analysis of MEF sets from part (A).

n=3/line at each timepoint, error bars represent SEM. Two-way ANOVA using Tukey’s multiple

comparisons test was performed on the growth curves. * P<0.05; ** P<0.01; *** P <0.001; ****

P<0.0001.

Table 2.2 – RNA-seq depth of coverage.

Page 41: Dissertation Cover pages v1.0 EDITS 122815

28

reasoning that these genes were not biologically relevant in this context and could significantly alter

downstream analysis, such as gene set enrichment analysis (GSEA). To find the proper RPKM cutoff, we

generated histograms of cumulative RPKM (summing the +PBRM1 and –PBRM1 conditions, and using

an average RPKM value for the PBRM1 shRNAs) for each cell line at each serum concentration (Figure

2.9). In all settings, ~80% of the mapped genes had a cumulative RPKM less than or equal to 10. We

excluded these low abundance transcripts and focused our analysis on the more highly expressed genes,

now termed the “expressed” genes (9,744 genes for 786-O normal serum; 9,846 genes for 786-O

reduced serum; 9,664 genes for A-704 normal serum; 9,798 genes for A-704 low serum). After filtering,

histograms of the fold-change values (-PBRM1 condition/+PBRM1 condition) were bell-curve shaped

(Figure 2.10).

To more accurately identify genes and pathways altered by PBRM1, and account for cell line-

specific effects, we wanted to limit our analysis to genes that were expressed in both 786-O and A-704

cells at a given serum concentration, and whose expression changed in the same direction with PBRM1

deficiency (i.e., same directionality of fold-change with both PBRM1 shRNAs in 786-O and A-704 EV

compared to A-704 WT). After filtering on these criteria, we were left with the following gene counts for

further analysis:

Gene # (direction change with PBRM1

deficiency)

Serum Condition UP DOWN TOTAL

Normal 2,777 1,542 4,319

Reduced 1,519 2,017 3,536

At normal serum, more genes had increased expression than decreased with PBRM1 deficiency,

indicating that PBRM1 was playing more of a repressive role under these conditions. However, at

reduced serum, the reverse was true, indicating that PBRM1 was playing more of a transcriptional

activator role under stressful conditions.

To quantify genes whose expression was most impacted by PBRM1 across cell lines, we then

performed a Fisher’s combined probability test at each serum condition. This method allows one to pool

the p-values from independent experiments and perform a meta-analysis, generating a “meta-p-value” for

Table 2.3 – Gene numbers after filtering.

Page 42: Dissertation Cover pages v1.0 EDITS 122815

29

each gene. We used the p-values generated from Cuffdiff comparing gene expression in the following

sample pairs: 786-O PBRM1 shRNA #1/control shRNA; 786-O PBRM1 shRNA #2/control shRNA; A-704

EV/WT. We will also analyze the RNA-seq data without combining later on.

Top-changed genes and pathways with PBRM1 alteration

With a single meta-p-value now associated with each gene, we next looked at the top genes with

expression changes. Figure 2.11 shows heatmaps of the log2(fold-change) for genes with meta-p-

values<0.01 at each serum condition. Numerous genes are among the top-changed genes at both serum

conditions: AIF1L, COL6A1, CLDN2, ID1, ALDH1A1, GPR56, AMACR, CLU, IL8, SERPINE1,

SNORD118, MXD1, UAP1, CTSS, and IFI27 appear in both heatmaps. We were particularly interested in

genes whose alteration would confer a growth advantage under stressful conditions, such as reduced

serum or a 3-D growth environment. With this in mind, we took special note of ALDH1A1 due to its

association with cancer-initiating cells (Pearce et al., 2005) (Ginestier et al., 2007) (Chen et al., 2009)

(van den Hoogen et al., 2010) (Su et al., 2010). Other notable genes that only appear on the normal

serum heatmap include IGFBP3 (a HIF target gene), HIF1A itself, CA2 (carbonic anhydrase II, a known

connection to ccRCC), EGFL7 (modulates vasculogenesis and Notch signaling), JAG1 (jagged1, a Notch

receptor ligand), and PMAIP1 (also known as NOXA, a pro-apoptotic factor).

To identify pathways and transcriptional programs altered by PBRM1, we then performed gene

set enrichment analysis (GSEA) using the meta-p-values associated with each gene and the pre-ranked

gene list analysis option. Numerous pathways were highly enriched at both serum conditions – the top 15

enriched gene sets for genes that are upregulated or downregulated with PBRM1 deficiency are shown in

Figure 2.12. At normal serum, upregulated genes were enriched for numerous gene sets related to

translational regulation (KEGG RIBOSOME, REACTOME PEPTIDE CHAIN ELONGATION, REACTOME

3 UTR MEDIATED TRANSLATIONAL, and REACTOME SRP DEPENDENT COTRANSLATIONAL

PROTEIN TARGETING TO MEMBRANE). Gene sets related to the classical subtype of glioblastoma

multiforme, defined by EGFR abnormalities, and retinoic acid signaling were also enriched (VERHAAK

GLIOBLASTOMA CLASSICAL and MARTENS TRETINOIN RESPONSE UP, respectively). At reduced

serum (Figure 2.12B), notable gene sets enriched among upregulated genes included those related to

Page 43: Dissertation Cover pages v1.0 EDITS 122815

30

Fig

ure

2.9

– H

isto

gra

ms

of

cu

mu

lati

ve

RP

KM

va

lue

fo

r d

iffe

ren

t s

ets

of

sa

mp

les

. C

um

ula

tive

RP

KM

va

lue

s w

ere

ca

lcu

late

d fro

m t

he

RN

A-s

eq

Cuff

diff o

utp

ut

by s

um

min

g t

he R

PK

M v

alu

es f

rom

th

e –

PB

RM

1 a

nd

+P

BR

M1

co

nditio

ns, usin

g

an

ave

rage

RP

KM

va

lue

fo

r th

e P

BR

M1

sh

RN

As in

78

6-O

(A

).

Bin

s o

f 1

0 w

ere

use

d to

gene

rate

th

e h

isto

gra

ms. T

he

dash

ed

line

sh

ow

s t

he

le

vel b

elo

w w

hic

h g

en

es w

ere

filt

ere

d f

rom

fu

rth

er

ana

lysis

.

Page 44: Dissertation Cover pages v1.0 EDITS 122815

31

Fig

ure

2.1

0 –

His

tog

ram

s o

f lo

g2 f

old

-ch

an

ge

in

RP

KM

aft

er

filt

eri

ng

lo

w-e

xp

res

se

d g

en

es

.

Fo

r 7

86

-O (

A)

an

d A

-70

4 (

B)

at

ea

ch s

eru

m c

once

ntr

atio

n, h

isto

gra

ms o

f th

e lo

g2 f

old

-ch

ang

e in

RP

KM

we

re g

en

era

ted

, a

fte

r filte

ring

out

low

-exp

resse

d g

en

es a

s

de

scri

bed

in

Fig

ure

2.9

. A

n a

ve

rag

e R

PK

M v

alu

e f

or

the

PB

RM

1 s

hR

NA

s in 7

86

-O w

as u

se

d.

Bin

s o

f 0

.1 w

ere

used

. T

he d

ash

ed

lines r

epre

se

nt

lo

g2(f

old

-cha

ng

es)

of-

0.4

and

0.4

, w

hic

h w

ill b

e u

se

d to

id

en

tify

diffe

rentia

lly e

xp

ressed

gen

es in

la

ter

an

aly

sis

.

Page 45: Dissertation Cover pages v1.0 EDITS 122815

32

Figure 2.11 – Heatmaps of the top changed genes with PBRM1 alteration at

each serum condition. Heatmaps of the log2(fold-change) of the genes with meta-

p-value<0.01 at each serum condition. The control conditions (786-O control

shRNA and A-704 EV) were set to 0.

Page 46: Dissertation Cover pages v1.0 EDITS 122815

33

Fig

ure

2.1

2 –

Ge

ne

se

t e

nri

ch

me

nt

an

aly

sis

of

RN

A-s

eq

me

ta-a

na

lys

is i

de

nti

fies

pa

thw

ays

sig

nif

ica

ntl

y a

lte

red

by P

BR

M1

. T

o id

en

tify

pa

thw

ays a

nd

tra

nscrip

tion

al pro

gra

ms a

lte

red

by P

BR

M1

, w

e p

erf

orm

ed

gen

e s

et

en

rich

me

nt

ana

lysis

(G

SE

A)

usin

g t

he m

eta

-p-v

alu

es

asso

cia

ted w

ith

ea

ch

gen

e a

nd

th

e p

re-r

an

ked

gen

e lis

t a

naly

sis

op

tion

(th

e in

ve

rse

of

the m

eta

-p-v

alu

e m

ultip

lied

by t

he d

ire

ctio

n o

f fo

ld-c

han

ge

wa

s u

se

d to

gen

era

te a

n in

tegra

ted

ra

nke

d lis

t o

f up

regu

late

d a

nd

do

wn

reg

ula

ted g

ene

s).

T

he

to

p 1

5 e

nriche

d g

en

e s

ets

am

on

g u

pre

gula

ted

(UP

) o

r d

ow

nre

gu

late

d (

DO

WN

) g

en

es a

t e

ach

se

rum

con

ditio

n a

re s

ho

wn

, a

lon

g w

ith

th

e n

orm

aliz

ed

en

richm

en

t sco

re (

NE

S),

no

min

al p

-va

lue

(NO

M p

-va

l),

an

d t

he

fals

e d

isco

ve

ry r

ate

q-v

alu

e (

FD

R q

-va

l).

Page 47: Dissertation Cover pages v1.0 EDITS 122815

34

EGFR (KOBAYAHSI EGFR SIGNALING 24HR UP and ZWANG TRANSIENTLY UP BY 1ST EGF PULSE

ONLY) and Wilm’s tumor (LI WILMS TUMOR VS FETAL KIDNEY 1 UP). In the EGFR-related genes

sets, some of the top upregulated genes include VAV3, CDK6, PDK2, and FGFR4. Among

downregulated genes, many enriched gene sets related to interferon signaling (HECKER IFNB1

TARGETS, SAMA RESPONSE TO IFNG UP, MOSERLE IFNA RESPONSE, BROWNE INTERFERON

RESPONSIVE GENES). EZH2-regulated genes also enriched as being downregulated (NUYTTEN EZH2

TARGETS UP).

As an alternative method to identify enriched pathways, we also performed gene ontology

analysis using the gene ontology biological processes (GOBP) database. We checked for enriched

processes among genes with meta-p-values<0.05 at each serum condition (Figure 2.13). At normal

serum, only six processes were significantly enriched and represented broad categories such as “singe

organism cellular process” and “small molecule metabolic process.” At reduced serum, however,

numerous processes were significantly enriched – the top 20 are shown in Figure 2.13B. Many of these

processes related to interferon signaling, defense response, and cytokine signaling, similar to the GSEA

output. A cell surface receptor signaling pathway was also enriched.

Microarray analysis of Pbrm1 conditional MEFs

To see what genes and pathways were altered in the Pbrm1 NULL MEFs, we used gene

expression microarrays to perform expression analysis on the three matched sets of MEFs described in

Figure 2.8. Aldh1a1 and Igfbp3 were among the top 50 most-altered genes in the MEFs (Figure 2.14A);

these genes were also found among the top-changed genes in the ccRCC RNA-seq analysis. GSEA of

the microarray data did not reveal any enriched pathways with low false-discovery rates (FDR q-

value<0.25) (Figure 2.14B). However, among the top-enriched pathways in the NULL MEFs, we saw

retinoic acid-related pathways (DELACROIX RAR TARGETS UP and DELACROIX RAR TARGETS

DOWN), and among the enriched pathways in the WT MEFs, we saw pathways related to epigenetic

regulation of gene expression (WANG LSD1 TARGETS UP and KAMMINGA EZH2 TARGETS). We also

performed gene ontology analysis, and saw modest enrichment of 5 pathways, including “immune system

process” (Figure 2.14C).

Page 48: Dissertation Cover pages v1.0 EDITS 122815

35

Fig

ure

2.1

3 –

Ge

ne

on

tolo

gy a

na

lys

is o

f R

NA

-se

q m

eta

-an

aly

sis

id

en

tifi

es

bio

log

ica

l p

roc

es

se

s s

ign

ific

an

tly a

lte

red

by P

BR

M1

. W

e

pe

rfo

rme

d g

en

e o

nto

log

y a

na

lysis

usin

g t

he

Gen

e O

nto

log

y B

iolo

gic

al P

rocesses d

ata

ba

se o

n g

en

es w

ith

me

ta-p

-va

lue

<0

.05

at

each

se

rum

co

nditio

n.

Fo

r n

orm

al se

rum

(A

), a

ll sig

nific

an

tly e

nrich

ed

path

wa

ys a

re s

ho

wn

(B

on

ferr

oni co

rrecte

d p

-va

lue <

0.0

5).

F

or

red

uce

d s

eru

m (

B),

th

e

top

20 e

nriche

d p

ath

wa

ys a

re s

ho

wn

.

Page 49: Dissertation Cover pages v1.0 EDITS 122815

36

Figure 2.14 – Significantly altered genes and pathways in Pbrm1 NULL MEFs. Microarray gene

expression analysis was performed on three sets of matched Pbrm1 conditional MEFs. A.) Heatmap of

the top 50 altered genes in the MEFs. B.) GSEA of the MEF microarray data. The top 15 pathways

enriched in the NULL MEFs are shown in the top panel, and the 14 pathways with P<0.05 enriched in the

WT MEFs are shown in the bottom panel. C.) Gene ontology analysis of MEF microarray data (P<0.05).

Page 50: Dissertation Cover pages v1.0 EDITS 122815

37

Overlap of genes with significant expression changes in 786-O, A-704, and MEFs

As an alternative expression analysis method to those prevented above, we identified genes

whose expression changed significantly in each individual cell line and then looked for any overlaps

between these genes. For the 786-O and A-704 cell lines at normal serum (similar condition to the

MEFs), we used a log2(fold-change) cut-off of 0.4 to call a gene as significantly altered, as shown by the

dotted lines in Figure 2.10. For the MEFs, we used a fold-change cut-off of 1.1 (due to the lower dynamic

range of microarray analysis). For the 786-O, A-704, and MEFs, 2,196, 1,394, and 433 genes met these

thresholds, respectively. Among these genes, there was significant overlap between the 786-O and A-

704 lines (355 genes), and less between these lines and the MEFs (23 and 15 genes), as might be

expected when comparing human cancer cell lines to non-transformed, primary mouse cells (Figure

2.15). Still, ten genes were found to be significantly altered in all settings: IGFBP3, ALDH1A1, and

EFNA3 were upregulated in all 3 cell lines; WBP5, ESCO2, and PBRM1 were downregulated; and

NIPAL2, DNAJB14, IL6, and CD47 changed in different directions.

Analysis of primary tumor datasets reveals that ALDH1A1 is overexpressed in PBRM1 mutant tumors

We next asked whether any expression changes could be seen in primary ccRCC tumors for the

six genes (IGFBP3, ALDH1A1, EFNA3, WBP5, ESCO2, and PBRM1) that were consistently upregulated

Figure 2.15 - Overlap of genes with significant expression changes in 786-O, A-704, and MEFs.

For 786-O, A-704, and MEFs, genes whose expression changed significantly with PBRM1 deficiency

were identified using the following cut-offs: log2(fold-change) > 0.4 for 786-O and A-704; and fold-

change > 1.1 for the MEFs. Gene overlap analysis was then performed. The ten genes changed in

all three lines are called-out and their fold-changes are listed.

Page 51: Dissertation Cover pages v1.0 EDITS 122815

38

or downregulated with PBRM1 deficiency in our cell culture systems. To answer this question, we used

two publicly available ccRCC primary tumor expression datasets: one from the TCGA available on the

cBioPortal for Cancer Genomics; and the other from the 2012 study from Peña-Llopis et al that profiled

BAP1 and PBRM1 mutant tumors/tumorgrafts (Pena-Llopis et al., 2012). We divided the tumors into

those with PBRM1 mutations (Mut) and those without (WT). In the TCGA dataset, 122 out of 417

sequenced tumors had PBRM1 mutations; in the Peña-Llopis et al dataset, 21 out of 53

tumors/tumorgrafts had PBRM1 mutations. In the TCGA dataset, 5/6 of the identified genes were

significantly altered in the PBRM1 mutant setting (Figure 2.16A). The changes in two of the genes

(EFNA3 and ESCO2) barely met significance (P=0.045 and P=0.047, respectively). ALDH1A1, WBP5,

and PBRM1, however, all had dramatic shifts in expression. The shift in PBRM1 expression suggests

that many of the mutations produced unstable transcripts that may undergo nonsense mediated decay.

In the Peña-Llopis et al dataset, only ALDH1A1 was among the significant genes that

distinguished PBRM1 mutant vs. non-mutant tumors. In fact, in this dataset, ALDH1A1 was even more

dramatically upregulated in the PBRM1 mutant tumors (Figure 2.16B). To see if this upregulation was

specific to PBRM1 mutant tumors, we divided the TCGA tumors by SETD2 and BAP1 mutation status and

saw no correlation with ALDH1A1 expression levels (Figure 2.16C). Based on this analysis, we

concluded that our identification of ALDH1A1 as a PBRM1-regulated gene also applied to primary ccRCC

tumors.

DISCUSSION

Like many other SWI/SNF subunits, PBRM1 is recurrently mutated in numerous cancers,

although it has a particularly high mutation rate in ccRCC (Varela et al., 2011) (Cancer Genome Atlas

Research, 2013). While previous studies have investigated PBRM1’s tumor suppressor functions, and

implicated PBRM1 in cell cycle control and chromosomal stability (Xia et al., 2008) (Varela et al., 2011),

we believe key genes and pathways under PBRM1 control have yet to be identified. Therefore, in this

chapter, we set out to create cell culture tools to study PBRM1 function in ccRCC. After characterizing

how PBRM1 deficiency impacted growth in our systems, we then performed gene expression profiling.

Page 52: Dissertation Cover pages v1.0 EDITS 122815

39

In the RCC cell lines 786-O and ACHN, stable knockdown of PBRM1 dramatically increased 2-D

growth in tissue culture plates, but only at reduced serum conditions (Figure 2.4A,B). In the A-704

ccRCC line, which possesses a homozygous truncating mutation of PBRM1, stable expression of wild-

type PBRM1 substantially reduced growth, but again only at reduced serum (Figure 2.4C). These

findings suggest that the growth advantages conveyed by PBRM1 loss may only emerge under stressful

conditions, such as those encountered by a nascent tumor. This conclusion is further supported by the

robust 3-D growth advantage of PBRM1 knockdown cells, as seen by colony formation in soft agar

Figure 2.16 – ALDH1A1 is upregulated in PBRM1 mutant tumors. A.) Provisional TCGA data for ccRCC

was taken from the cBioPortal for Cancer Genomics in November 2015 – 417 tumor samples had

sequencing and expression data available. We divided the tumors into those with PBRM1 mutations (Mut)

and those without (WT) – 122/417 tumors were mutated. mRNA z-score levels are shown for the 6 listed

genes. B.) We used ccRCC expression data from Peña-Llopis et al to see if any of the genes from (A) was

among the significant genes that distinguished PBRM1 mutant vs. non-mutant tumors – only ALDH1A1 was

differentially expressed (21/53 tumors had PBRM1 mutations). Relative mRNA levels are shown. C.) The

TCGA data from (A) was divided based on SETD2 and BAP1 mutation status – 52/417 tumors had SETD2

mutations, and 36/417 had BAP1 mutations. ALDH1A1 mRNA z-score levels are shown. All plots

represent Tukey plots with the outliers not shown. Statistical testing was performed using Mann-Whitney

tests.

Page 53: Dissertation Cover pages v1.0 EDITS 122815

40

(Figure 2.6). Additionally, tumor xenograft studies using the 786-O line revealed that PBRM1 knockdown

cells grow significantly faster and form significantly larger tumors than control cells in vivo (Figure 2.7).

Together, these data suggest that PBRM1 deficiency helps a cell adjust to stressful environments,

whether it be reduced serum, anchorage independence, or the subcutaneous compartment of a nude

mouse, and continue to grow. Investigating genes and pathways influenced by PBRM1 that may

contribute to this growth advantage will be the subject of the next chapter.

In the A-704 line, stable expression of cancer-associated mutant forms of PBRM1 (H1204P and

T232P) resulted in faster growth than expressing WT PBRM1, but still slowed growth compared to the EV

control line (Figure 2.5). Although the large majority of PBRM1 mutations in primary tumors are

nonsense, truncating mutations, which are expected to be true loss of function mutations, missense

mutations in the context of LOH do occur. This growth finding suggests that at least some of these

mutations may function as hypomorphs, in which some functions of PBRM1 may be lost while others

remain intact. Further exploring these mutants and their effects compared to WT PBRM1 may aid in the

discovery of structure:function relationships, and will be done in experiments in later chapters.

Additionally, some of the PBRM1 mutants could not be stably expressed, such as the A597D

mutant, or could only be expressed at very low levels, such as the 6AAD mutant. While this could be due

to instability of the produced protein, it could also result from failure to incorporate into the PBAF complex,

eventually leading to protein degradation. We also did not explore if other types of PBRM1 mutations,

such as some of the truncating mutations, lead to stable expression of PBRM1 protein fragments, and if

so, how these proteins affect PBAF assembly. As truncating mutations are much more prevalent than

missense mutations, this would be an interesting avenue for further exploration.

Due to the widening growth gap at reduced serum conditions, we decided to perform RNA-seq

expression profiling on the 786-O and A-704 lines grown at both normal and reduced serum conditions.

We believed that doing so would give us a better chance of identifying key growth and cancer-related

pathways under PBRM1 regulation. We also performed Fisher’s combined probability test to conduct a

meta-analysis in which we could combine the RNA-seq results from the two separate cell lines, ones in

which we approached PBRM1 alteration from opposing directions – knocking down in the WT setting

(786-O), or restoring in the mutant setting (A-704). We hoped that this approach would allow us to

Page 54: Dissertation Cover pages v1.0 EDITS 122815

41

account for cell-line specific effects and biases related to using shRNA vectors or over-expressing a gene

of interest.

After processing the RNA-seq data in this fashion, we performed GSEA and gene ontology

analysis to identify enriched pathways and biological processes (Figure 2.12 and 2.13). Although

numerous pathways and processes were altered, a few themes emerged from this analysis. First, at

normal serum, among upregulated genes with PBRM1 deficiency, numerous pathways enriched related

to translational regulation. Although we don’t follow up on this finding in this dissertation, future studies

could investigate PBRM1’s role in translational control, as this would be expected to have wide-ranging

effects on a cell. Second, at both normal and reduced serum, among upregulated genes, pathways

enriched related to the EGFR pathway. Given the EGFR pathway’s well-known roles in growth and

cancer development, and the genetic antagonism between the pbap complex and the EGFR pathway

previously found in flies, we will investigate this pathway further in the next chapter (Rendina et al., 2010)

(Curtis et al., 2011). Lastly, at reduced serum conditions, among downregulated genes, numerous

enriched GSEA pathways and GO processes related to interferon signaling. One of the top

downregulated genes at both serum conditions was IFI27, or Interferon, Alpha-Inducible Protein 27, a

supposed pro-apoptotic interferon-response gene (Figure 2.11). We will also investigate this pathway in

relation to PBRM1 in Chapter 3.

We also conducted expression profiling in our conditional Pbrm1 MEFs, a non-transformed,

primary mouse line (Figure 2.14). We hypothesized that any genes whose expression changed across all

three of these systems (786-O, A-704, and MEFs) might be true PBRM1-regulated genes. Indeed, we

saw ten genes regulated across all systems, including six that changed in the same direction with PBRM1

deficiency (Figure 2.15). PBRM1’s presence on this list as a consistently downregulated gene served as

a nice control for this analysis method. Finally, we used two publicly available ccRCC datasets to see if

any of these genes were differentially expressed in primary tumors with PBRM1 mutations. We found

that ALDH1A1 was significantly upregulated in PBRM1 mutant tumors in both datasets (Figure 2.16). In

the next chapter, we intend to further explore PBRM1 regulation of ALDH1A1 and its role in tumor control.

Page 55: Dissertation Cover pages v1.0 EDITS 122815

42

CHAPTER 3: PBRM1 suppresses tumorigenic potential through regulation of

ALDH1A1 expression, and the EGFR and IFN pathways

Page 56: Dissertation Cover pages v1.0 EDITS 122815

43

BACKGROUND AND SIGNIFICANCE

In Chapter 2, we created cell culture tools to study PBRM1 function in ccRCC, and subsequently

conducted gene expression profiling using these tools. We analyzed the expression data to identify the

top-changed genes and enriched pathways and processes. These efforts yielded some interesting

results. Across our cell line systems, a few genes were consistently upregulated or downregulated with

PBRM1 deficiency (IGFBP3, ALDH1A1, EFNA3, WBP5, ESCO2, and PBRM1). Previously, PBRM1 had

not been connected to the regulation of any of these genes. We then asked whether any of these genes

were differentially expressed in primary tumors, and found that ALDH1A1 expression was increased in

PBRM1 mutant tumors.

ALDH1A1 is also known as aldehyde dehydrogenase 1 or retinaldehyde dehydrogenase 1. It is

one of a class of oxidizing enzymes that convert aldehydes to carboxylic acids (Xu et al., 2015). It is part

of the retinoic acid metabolic pathway and irreversibly converts retinaldehyde to retinoic acid (ALDH1A2

and ALDH1A3 can also perform this step). Aldehyde dehydrogenases (ALDHs) also perform other

functions in a cell, such as protecting against a variety of environmental stressors by serving as aldehyde

scavengers (Singh et al., 2013). For example, ALDHs are thought to protect against oxidative stress and

UV damage and to help catalyze the breakdown of lipid peroxides.

ALDH1A1 has known roles in hematopoetic stem cell development (Ghiaur et al., 2013) (Chanda

et al., 2013), white vs. brown fat programming (Kiefer et al., 2012b), and insulin signaling (Kiefer et al.,

2012a). It has also been identified as a marker of tumor-initiating cells or cancer-stem cells in a variety of

cancer types (Pearce et al., 2005) (Ginestier et al., 2007) (Chen et al., 2009) (van den Hoogen et al.,

2010) (Su et al., 2010). The cancer stem cell theory holds that a small population of cells within the

cancer actually drive the cancer, and that these cells possess unique properties, related to such things as

self-renewal capacity, multipotency, resistance to therapeutics, and ability to seed new tumors (Nguyen et

al., 2012). This association between ALDH1A1 and cancer stem cells seems paradoxical at first glance,

as retinoic acid signaling is traditionally associated with a more differentiated state. Various mechanisms,

however, have been proposed to explain ALDH1A1’s association with increased tumorigenicity. For

example, increased retinoic acid formed from higher ALDH1A1 activity could participate in non-classical

retinoic acid signaling, whereby PPARβ/δ activation has anti-apoptotic, pro-growth effects (Schug et al.,

Page 57: Dissertation Cover pages v1.0 EDITS 122815

44

2007). ALDH1A1’s cancer-promoting effects could also be mediated through non-retinoic acid related

mechanisms, such as protection from reactive oxygen species or limiting the toxicity of drugs (Singh et

al., 2013) (Huang et al., 2013) (Corominas-Faja et al., 2013) (Raha et al., 2014). In this chapter, we will

explore PBRM1 regulation of ALDH1A1 and how it impacts tumorigenic potential.

We also identified the EGFR and IFN pathways as being potentially affected by PBRM1 status.

Both these pathways not only have relevance to cancer, but have been studied before in relation to

PBRM1-containing complexes (Rendina et al., 2010) (Curtis et al., 2011) (Yan et al., 2005). The EGFR

pathway is a widely studied growth pathway that is frequently altered in cancer (Chong and Janne, 2013).

Its relevance to cancer is evident by the numerous EGFR inhibitors and blocking antibodies that are

clinically approved for the treatment of various cancer types. Although EGFR is not amplified in ccRCC, it

is frequently overexpressed, and the downstream PI3K pathway is recurrently activated through low

mutation rates in various other components (Cancer Genome Atlas Research, 2013) (Sato et al., 2013).

EGFR inhibition in metastatic ccRCC, however, has largely yielded disappointing clinical results, with little

or no benefit over cytokine therapy (e.g., IFNα) (Drucker et al., 2003) (Ravaud et al., 2008). IFNα is the

standard of care therapy for metastatic ccRCC that EGFR inhibitors and other potential new therapies are

typically compared against. However, it has limited efficacy and only extends overall survival by a couple

months (Institute, 2015). Exploring PBRM1’s role in modulating these pathways may yield clinically

relevant insights into mechanisms of treatment resistance, at least among this genetic subtype of ccRCC.

RESULTS

PBRM1 status affects ALDH1A1 mRNA, protein, and activity levels

Having established a connection between PBRM1 and ALDH1A1 via RNA-seq in our cell culture

systems and primary tumors, we first confirmed via qRT-PCR that ALDH1A1 mRNA levels were changing

(Figure 3.1A). As expected, PBRM1 knockdown in 786-O increased ALDH1A1 mRNA levels, while

expression of WT PBRM1 in A-704 decreased ALDH1A1 mRNA levels. Additionally, the Pbrm1 NULL

MEFs had significantly higher Aldh1a1 mRNA levels. We next asked whether these mRNA changes

extended to protein levels and enzyme activity in our ccRCC cell lines. In 786-O, both PBRM1 shRNA

lines expressed significantly higher protein levels of ALDH1A1 compared to the control line (Figure 3.1B).

Page 58: Dissertation Cover pages v1.0 EDITS 122815

45

As an additional check, we also probed ALDH1A1 protein levels in the ACHN lines we created, and saw

similar ALDH1A1 changes (Figure 3.1D). Alternatively, in A-704, expression of WT PBRM1, but not

cancer-associated mutant PBRM1, lowered ALDH1A1 protein levels.

ALDH1-class enzyme activity can be measured using the ALDEFLUOR assay, a flow-cytometry

based method that measures the conversion of a fluorescently-tagged aldehyde (bodipy-

aminoacetaldehyde) to an acid (bodipy-aminoacetate) (Corti et al., 2006). While the aldehyde can freely

diffuse into and out of the cell, the acid becomes trapped with the cell, leading to higher levels of

fluorescence. The ALDH1-class chemical inhibitor 4-Diethylaminobenzaldehyde (DEAB) is used to set a

negative baseline. We used this assay to measure ALDH1-class activity in the 786-O and A-704 lines.

The results mirrored those seen for ALDH1A1 protein levels: PBRM1 knockdown in 786-O increased the

ALDEFLUOR-positive population of cells by around 50%; while WT PBRM1 expression in A-704

Figure 3.1 – PBRM1 status affects ALDH1A1 mRNA, protein, and activity levels. A.) qRT-PCR analysis

of ALDH1A1 mRNA levels (relative to GAPDH) in the indicated cell lines. For 786-O and A-704, error bars

represent standard deviations for n=3. For MEFs, errors bars represent SEM for n=3. The delta-delta-Ct

method was used to analyze the data, with 786-O control shRNA, A-704 EV, and WT MEF lines set to 1.

Unpaired t-tests were performed. B.) and D.) Western blot analysis of ALDH1A1 protein levels in the

indicated cell lines. C.) Percentage of cells that were ALDEFLUOR (+) using the ALDEFLUOR assay, in

which the chemical inhibitor DEAB was used to set a negative baseline of fluorescence for each cell line.

n=3 for each line, error bars represent SEM, and unpaired t-tests were performed.

Page 59: Dissertation Cover pages v1.0 EDITS 122815

46

decreased the ALDEFLUOR-positive population by ~33%, while the cancer mutants had no impact

(Figure 3.1C).

PBRM1 knockdown in 786-O increases tumorsphere-forming capacity

As ALDH1A1 has been linked to tumor-initiating cells, we next sought to establish whether

PBRM1 deficiency increased the tumorsphere-forming capacity of cells. In this assay, the cells are plated

as single cells at low density on ultra-low attachment plates using serum-free tumorsphere media

supplemented with growth factors. The ability of the cells to propagate and form free-floating masses of

condensed cells (tumorspheres) can then be evaluated. This method was initially used for the isolation

and characterization of neural stem cells (neurospheres) (Reynolds and Weiss, 1992), and was later

adapted for the study of mammary stem cells and breast cancer stem cells (mammospheres) (Dontu et

al., 2003) (Ponti et al., 2005) (Farnie et al., 2007). It has subsequently been used for the in vitro study of

cancer stem or stem-like cells of various lineages, and, like other anchorage-independent growth-assays,

is thought to represent a good surrogate measure of in vivo tumorigenicity (Freedman and Shin, 1974)

(Yen et al., 2012) (Zhang et al., 2012) (Qiu et al., 2012) (Kim and Alexander, 2014).

PBRM1 knockdown significantly increased the ability of 786-O cells to form free-floating

tumorspheres (Figure 3.2A, left panel). When we tried to passage the control and PBRM1 shRNA #2

tumorspheres (by dissociating the cells and replating at the original density), the PBRM1 shRNA cells

maintained a substantially higher sphere-forming capacity (Figure 3.2A, right panel), indicating a higher

self-renewal rate (Shaw et al., 2012). As part of the passaging process, the number of cells per well after

dissociation were counted for the control and PBRM1 shRNA #2 tumorspheres. There were significantly

more PBRM1 shRNA cells present, indicating that increased tumorsphere formation was not just due to

increased association of individual cells (Figure 3.2B). Representative tumorsphere pictures are shown in

Figure 3.2C. At the second passage, hardly any tumorspheres formed for the control cells – the picture

shows a few individual cells clumped together, which do not meet the criteria for a tumorsphere.

Expression of the T232P PBRM1 cancer mutant increases tumorsphere formation in A-704 cells

Page 60: Dissertation Cover pages v1.0 EDITS 122815

47

We also tested the tumorsphere-formation capacity of the A-704 cells. Overall, the A-704 cells,

including the control EV line, formed fewer tumorspheres than the 786-O cells (Figure 3.3). Expression of

both WT and H1204P PBRM1 qualitatively lowered tumorsphere formation compared to the EV line, but

the differences were not significant. This could be due to the low overall tumorsphere-forming capacity of

the EV line. Interestingly, the T232P mutant formed significantly more tumorspheres. This suggests that

the T232P mutant may not behave as a simple loss-of-function mutant, but may have neomorphic

properties. However, due to the low overall tumorsphere formation rates in these cells, we will perform

subsequent tumorsphere experiments using the 786-O lines.

Figure 3.2 – PBRM1 knockdown in 786-O increases tumorsphere-forming capacity. A.) Tumorsphere-

forming capacity of 786-O lines, Left panel - # of tumorspheres formed initially (1st passage); Right panel - #

of tumorspheres formed for control and PBRM1 shRNA #2 after dissociating cells from 1st passage and

replating (2nd passage). B.) Cell numbers at time of passaging for control and PBRM1 shRNA #2 lines. C.)

Representative images of tumorspheres for control and PBRM1 shRNA cells, at 1st and 2nd passage. Error

bars represent SEM for n=5-6; unpaired t-tests were used for statistical testing.

Page 61: Dissertation Cover pages v1.0 EDITS 122815

48

Anchorage-independent growth in 786-O

requires ALDH1-class enzyme activity

Having established that PBRM1

knockdown cells have increased tumorigenic

potential as seen by colony formation in soft-

agar (Figure 2.6.) and tumorsphere-forming

capacity and self-renewal (Figure 3.2), we

next asked if ALDH1A1 contributed to this

process. We performed tumorsphere assays

in the 786-O control and PBRM1 shRNA lines

to which we added the ALDH1-class inhibitor

DEAB (or 95% ethanol solution as a vehicle

control) (Figure 3.4A, left panel). We used

the same concentration of DEAB that is used to set the negative baseline in the ALDEFLUOR assay

(15uM). The addition of DEAB almost completely abrograted the ability of either line to form

tumorspheres, indicating that some level of ALDH1-class activity is required for tumorsphere formation.

To demonstrate that this effect was due to ALDH1A1 inhibition specifically, we knocked down

ALDH1A1 in each line using three different siRNAs (Figure 3.4B). Like we saw with DEAB addition,

ALDH1A1 knockdown reduced the ability of both lines to form tumorspheres (Figure 3.4A, right panel).

The level of reduction, however, was not as dramatic as that seen with DEAB. This suggests that either

siRNA knockdown of ALDH1A1 is not as effective a means of blocking ALDH1A1 as DEAB addition, or

that other ALDH1-class enzymes (ALDH1A2 or ALDH1A3) may be involved.

To show that these effects were not specific to the tumorsphere assay, we also performed soft-

agar assays in which we titrated in higher doses of DEAB, up to the 15uM dose (Figure 3.4D). We saw a

dose-dependent inhibition of colony formation in the PBRM1 shRNA line. At the highest dose of DEAB,

an equally low number of colonies could form for the control and PBRM1 shRNA lines.

Figure 3.3 – PBRM1 mutant T232P increases

tumorsphere-forming capacity in A-704 cells.

Tumorsphere-formation capacity of A-704 lines. The

differences between the EV and WT/1204P lines were not

statistically significant. n=3/line; unpaired t-tests were

used for statistical testing.

Page 62: Dissertation Cover pages v1.0 EDITS 122815

49

To rule out that the DEAB was killing the cells or inhibiting proliferation, we performed growth

curve analysis on these lines with increasing doses of DEAB (Figure 3.4C). Importantly, DEAB had no

effects on 2-D cellular growth, even at 100uM. Overall, these findings suggest that ALDH1-class activity

is specifically required for anchorage-independent growth, whether measured by the tumorsphere or soft-

agar assays.

ALDH1A1 overexpression increases tumorsphere-forming capacity

We next asked whether ALDH1A1 overexpression on its own could increase tumorsphere-

forming capacity. In the 786-O control line, we transfected in either an empty-vector control plasmid, or a

plasmid containing HA-tagged ALDH1A1. Western blot analysis revealed successful overexpression of

Figure 3.4 – Anchorage-independent growth in 786-O requires ALDH1-class enzyme activity. A.)

Tumorsphere assays in 786-O control and PBRM1 shRNA #2 lines. Left panel – Vehicle control or DEAB (15uM)

was added to the tumorsphere media at time of plating. Right panel – Cells were transfected with non-targeting

siRNA (C) or 1 of 3 ALDH1A1-targting siRNAs (#2, #5, or #7) 24hrs before plating for tumorsphere assay.

n=3/condition; unpaired t-tests were used for statistical testing. B.) Western blot analysis of ALDH1A1 expression

levels in same cells used for tumorsphere assay with ALDH1A1 siRNA knockdown at time of plating. C.) Growth

curve analysis, as measured by Incucyte ZOOM % confluence, of 786-O lines treated with increasing doses of

DEAB or vehicle control. n=3/dose at each timepoint. D.) Colony formation in soft-agar of 786-O control and

PBRM1 shRNA #1 lines. Increasing doses of DEAB, or vehicle control, were mixed in with the soft-agar at time of

plating. n=3/line at each dose, error bars represent SEM, and unpaired t-tests of the 0uM treated condition vs.

higher doses within a cell line were performed.

Page 63: Dissertation Cover pages v1.0 EDITS 122815

50

the ALDH1A1-HA, although expression levels were still lower than the endogenous ALDH1A1 levels seen

with PBRM1 knockdown (Figure 3.5, right panel). Compared to the EV-control, ALDH1A1-HA

overexpression nearly doubled the number of tumorspheres that formed (Figure 3.5, left panel). This

increased tumorsphere-forming capacity was still lower than that seen with PBRM1 knockdown, in line

with the level of ALDH1A1 overexpression.

PBRM1 deficiency increases sensitivity to EGF stimulation

PBRM1 status affects total EGFR levels and downstream signaling

GSEA and gene ontology analysis of the RNA-seq data suggested that PBRM1 influences

multiple pathways, including the EGFR pathway. To test this, we placed the 786-O and A-704 lines in

serum-free conditions, stimulated with EGF, and checked for differences in downstream signaling events.

At serum-free conditions, the 786-O PBRM1 shRNA lines had much higher total EGFR levels (Figure

3.6A). These lines were also more sensitive to EGF stimulation compared to the control line: p-EGFR

(Y1143) levels were elevated, as were p-AKT (T308) and p-ERK levels. This differential sensitivity was

most apparent at 20 ng/mL EGF stimulation. In this cell line, p-AKT levels at S473 were similar. In the

Figure 3.5 – ALDH1A1 overexpression increases tumorsphere-forming capacity. 786-O

control shRNA cells were transfected with either an empty-vector control plasmid (pcDNA-EV), or

a plasmid expressing an HA-tagged ALDH1A1 (pcDNA-ALDH1A1-HA). Right panel – Western

blot analysis showing level of expression of ALDH1A1-HA, in comparison to EV-control

transfected cells (left lane), or endogenous ALDH1A1 levels in the PBRM1 shRNA #2 line (right

lane). Left panel – tumorsphere formation in 786-O control cells transfected with the indicated

plasmids. n=3/line; an unpaired t-test was used for statistical testing.

Page 64: Dissertation Cover pages v1.0 EDITS 122815

51

A-704 lines, similar trends were evident (Figure 3.6B). At baseline, expression of WT PBRM1 resulted in

lower total EGFR levels compared to the EV control or PBRM1 mutant lines. When stimulated with EGF,

the WT line responded with lower levels of p-EGFR (Y1173), p-AKT (at both T308 and S473), and p-ERK,

compared to the other lines. In their responses, the PBRM1 mutant lines resembled the EV control.

PBRM1 status affects growth responsiveness to EGF stimulation

Having seen differences in downstream signaling in response to EGF stimulation, we

hypothesized that PBRM1 status would also affect how the cells grew in response to EGF. To test this,

we grew the cells at very low concentrations of serum (0.1% and 2.5% serum for 786-O and A-704,

respectively), such that they didn’t die but only grew in a limited fashion. Without EGF stimulation, the

786-O PBRM1 shRNA lines grew slightly faster than the control line (Figure 3.7A – solid lines). Likewise,

the A-704 EV line had a slight growth advantage over the WT line (Figure 3.7B – solid lines). When

stimulated with EGF, both 786-O PBRM1 shRNA lines responded with faster growth; the control line, on

the other hand, did not respond at all (dashed lines). In the A-704 lines, EGF stimulation produced even

Figure 3.6 – PBRM1 status affects total EGFR levels and downstream signaling. A.) Western blot

analysis of 786-O lines stimulated with the indicated concentration of EGF for 30’ under serum-free

conditions. B.) Western blot analysis of A-704 lines stimulated with 1ng/mL EGF for 30’ under serum-

free conditions.

Page 65: Dissertation Cover pages v1.0 EDITS 122815

52

more divergent outcomes: the EV line responded with a slight growth increase, while EGF stimulation had

a growth inhibitory effect on the WT line (dashed lines). An anti-growth EGF effect in seen in some cell

lines (Barnes, 1982) (Xie et al., 1997) (Yamasaki et al., 2003), including sometimes for ccRCC (Ramp et

al., 1997).

PBRM1 knockdown in 786-O renders cells more resistant to EGFR inhibition

We next explored whether PBRM1 knockdown influences sensitivity to EGFR inhibition using the

drug erlotinib. Having established that PBRM1 knockdown has particularly large effects on 3-D growth,

we tested how EGFR inhibition affected colony growth in soft-agar in the 786-O lines. As expected, 786

PBRM shRNA #1 cells formed significantly more colonies compared to control cells when treated with

vehicle (Figure 3.8). When treated with an intermediate dose of erlotinib (1uM), the control cells almost

completely lost their ability to form colonies, while the knockdown cells still formed significantly more

colonies, albeit at a reduced level. At a higher dose of erlotinib (10uM), colony growth was severely

limited in both lines.

Decreased sensitivity to IFNα with PBRM1 deficiency

PBRM1 deficiency reduces basal and IFNα-induced IFI27 mRNA levels

Figure 3.7 – PBRM1 status affects growth responsiveness to EGF stimulation. A.) 2-D growth curves of

786-O lines grown on 48-well tissue culture plates at 0.1% serum in the presence of PBS (0 EGF, solid lines) or

50 ng/mL EGF (50 EGF, dotted lines). B.) 2-D growth curves of A-704 lines grown on 48-well tissue culture plates

at 2.5% serum in the presence of PBS (0 EGF, solid lines) or 10 ng/mL EGF (10 EGF, dotted lines). n=6 per

timepoint for each line. Two-way ANOVAs using Tukey’s multiple comparisons test was performed on each set of

growth curves, and significant differences between the untreated and EGF stimulated condition for a given line

are shown. * P<0.05; ** P<0.01; *** P <0.001; **** P<0.0001.

Page 66: Dissertation Cover pages v1.0 EDITS 122815

53

Gene expression profiling of our cell

lines at low serum suggested that interferon-

response pathways were also perturbed with

PBRM1 alteration. At both normal and

reduced serum, one of the top downregulated

genes with PBRM1 deficiency was IFI27,

also known as Interferon, Alpha-Inducible

Protein 27 or ISG12. IFI27 is a thought to be

a pro-apoptotic interferon-response gene that

localizes to the mitochondrial membrane and

can enhance cytochrome c release and

apoptosis in conjunction with other pro-

apoptotic stimuli (Rosebeck and Leaman,

2008) (Cheriyath et al., 2011). Some reports

have implicated IFI27 as a key mediator of TRAIL-induced apoptosis (Liu et al., 2014b) (Liu et al., 2014a).

While decreased IFI27 levels have been found in some cancers (Mihalich et al., 2012), other studies have

reported increased IFI27 levels (Suomela et al., 2004) (Wenzel et al., 2008). In connection to the

SWI/SNF complex, one previous study has linked IFI27 expression to the activity of BRG1 downstream of

IFNα and STAT2 signaling (Huang et al., 2002), while another study saw no IFI27 dependence on ARID2

(Yan et al., 2005).

To determine PBRM1’s relationship to IFN signaling and IFI27 induction, we treated the 786-O

and A-704 lines with IFNα and measured IFI27 mRNA levels at various timepoints. In agreement with the

RNA-seq data, IFI27 mRNA levels were significantly lower at baseline in the 786-O shRNA #1 line and

the A-704 EV line (Figure 3.9A and B). When stimulated with IFNα, IFI27 mRNA levels rose dramatically

in the 786-O control line, cresting at a nearly 80-fold induction at 24hrs (Figure 3.9A). The 786-O PBRM1

shRNA line was much less responsive at all time points. IFI27 was not as highly induced in the A-704

lines with IFNα stimulation, but WT PBRM1 expression still heightened the response (Figure 3.9B). At all

Figure 3.8 – PBRM1 knockdown in 786-O renders

cells more resistant to EGFR inhibition. Colony

formation in soft-agar of 786-O control and PBRM1

shRNA #1 lines. Increasing doses of erlotinib, or vehicle

control, were mixed in with the soft-agar at time of plating.

n=3/line at each dose, error bars represent SEM, and

unpaired t-tests using the Holm-Sidak method to correct

for multiple comparisons between cell lines at a given

dose were performed.

Page 67: Dissertation Cover pages v1.0 EDITS 122815

54

timepoints except 48hrs, IFI27 was induced to a higher degree in the WT line compared to the EV (at

48hrs, there was a trend towards higher expression).

IFI27 mRNA levels are lower in PBRM1 mutant tumors

We next asked whether IFI27 expression changes could be seen in primary ccRCC tumors based

on PBRM1 status. We applied the same methodology we used in Chapter 2 (Figure 2.16). In the TCGA

dataset, IFI27 expression levels were significantly lower in PBRM1 mutant tumors (Figure 3.10), in line

with our cell culture models. However, in the Peña-Llopis et al dataset, IFI27 was not among the

significant genes that distinguished PBRM1 mutant vs. non-mutant tumors.

A-704 cells expressing PBRM1 are more growth inhibited by IFNα

To test whether the differential response to IFNα had any functional consequences, we

performed growth-curve analysis on the A-704 lines in the presence of increasing doses of IFNα. The

WT PBRM1 expressing line was more growth inhibited by IFNα – even the lowest dose of 10 units/mL

slowed growth to some extent, while the higher doses (200 and 500 units/mL) almost completely inhibited

Figure 3.9 – PBRM1 deficiency reduces basal and IFNα-induced IFI27 mRNA levels. qRT-PCR analysis of

IFI27 mRNA levels (relative to GAPDH) in the 786-O (A) and A-704 (B) lines after treatment with 100 units/mL of

IFNα for the indicated times. The delta-delta-Ct method was used to analyze the data, with timepoint 0hr for the

786-O control shRNA and A-704 EV lines set to 1. Error bars represent standard deviations for n=3. Unpaired t-

tests were performed comparing the control and PBRM1-altered lines at a given timepoint. * P<0.05; ** P<0.01; ***

P <0.001; **** P<0.0001.

Page 68: Dissertation Cover pages v1.0 EDITS 122815

55

growth (Figure 3.11, right panel). Meanwhile,

10 units/mL had no effect on the EV line, and

the highest doses slowed but did not halt

growth (Figure 3.11, right panel).

Figure 3.10 – IFI27 mRNA levels are lower in PBRM1

mutant tumors. Provisional TCGA data for ccRCC was

taken from the cBioPortal for Cancer Genomics in

November 2015 – 417 tumor samples had sequencing

and expression data available. We divided the tumors

into those with PBRM1 mutations (Mut) and those without

(WT) – 122/417 tumors were mutated. mRNA z-score

levels are shown for IFI27 in the form of a Tukey plot with

the outliers not shown. Statistical testing was performed

using the Mann-Whitney test.

Figure 3.11 – A-704 cells expressing PBRM1 are more growth inhibited by IFNα. 2-D growth curves of A-704

lines (EV, left, and WT, right) grown on 48-well tissue culture plates at full serum in the presence of the indicated

doses of IFNα or vehicle control. Growth data was collected using an Incucyte Zoom machine as a % confluence

of a well. n=3 per timepoint per dose for each line.

Page 69: Dissertation Cover pages v1.0 EDITS 122815

56

DISCUSSION

In this chapter, we set out to confirm and further explore the connection between PBRM1 and

some of the genes and pathways implicated from our gene expression analysis in Chapter 2. First, we

investigated PBRM1 regulation of ALDH1A1, a putative marker of tumor-initiating cells. We verified via

qRT-PCR that ALDH1A1 mRNA levels increased with PBRM1 deficiency in our cell culture systems, and

that this translated into increased protein expression in our ccRCC lines (Figure 3.1). We then went a

step further and used the ALDEFLUOR assay to show that ALDH1-class enzyme activity increased with

PBRM1 deficiency.

We then demonstrated that this ALDH1A1 upregulation has functional consequences for

tumorigenic potential. In two different anchorage-independent growth assays (tumorsphere formation and

colony formation in soft agar), we chemically inhibited ALDH1-class enzyme activity with DEAB and

abrogated 3-D growth (Figure 3.4). In the tumorsphere assay, this inhibition occurred in both the 786-O

control and PBRM1 knockdown lines, indicating that PBRM1 knockdown cells had not become uniquely

dependent on ALDH1-activity. Instead, these findings imply that some minimal level of ALDH1-activity is

required for anchorage-independent growth, and suggest more of a dose-response relationship, where

having more ALDH1-activity translates into more 3-D growth. The ALDH1A1 overexpression experiment

in the control cells further supports this model, where higher expression of ALDH1A1 led to increased

tumorsphere-forming capacity (Figure 3.5). ALDH1-effects seem to be limited to anchorage-independent

growth, as even very high doses of DEAB had no effect on 2-D growth on a plate.

The overexpression experiment also indicated that this effect is specific to ALDH1A1, at least in

part (versus ALDH1A2 or ALHD1A3). Decreased tumorsphere formation with siRNA knockdown of

ALDH1A1 provides additional evidence of an ALDH1A1-specific effect. However, the level of

tumorsphere inhibition with siRNA was not as great as with DEAB, suggesting either less effective

inhibition of ALDH1A1 with siRNA, the involvement of other ALDH1-class enzymes, or off-target effects

by DEAB. While the RNA-seq analysis indicates that ALDH1A2 and ALDH1A3 mRNA levels did not

change with PBRM1 alteration, future experiments could more definitively determine if they were

contributing to 3-D growth processes in these cells.

Page 70: Dissertation Cover pages v1.0 EDITS 122815

57

While our findings suggest that ALDH1A1 expression and activity are increased with PBRM1

deficiency, and that this increases tumorigenic potential, we did not explore how ALDH1A1 could be

mediating this effect. Although prior studies offer some possible explanations, such as non-canonical

retinoic acid signaling through PPARβ/δ, or increased protection from oxidative damage, we do not know

what factors are involved in this setting. Future experiments could investigate how ALDH1A1 increases

tumorigenic potential in our ccRCC cell lines. For example, the role of retinoic acid signaling could be

further explored. Total levels of retinoic acid (all-trans or cis) could be measured and compared in the

minus- and plus-PBRM1 conditions. The mRNA levels of known targets of canonical vs. non-canonical

retinoic acid signaling could be quantified. Retinoic acid receptor agonists and antagonists could be

added to the anchorage-independent growth assays. Other experiments could investigate the role of

reactive oxygen species and oxidative protection by ALDH1A1.

Prior studies investigating SWI/SNF components have often found that they play wide-ranging

roles in a cell – ChIP-seq and gene expression profiling experiments suggest that SWI/SNF complexes

bind across the genome and modulate a multitude of genes and pathways (Ho et al., 2009) (Euskirchen

et al., 2011). We too have found this to be the case for PBRM1. Our RNA-seq analysis suggested that

PBRM1 was affecting the EGFR pathway, and we confirmed that in this chapter. After EGF stimulation,

we saw heightened downstream signaling and growth responsiveness in the lines with PBRM1 deficiency

(Figures 3.6 and 3.7). At baseline, these lines had dramatically higher total EGFR protein levels,

providing a possible explanation for this increased sensitivity. Based on the RNA-seq data, EGFR mRNA

levels were also moderately increased, suggesting that this effect is at least partially transcriptional

(normal serum RPKM values: 786-O control shRNA - 37 ; PBRM1 shRNA #1 - 50; PBRM1 shRNA #2 -

58; A-704 EV - 205; A-704 WT - 178). However, this does not rule out post-translational events, such as

protein modification, altered EGFR cycling to the membrane, or reduced ubiquitination and proteosomal

degradation. Future experiments could discern why EGFR protein levels were increasing so much.

These changes in EGFR signaling, however, did not translate into an “oncogene addiction,” at

least for the 786-O cells. When treated with erlotinib, the PBRM knockdown cells were more resistant to

3-D growth inhibition – at the intermediate dose of erlotinib, they could still form colonies in soft agar,

while the control cells had largely lost this ability (Figure 3.8). This suggests that EGFR inhibitors might

Page 71: Dissertation Cover pages v1.0 EDITS 122815

58

have better clinical efficacy in non-PBRM1 mutant tumors, an idea that could be further explored using

tissue culture models and xenograft studies before being put to the test in the clinic.

Lastly, we explored the connection between PBRM1 and IFN signaling suggested by the low-

serum RNA-seq analysis. We found that PBRM1 deficiency lowered the induction of the reportedly pro-

apoptotic interferon response gene IFI27 with IFNα stimulation, and that expression of WT PBRM1 in A-

704 cells increased growth inhibition by IFNα (Figures 3.9 and 3.11). In light of previous studies with

mixed findings on SWI/SNF control of IFI27 induction (Huang et al., 2002) (Yan et al., 2005), our results

suggest some degree of cell-type specificity in terms of what particular IFN response genes are SWI/SNF

dependent. For example, we saw no changes in IFITM1, the IFN response gene found to be ARID2

dependent in Yan et al, 2005, in our RNA-seq analysis. As reports describe IFI27 as promoting apoptosis

in conjunction with other stimuli, such as DNA-damage or TRAIL-treatment, future experiments could

measure how PBRM1 status affects apoptosis levels in response to some of these combinations (e.g.,

IFNα + TRAIL). As IFN therapy is still widely used for the treatment of ccRCC, these findings have

clinical implications and suggest that PBRM1 mutant tumors would be more resistant to IFN therapy.

They also imply that PBRM1 status may impact therapeutic response to the emerging class of immune

checkpoint therapies, as PBRM1 mutant tumor cells may be less susceptible to some of the death-

inducing mechanisms of newly activated T-cells. It will be interesting to see if ccRCC patients that

respond to immune-checkpoint therapy are enriched for non-PBRM1 mutant tumors.

In this chapter, we have confirmed that PBRM1 plays a role in the regulation of ALDH1A1

expression, and the EGFR and IFN pathways, and that this has important functional consequences.

However, we have not yet explored the molecular mechanisms through which PBRM1 could be exerting

such control. That will be the subject of the next chapter.

Page 72: Dissertation Cover pages v1.0 EDITS 122815

59

CHAPTER 4: With PBRM1 deficiency, ARID2 still binds in a SWI/SNF complex, but

with altered genomic localization associated with differential gene expression

Page 73: Dissertation Cover pages v1.0 EDITS 122815

60

BACKGROUND AND SIGNIFICANCE

In Chapters 2 and 3, we examined genes and pathways regulated by PBRM1 that contribute to its

tumor suppressor function. So far, this analysis has treated PBRM1 more as a single protein that

represses or activates transcription on its own. However, when studying PBRM1, it is important to keep

in mind that it is part of a multi-subunit complex, where loss of a single subunit could have complex

effects on the remaining subunits’ assembly and action. For example, SNF5 loss is only transforming

when BRG1 is present, implying that aberrant SWI/SNF activity, but not the total lack of activity, is

oncogenic (Wang et al., 2009). A recently identified SWI/SNF complex member, SS18, also dramatically

alters complex formation in cancer (Kadoch and Crabtree, 2013). Numerous recent reports have also

demonstrated a synthetic lethal relationship between homologous SWI/SNF subunits. For example, two

independent studies identified BRM as an essential gene in BRG1 mutant cancers (Hoffman et al., 2014)

(Wilson et al., 2014). ARID1A and ARID1B, mutually exclusive subunits in the BAF complex, exhibit a

similar relationship (Helming et al., 2014b). These studies indicate that some level of SWI/SNF activity

may be essential for growth in cells, and suggest specific cancer vulnerabilities that could be targeted

therapeutically.

It has previously been shown that, at least in some cell lines, PBRM1 is not required for PBAF

complex assembly (Yan et al., 2005). In HCC1143 cells, a breast cancer line where PBRM1 is not

expressed, ARID2 was expressed and was still capable of associating in a complex with other SWI/SNF

components. In HeLa cells, ARID2 was required for PBAF complex assembly – without it, the complex

fell apart and PBRM1 was degraded. However, the reverse was not true – PBRM1 knockdown did not

cause a reciprocal decline in ARID2 levels. These findings suggest that a PBAF-like complex remains

even after PBRM1 loss.

In this chapter, we will explore how PBRM1 deficiency in ccRCC impacts some of the remaining

subunits of the PBAF complex, such as ARID2. In doing so, we hope to better understand the molecular

mechanisms that lead to some of the transcriptional changes previously described.

RESULTS

PBRM1 deficiency increases ARID2 protein levels

Page 74: Dissertation Cover pages v1.0 EDITS 122815

61

We first asked how PBRM1 deficiency impacts the protein levels of some of the remaining PBAF

subunits in our cell line systems. In 786-O cells, PBRM1 knockdown led to a large increase in ARID2

protein levels (Figure 4.1A). SNF5 protein levels also increased in both PBRM1 shRNA lines. In the A-

704 cells, stable expression of WT PBRM1 resulted in lower ARID2 protein levels, whereas levels

remained stable with expression of PBRM1 cancer-associated mutants. SNF5 levels did not differ among

the A-704 lines. Also, in both 786-O and A-704 lines, BRG1 levels did not change consistently.

We also checked PBAF-subunit protein levels in ACHN cells (Figure 4.1B). PBRM1 knockdown

led to even more dramatic increases in ARID2 protein levels. SNF5 levels did not change consistently

between the PBRM1 hairpins, and BRG1 levels remained stable. We concluded that PBRM1 deficiency

results in increased protein levels of ARID2.

Increased ARID2 levels are at least partially due to increased transcription

To see if these ARID2 changes happened at the mRNA level, we performed qRT-PCR analysis in

the 786-O and A-704 lines. As we saw at the protein level, PBRM1 knockdown in the 786-O cells led to

increased ARID2 mRNA levels, while WT PBRM1 expression in A-704 cells led to decreased mRNA

levels (Figure 4.2A). While these mRNA changes were modest, they suggest that increased ARID2

levels are at least partially due to increased transcription.

Figure 4.1 - PBRM1 deficiency increases ARID2 protein levels. Western blot analysis of the indicated

PBAF complex components in 786-O and A-704 (A) and ACHN cell lines (B).

Page 75: Dissertation Cover pages v1.0 EDITS 122815

62

We next asked whether we could see similar changes in ccRCC primary tumors using the publicly

available datasets. In both datasets, PBRM1 mutation was associated with significantly higher ARID2

mRNA levels (Figure 4.2B).

In the absence of PBRM1, ARID2 still associates with SWI/SNF components

Having seen increased ARID2 levels with PBRM1 knockdown, we next tested whether this ARID2

was assembling into a SWI/SNF complex. We performed immunoprecipitation (IP) experiments using an

antibody against BRG1 and checked for binding of ARID2, SNF5, and PBRM1. In the 786-O cells, high

levels of ARID2 still bound to BRG1 after PBRM1 knockdown (Figure 4.3A). Similarly, in the A-704 cells,

ARID2 still bound to BRG1, even in the absence of PBRM1 (the EV line) (Figure 4.3B). These results are

in agreement with previous studies that found that PBRM1 was dispensable for the rest of the PBAF

complex assembling (Yan et al., 2005).

While these IP experiments indicated that some ARID2 still associated with other SWI/SNF

components, they did not preclude the possibility that ARID2 was binding to other proteins outside the

PBAF complex. We performed glycerol gradient fractionation experiments in the 786-O lines to determine

if this was the case. If ARID2 was binding outside of the PBAF complex, we would expect to see some of

Figure 4.2 - Increased ARID2 levels are at least partially due to increased transcription. A.) qRT-PCR

analysis of ARID2 mRNA levels (relative to GAPDH) in the indicated cell lines. Error bars represent

standard deviations for n=3, the delta-delta-Ct method was used to analyze the data, with 786-O control

shRNA and A-704 EV lines set to 1, and unpaired t-tests were performed. B.) Provisional TCGA data for

ccRCC was taken from the cBioPortal for Cancer Genomics in November 2015 – 417 tumor samples had

sequencing and expression data available (mRNA z-scores). ccRCC expression data was also taken from

Peña-Llopis et al (relative mRNA expression levels). We divided the tumors into those with PBRM1

mutations (Mut) and those without (WT) – in the TCGA data,122/417 tumors were mutated, while 21/53

tumors had PBRM1 mutations in Peña-Llopis et al. ARID2 levels are shown in the form of Tukey plots with

the outliers not shown. Statistical testing was performed using the Mann-Whitney test.

Page 76: Dissertation Cover pages v1.0 EDITS 122815

63

it eluting in fractions separate from other SWI/SNF subunits. Based on published reports of glycerol

gradient fractionation of SWI/SNF complexes, we used a 10-30% glycerol gradient, and spun for 18 hours

to ensure good separation of lower molecular weight complexes where we hypothesized ARID2 might be

relocating (Lessard et al., 2007) (Kadoch and Crabtree, 2013). Looking from fractions 2-24 in the control

cells (out of 24 total fractions), ARID2 eluted in the heavier fractions, mostly in fraction 2, with some

protein in fractions 4 and 6 (Figure 4.4A-left panel). PBRM1 showed the same pattern. BRG1 and SNF5

were found at slightly lighter fractions, both peaking in fraction 4 and extending to fraction 8.

In both PBRM1 knockdown lines, there were subtle shifts in the elution pattern (Figure 4.4A –

middle and right panels). ARID2 now peaked in fraction 4, a slightly lighter fraction. Likewise, the peaks

of BRG1 and SNF both shifted to a slightly lighter fraction, fraction 6. With these shifts, there was now

more overlap between ARID2 and BRG1 and SNF5, indicating that with PBRM1 deficiency ARID2 could

be more bound in SWI/SNF complexes. Additionally, no ARID2 was found in lighter fractions without

Figure 4.3 – In the absence of PBRM1, ARID2 still associates with SWI/SNF components.

BRG1 IP experiments were performed on 0.5mg of protein from nuclear extracts of 786-O (A) and

A-704 (B) cell lines. A matched isotype IgG was used for control IPs. Inputs are aliquots taken

from pre-cleared nuclear extracts before the IPs were performed.

Page 77: Dissertation Cover pages v1.0 EDITS 122815

64

BRG1 and SNF5, suggesting that it was not binding in new complexes. As expected, PBRM1 levels were

barely detectable but when visible displayed the same pattern as the control cells.

Because we did not see ARID2 in lighter fractions, we also looked at ARID2 elution patterns at

higher resolution in the heavier fractions (Figure 4.4B). In the control cells, ARID2 could be found in

fractions 1-5, but peaked in fraction 1. BRG1 and SNF5 were found mostly in fractions 4-6. These

results imply that the majority of ARID2 is found outside the PBAF complex in the control cells. After

PBRM1 knockdown, ARID2 shifted to lighter fractions, and could be found in fractions 2-6, peaking in

fractions 4-5. BRG1 and SNF also shifted to slightly lighter fractions, and could now be found in fractions

4-7, with peaks in fraction 5. Overall, these results support the IP findings and suggest that after PBRM1

knockdown, more ARID2 is bound in a PBAF-like complex that also contains BRG1 and SNF5. This

complex elutes in slightly lighter fractions, perhaps reflecting the loss of PBRM1 inclusion.

Subcellular fractionation experiments reveal that ARID2 increases in both the soluble and

insoluble nuclear fractions

As the IP and glycerol gradient experiments were both performed on the soluble nuclear fraction

of the 786-O cells, we next performed subcellular fractionation experiments to see if ARID2 was transiting

to other cellular compartments (Figure 4.5). We used tubulin (TUBB) as a cytosolic marker, MYC as

Figure 4.4 – Glycerol gradient fractionation of 786-O cell lines. For each cell line, glycerol gradients were

made using 10% and 30% glycerol solutions, to a total volume of 11.5 mL. Nuclear extracts of the 786-O

lines were collected, they were diluted to 1.7mg/mL, and 0.5 mL was carefully layered on top of the glycerol

gradients. The gradients were spun for 18 hrs in a swinging bucket rotor in an ultracentrigure, and 0.5 mL

fractions were collected from the bottom (heavier fractions) of the tube (24 fractions total). Aliquots of the

fractions were combined with 2x sample buffer and subjected to Western Blot analysis. A.) Wider view of the

fractionation, from fractions 2-24, skipping every other fraction, except in the middle. B.) Higher resolution

analysis of the heavy fractions, fractions 1-10.

Page 78: Dissertation Cover pages v1.0 EDITS 122815

65

marker of both the soluble and insoluble nuclear fractions, and lamin B1 (LMNB1) and H3 as markers of

the insoluble nuclear fraction. Before being knocked down, PBRM1 could be found in both the soluble

and insoluble nuclear fractions, as could ARID2. With PBRM1 knockdown, ARID2 levels increased in

both the soluble and insoluble nuclear fractions. Additionally, after PBRM1 knockdown, less SNF5

seemed to locate to the insoluble nuclear fraction. No BRG1 could be detected in the insoluble nuclear

fraction in any line. No SWI/SNF component could be found in the cytoplasmic fraction.

Along with the IP and glycerol gradient findings, these data suggest that after PBRM1

knockdown, some ARID2 still associates with SWI/SNF components in the soluble nuclear fraction, while

other ARID2 protein transits to the insoluble nuclear fraction, perhaps outside of the SWI/SNF complex.

More experiments are needed to clarify the role of ARID2 in the insoluble nuclear fraction.

Figure 4.5 – Subcellular fractionation of the 786-O lines. 786-O cells

were subjected to subcellular fractionation to separate the cytosolic and

nuclear fractions. The insoluble nuclear fraction was resuspended in 2x

sample buffer, subject to brief sonication, and boiled for 15 minutes to be

able to run on a gel. Western blot analysis of the different fractions was

then performed.

Page 79: Dissertation Cover pages v1.0 EDITS 122815

66

ARID2 ChIP-seq indicates altered genomic binding of the PBAF complex that remains after

PBRM1 knockdown

After PBRM1 knockdown, ARID2 binds to fewer areas of the genome

Based on its domain structure and the fact that it’s one of the defining subunits of the PBAF

complex, PBRM1 has been proposed to be a “targeting” SWI/SNF subunit, helping direct the complex to

particular genomic loci. Because of this and our finding that a PBAF-like complex containing ARID2

remained intact after PBRM1 knockdown, we decided to probe the genomic binding of ARID2 before and

after PBRM1 knockdown by performing ARID2 chromatin immunoprecipitation followed by sequencing

(ChIP-seq) in the 786-O lines. If PBRM1 was serving as a “targeting” subunit, we would expect to see

altered ARID2 genomic binding after PBRM1 knockdown.

We used 75bp single-end sequencing on our ChIP-seq libraries, and achieved good depth of

coverage on all samples:

We aligned our reads using Bowtie 2 (Langmead and Salzberg, 2012), and then visualized the read pile-

ups on the UCSC genome browser. In general, the binding profiles were broad and seemed to resemble

wide binding domains rather than sharp peaks, similar to the profile seen for some histone marks, such

as H3K27Me3. Looking at the input DNA, serving as controls, we saw no such patterns. For this reason,

we decided to call peaks using SICER, which was designed to recognize and call disperse ChIP-enriched

regions with high sensitivity and specificity (Zang et al., 2009). We also used SICER to call differentially

enriched regions between the samples.

First, we looked at the broad binding patterns of ARID2 across the genome. There was a general

loss of ARID2 binding with PBRM1 knockdown: 21,704 peaks were called in the control line, while only

9,161 and 5,456 peaks were called in the PBRM1 shRNA #1 and #2 lines, respectively (Figure 4.6A). We

used HOMER to annotate the genomic feature (e.g., intron, exon, TSS) most closely associated with

ChIP-seq depth of coverage

(million reads):

786-O Line ARID2 INPUT

Control shRNA 28.6 33.5

PBRM1 shRNA #1 32.4 28.6

PBRM1 shRNA #2 23.2 29.6

Table 4.1 – ChIP-seq depth of coverage.

Page 80: Dissertation Cover pages v1.0 EDITS 122815

67

each called peak (Heinz et al., 2010). Looking across the lines, we saw subtle shifts in the genomic

regions associated with ARID2 peaks (Figure 4.6A). With PBRM1 knockdown, ARID2 was proportionally

slightly more bound to intergenic regions, and slightly less bound to intronic, promoter, and 5UTR regions.

Overall, although these shifts were small, they were significantly different via the Chi-square test. More

strikingly, absolute numbers of ARID2 peaks decreased at every genomic feature with PBRM1

knockdown (Figure 4.6B). We also used SICER to identify regions with differential ARID2 binding

depending on PBRM1 status. Many more ARID2 peaks were lost than gained in both PBRM1 hairpin

lines compared to the control (Figure 4.6C).

To confirm that SICER was accurately calling ARID2 peaks and differential regions between the

samples, we looked at the average binding enrichment profiles for the regions of differential binding called

by SICER (Figure 4.7). There were four sets of regions of differential binding: regions with gained ARID2

peaks in the PBRM1 shRNA #1 or #2 lines; and regions with lost ARID2 peaks in the PBRM1 shRNA #1

Figure 4.6 – Genomic annotation of ARID2 binding in 786-O lines. HOMER was used to annotate the genomic

feature most closely associated with each called peak in the 786-O lines. A.) Pie charts of the distribution of ARID2

binding across genomic features. Those features associated with 0 peaks in all lines are not shown. The absolute

numbers of peaks associated with each feature are shown in the legends, including the total number of called

peaks. B.) Intra-line comparison of genomic annotation of ARID2 peaks in 786-O cells – the y-axis is in log-scale.

C.) Table of number of gained and lost ARID2 peaks in the PBRM1 shRNA lines vs. the control, called by SICER.

Page 81: Dissertation Cover pages v1.0 EDITS 122815

68

or #2 lines. When we looked at the average binding enrichment for each cell line at these genomic

locations, we saw that differential peaks were accurately being called. The lost peak regions for both

PBRM1 hairpins produced sharp binding enrichment profiles only in the control sample (left panel), while

the gained peak regions produced binding enrichment peaks only in the corresponding hairpin line

(middle and right panels). Additionally, there does not appear to be much overlap in the differential

ARID2 peaks between the two PBRM1 hairpin lines, as the gained peak regions for one hairpin did not

show an enrichment in the other hairpin line.

k-means clustering of ARID2 binding enrichment reveals broad shifts in binding patterns with PBRM1

knockdown

While our analysis so far indicated a general loss of ARID2 binding to the genome with PBRM1

knockdown, we were curious whether this occurred at a subset of genomic regions, or whether there was

broader shift in binding patterns. To answer this question, we performed k-means clustering analysis on

the ARID2 binding enrichment in the control sample. We limited the analysis to the genomic locations (+/-

2kb) of only the transcribed genes in 786-O, as defined in Chapter 2. We set cluster number (k) equal to

5, because at higher k’s the binding enrichment profiles became more finely segmented but did not differ

Figure 4.7 – ARID2 binding enrichment profiles for ARID2 peaks gained or lost with PBRM1

knockdown. SICER was used to call ARID2 ChIP-seq binding peaks, and to identify regions of differential

binding between the control line and the PBRM1 shRNA lines. The average ARID2 binding enrichment for

each line for these differential regions was then generated using Galaxy’s deepTools. The plots illustrate the

average binding enrichment from 5kb upstream to 5kb downstream of the peak center. All gained or lost

peaks were in the PBRM1 shRNA line (#1 or #2) compared to the control line. The numbers in parenthesis are

the number of gained or lost peaks.

Page 82: Dissertation Cover pages v1.0 EDITS 122815

69

in the general pattern seen at 5 clusters. As shown in Figure 4.8A, the clustering analysis produced

clusters of regions with distinct ARID2 binding enrichment profiles: cluster 1 had high overall binding

enrichment, with both upstream and downstream peaks; cluster 2 regions were defined by a sharp peak

about 1000 kb upstream of the transcriptional start site (TSS); cluster 3 had a sharp binding peak

immediately before the TSS; cluster 4 regions were distinguished by peaks downstream of the

Figure 4.8 – k-means clustering of ARID2 binding enrichment in 786-O control cells. Using Galaxy’s

deepTools, we performed k-means clustering of the ARID2 binding enrichment in the 786-O control cells. K

was set equal to 5, and we looked at the regions surrounding all transcribed genes (+/- 2kb) in the 786-O cells,

as defined by the RNA-seq analysis in Chapter 2. A.) Left panel - average binding enrichment for the 5 clusters

identified in the control cells. The same cluster sets were then applied to the PBRM1 shRNA ARID2 ChIP-seq

results (right panels). B.) Tukey plots (with outliers shown) of the expression changes for the genes in each

cluster defined in (A). One-way ANOVA with multiple comparisons correction statistical testing was performed

between the clusters. C.) Gene ontology analysis showing the top 12 enriched biological processes for the

genes comprising Clusters 3 and 4. TSS=transcriptional start site; TES=transcriptional end site.

Page 83: Dissertation Cover pages v1.0 EDITS 122815

70

transcriptional end site (TES); and cluster 5 was defined by low binding with no peaks. When we applied

these clusters to the PBRM1 shRNA lines, there was a general loss of binding patterns across all clusters

(Figure 4.8A – right panels). This suggests that in addition to ARID2 binding less to the genome, there is

a broad shift in where it does bind after PBRM1 knockdown.

We also wondered whether there were any gene expression differences between the clusters.

We used the log2(fold-change) values from the normal serum 786-O RNA-seq analysis to test this

(PBRM1 shRNA/control shRNA fold-change values). Although we did not see dramatic differences,

clusters 3 and 4, defined by sharp peaks immediately upstream and downstream of genes, respectively,

had significantly higher gene expression changes compared to cluster 5, defined by low overall binding

enrichment (Figure 4.8B). This means that in the control cells, genes in clusters 3 and 4 are being more

repressed (i.e., with PBRM1 knockdown, they become more upregulated). Gene ontology analysis of the

genes in these clusters revealed that they are enriched for genes related to RNA splicing, DNA repair, cell

cycle progression, and ribonucleoprotein complex assembly (Figure 4.8C).

Having thus far seen a general loss of ARID2 binding in the PBRM1 shRNA lines, we wanted to

check if there were some regions with binding enrichment in these lines. Clustering analysis of the

PBRM1 shRNA lines revealed that regions could be clustered based on ARID2 binding enrichment in

these lines as well (Figure 4.9A and B). In fact, the clusters had enrichment profiles that were remarkably

similar to the control line clusters. However, the clusters generated consisted of entirely different sets of

regions, as evident by the results when these clustering solutions were applied to the other lines. The

PBRM1 shRNA #1 clusters produced flat profiles in the control and shRNA #2 lines, and the PBRM1

shRNA #2 clusters did the same in the control and PBRM1 shRNA #1 lines. These results indicate that

some ARID2 still binds to the genome after PBRM1 knockdown, but in broadly shifted patterns. The low

level of overlap in binding enrichment between the two PBRM1 shRNA lines (Figures 4.7 and 4.9)

suggests that ARID2 genomic binding without PBRM1 may be stochastic in nature.

Page 84: Dissertation Cover pages v1.0 EDITS 122815

71

ARID2 binding differences correlate with gene expression changes

The clustering analysis suggested that there might be an association between ARID2 binding

patterns and gene expression changes. To further explore this relationship, we plotted the average

ARID2 binding score for each cell line for groups of genes with different expression changes (Figure

4.10A). For the genes that are most upregulated (top quartile) with PBRM1 knockdown, there was a peak

of ARID2 binding immediately preceding the TSS in the control line (left panel). In the PBRM1 shRNA #1

line, this peak is lost, and in the shRNA #2 line, it is greatly diminished. For the genes that are most

downregulated (bottom quartile) with PBRM1 knockdown, similar changes were visible: in the control

cells, there is a peak of ARID2 binding preceding the TSS, which is lost or greatly reduced with PBRM1

Figure 4.9 – k-means clustering of ARID2 binding enrichment in 786-O PBRM1 shRNA lines. Using

Galaxy’s deepTools, we performed k-means clustering of the ARID2 binding enrichment in the 786-O

PBRM1 shRNA lines. K was set equal to 5, and we looked at the regions surrounding all transcribed genes

(+/- 2kb) in the 786-O cells, as defined by the RNA-seq analysis in Chapter 2. A.) Left panel - average

binding enrichment for the 5 clusters identified in the PBRM1 shRNA #1 cells. The same cluster sets were

then applied to the PBRM1 control and shRNA #2 ChIP-seq results (right panels). B.) Left panel - average

binding enrichment for the 5 clusters identified in the PBRM1 shRNA #2 cells. The same cluster sets were

then applied to the PBRM1 control and shRNA #1 ChIP-seq results (right panels).

Page 85: Dissertation Cover pages v1.0 EDITS 122815

72

knockdown (middle panel). The downregulated genes also have an ARID2 binding plateau in the region

downstream of the TES in the control cells which is largely lost with PBRM1 knockdown. As a control, we

looked at the average binding profile among genes that we classified as unexpressed in the 786-O lines

(right panel) (see Chapter 2 for explanation). Although there was lower average binding in general for the

PBRM1 shRNA lines, no cell line displayed peaks of binding for this set of genes. As an additional

control, we performed the same analysis on the input DNA for these gene sets and saw no peak patterns

or major differences between the cell lines (Figure 4.10B).

As an alternative method to assess how ARID2 binding changes impacted gene expression, we

used the publicly available software package BETA (Binding and Expression Target Analysis) (Wang et

al., 2013). BETA makes predictions about the regulatory potential of a binding factor on a certain gene

based on the level of binding around that gene, taking into account all binding sites within a certain

distance from the TSS and their relative distances, and gene expression changes. It ranks genes based

on the regulatory potential of factor binding and on differential gene expression, and then calculates the

rank product (used as a p-value) to predict direct gene targets. It also ascertains whether factor binding

Figure 4.10 –ARID2 and input DNA average binding profiles for the top upregulated, top downregulated, and

unexpressed genes. A.) The average ARID2 binding score for each 786-O line was generated using Galaxy’s

deepTools for the top quartile of upregulated genes (left panel), the top quartile of downregulated genes (most

downregulated – middle panel), and genes that did not meet our threshold for expression (summed RPKM<10)

(right panel). The plots illustrate the average binding score from 2kb upstream of the TSS to 2kb downstream of the

TES. B.) As controls, the same plots as in (A) for the average input DNA scores.

Page 86: Dissertation Cover pages v1.0 EDITS 122815

73

upregulates and/or downregulates genes using a non-parametric statistical test comparing the binding

factor regulatory potentials for genes that are differentially expressed versus those that don’t change.

We used the 786-O Cuffdiff output from the RNA-seq analysis as the gene expression input file

(see Chapter 2). As binding region inputs, we separately used the four different gained or lost ARID2

peak files from Figure 4.7. Using the ARID2 differential peak files separately would allow us to discern

how ARID2 gain or loss contributed to gene expression changes. The BETA activating/repressive

function predictions of ARID2 peak changes are shown in Figure 4.11. The blue and red lines represent

the upregulated and downregulated genes, respectively, from the 786-O RNA-seq analysis. The dashed

line represents the genes whose expression did not change, which served as a background for

comparison. Genes are ranked based on the regulatory potential score of ARID2 binding, from high to

low, and are then cumulated for the different expression sets – the quicker the genes cumulate, the

sharper the rise of the curve, implying more of an association between ARID2 binding and the specified

expression change. The P values represent the significance of the upregulate or downregulate gene set

distributions as compared to the background set by the Kolmogorov-Smirnov test.

The results reveal that ARID2 peak changes are significantly correlated with gene expression

changes – in all four ARID2 peak datasets, upregulated and downregulated genes accumulated more

rapidly than static genes. The data also indicate that ARID2 binding loss or gain can lead to both

increased and decreased gene expression, suggesting that additional factors may be involved in

determining the direction of expression changes, such as the histone modification context or presence of

co-factors. For both PBRM1 hairpins, there was a slightly stronger association between losing ARID2

peaks and differential expression, particularly upregulation (Figure 4.11A and B, left panels). This may

reflect the fact that significantly more ARID2 peaks were lost than gained with PBRM1 knockdown. For

this reason, we were particularly interested in some of the putative direct gene targets that lose ARID2

binding and have increased expression. The top 50 of these genes from the BETA output are shown for

each hairpin in Figure 4.12. Among this set, we see many genes relevant to cancer, including some that

we had already noted in our RNA-seq meta-analysis, such as EGFL7, CA2, ID1, and JAG1 (Figure 2.11).

Although ALDH1A1 was not among the top 50 genes, it too was identified as a direct target gene in the

shRNA #1 and #2 lines, with rank products of 1.32e-03 and 1.76e-03, respectively.

Page 87: Dissertation Cover pages v1.0 EDITS 122815

74

Figure 4.11 – BETA analysis of ARID2 binding changes and differential expression. The publicly available

software program BETA (Binding and Expression Target Analysis) was used to assess the association between

ARID2 binding changes and differential gene expression. The 786-O Cuffdiff output from the RNA-seq analysis

was used as the differential gene expression input file, and the ARID2 peaks gained or lost from Figure 4.6 were

used separately for the binding region inputs. The BETA activating/repressive function prediction of ARID2 peak

changes for the PBRM1 shRNA #1 line (A) and shRNA #2 line (B) are shown. The blue and red lines represent

the upregulated and downregulated genes, respectively, from the 786-O RNA-seq analysis. The dashed line

represents the genes whose expression did not change, which served as a background for comparison. Genes

are ranked based on the regulatory potential score of ARID2 binding, from high to low, and are then cumulated

for the different expression sets – the quicker the genes cumulate, the sharper the rise of the curve, implying

more of an association between ARID2 binding and the specified expression change. The P values (in

parentheses) represent the significance of the upregulate or downregulate gene set distributions as compared to

the background set by the Kolmogorov-Smirnov test.

Page 88: Dissertation Cover pages v1.0 EDITS 122815

75

Figure 4.12 – BETA-identified direct

gene targets that lose ARID2 binding

and have increased gene expression

with PBRM1 knockdown. The top 50

BETA-identified direct gene targets that

lose ARID2 binding (DECREASED peaks)

and have increased gene expression

(upregulate) in each PBRM1 shRNA line

(#1 – left table; #2 – right table). The

genes are ordered by the rank product,

which BETA uses as a p-value.

Page 89: Dissertation Cover pages v1.0 EDITS 122815

76

ARID2 binding patterns at specific loci

We next looked at the ARID2 peak patterns around some of these genes using the UCSC

genome browser (Figure 4.13). ARID2 binding enrichment levels (compared to input DNA) for all peaks

(FDR<0.05) are shown. Around the ALDH1A1 locus, ARID2 binding patterns shift with PBRM1

knockdown (Figure 4.13A). In the control cells, ARID2 peaks can be found both upstream (~60kb) and

downstream (~35kb) of the gene TSS and TES, respectively. With PBRM1 knockdown, these peaks

seems to relocate closer to the gene body, and can be found around the TSS and within the gene body

for shRNA #2.

Broad shifts in ARID2 binding can also be seen around the EGFR and CA2 loci (Figure 4.13B

and C, respectively). ARID2 seems to bind widely around the EGFR locus in the control cells – peaks

can be found upstream, at the TSS, in the gene body, and downstream of the TES. With PBRM1

knockdown, many of these peaks are lost, and the bound ARID2 that remains can be found mostly in the

gene body. At the CA2 locus, ARID2 enriches around and downstream of the TES in the control cells. In

the PBRM1 knockdown cells, many of these downstream peaks are lost, particularly for shRNA #2.

For other genes, ARID2 appears bound around the TSS when PBRM1 is present. When PBRM1

is lost, ARID2 binding at the TSS is concomitantly lost. ID1, JAG1, and EGFL7 display this pattern of

binding (Figure 4.13D-F, respectively).

Lastly, for some genes with differential expression, such as ARID2 and IFI27, there was no

detectable ARID2 binding around the gene locus in any line (Figure 4.13G and H, respectively). This

pattern of binding suggests that these genes may not be under direct transcriptional control by PBRM1,

but instead may be affected through indirect means.

In total, these snapshots of ARID2 binding support our conclusions that ARID2 binding is broadly

shifted in the absence of PBRM1, although the exact pattern of change depends on the genomic context.

Page 90: Dissertation Cover pages v1.0 EDITS 122815

77

A.

B.

C.

Page 91: Dissertation Cover pages v1.0 EDITS 122815

78

E.

F.

D.

Page 92: Dissertation Cover pages v1.0 EDITS 122815

79

PBRM1 knockdown results in widespread increases in H3K4 trimethylation

As SWI/SNF mutation in cancer has previously been noted to result in altered histone

modifications, we also performed ChIP-seq on various histone marks in the 786-O lines to assess

whether PBRM1 knockdown changed the histone landscape. We performed native ChIP-seq for these

studies, where the cells are not fixed and mononucleosomes are isolated via the action of micrococcal

nuclease. This method is thought to yield higher resolution mapping of histone modifications on single

nucleosomes and to avoid non-specific signal from modifications on nearby or linked nucleosomes

(Schones et al., 2008) (Cuddapah et al., 2009). We probed for the following marks: H3K4Me3, a marker

H.

Figure 4.13 - ARID2 binding patterns at specific loci. ARID2 binding enrichment levels (compared to input

DNA) for all peaks (FDR<0.05) at specific loci, as visualized on the UCSC genome browser. For a specific gene,

the scales of binding enrichment have been set equal to each other across all lines. ARID2 binding around the

following genes are shown (A-H): ALDH1A1, EGFR, CA2, ID1, JAG1, EGFL7, ARID2, and IFI27.

G.

Page 93: Dissertation Cover pages v1.0 EDITS 122815

80

of active promoters; H3K4Me1, a marker of enhancers; H3K27Ac, a marker of active enhancers and

promoters; H3K27Me3, a repressive mark; and H3K9Ac, a marker of active enhancers and promoters.

We aligned our results with Bowtie 2 and performed peak-calling with MACS2 (Zhang et al.,

2008). To get an overall picture of histone mark changes with PBRM1 knockdown, we used the MACS2

tool bdgdiff, which identifies common, gained, and lost peaks between samples. For this analysis, we

treated the two PBRM1 shRNA lines as replicates and compared them to the control line. The results are

shown in Table 4.2, where the absolute number of common, gained, or lost peaks are listed. There was a

striking gain of H3K4Me3 peaks with PBRM1 knockdown (31,155 peaks), while 0 peaks were lost. There

was also a notable loss of H3K4Me1 and H3K9Ac peaks, with very few gains. For the H3K27Ac and

H3K27Me3 marks, very few gains or losses occurred with PBRM1 knockdown.

Peaks

Mark Common Gained Lost

H3K4Me3 2,067 31,155 0

H3K4Me1 78,151 515 9,987

H3K27Ac 1,906 8 151

H3K27Me3 8,087 138 122

H3K9Ac 16,472 1 1,029

We next asked how relevant these histone mark changes were to differential gene expression in

these cells. We ran BETA again, using the gained or lost peak files for each histone mark as the binding

factor datasets. Gained H3K4Me3 peaks were highly associated with expression changes (Figure

4.14A). Interestingly, both upregulated and downregulated genes showed this association (P=2.58e-128

and P=9.51e-184, respectively). As H3K4Me3 is traditionally associated with active transcription and

increased gene expression, these results suggest the involvement of other factors that could further

shape expression outcomes, such as the binding of inhibitory co-factors. H3K4Me3 loss could not be

investigated because there were no lost peaks.

Additionally, H3K4Me1 lost peaks were significantly associated with downregulated genes (Figure

4.14B). As this mark is traditionally associated with enhancers, its loss would be expected to result in

decreased gene expression. For all the other marks, peak changes showed no correlation with

Table 4.2 – Number of peak changes identified by

MACS2 bdgdiff for histone marks

Page 94: Dissertation Cover pages v1.0 EDITS 122815

81

differential expression in this system (Figure 4.15A-C). H3K9Ac gain could not be probed because there

was only 1 gained peak.

As changes in H3K4Me3 and H3K4Me1 seem to be the most relevant with PBRM1 knockdown in

these cells, we next looked at the top target genes with changes in these marks (gains for H3K4Me3, and

losses for H3K4Me1) and differential expression (Figure 4.16). Among the upregulated genes with

H3K4Me3 gains, we saw many previously noted genes, including CA2, ID1, JAG1, and ALDH1A1.

We next looked at the H3K4Me3 levels at the TSS of ALDH1A1, CA2, ID1, and JAG1 (Figure

4.17A-D, respectively). H3K4Me3 binding enrichment levels (compared to input DNA) are shown. For

these genes, the control cells were not devoid of H3K4Me3 binding, but instead had H3K4Me3

enrichment at the TSS. PBRM1 knockdown led to a strengthening of this signal, and/or extension into the

gene body. These changes are particularly apparent for ALDH1A1 and CA2: for ALDH1A1, the signal is

both strengthened at the TSS and the H3K4Me3 mark extends further into the gene body; while for CA2,

the level of H3K4Me3 is actually decreased at the TSS, but extension is increased. The expression

values from the RNA-seq analysis show similar patterns of change: these genes are expressed in the

control cells, but expression is enhanced following PBRM1 knockdown.

ARID2 is required for some of the pro-tumorigenic effects of PBRM1 deficiency

ALDH1A1 upregulation with PBRM1 deficiency requires ARID2

Having seen ARID2 upregulation and continued association with SWI/SNF components in

PBRM1 deficient settings, but with altered genomic binding, we next asked what role ARID2 played in the

pro-tumorigenic changes we previously associated with PBRM1 loss. When we knocked down ARID2

using three different siRNAs in the 786-O shRNA #1 cells, ALDH1A1 protein expression was no longer

detectable (Figure 4.18A). In the control cells, the little ALDH1A1 protein visible also disappeared with

ARID2 knockdown. PBRM1 protein levels declined after ARID2 knockdown in these cells, confirming

previous findings that ARID2 is required for PBAF-complex stability. We confirmed that the ALDH1A1

changes were happening at the mRNA level (Figure 4.18B). We saw similar declines in ALDH1A1

expression with ARID2 knockdown in the PBRM1 shRNA #2 cells (Figure 4.18C). In the A-704 EV line,

ARID2 knockdown likewise decreased ALDH1A1 protein expression, but not as completely as in the 786-

Page 95: Dissertation Cover pages v1.0 EDITS 122815

82

O shRNA lines (Figure 4.18D). Again, ARID2 knockdown in cells expressing PBRM1 (the A-704 WT line)

led to loss of PBRM1 expression, demonstrating that even exogenously expressed PBRM1 requires

ARID2 for stability.

Figure 4.14 – BETA analysis of H3K4Me3 and H3K4Me1 changes and differential

expression. BETA analysis as in Figure 4.11 for changes in the histone marks H3K4Me3 (A)

and H3K4Me1 (B). The gained or lost peak files generated from the bdgdiff analysis of

histone mark binding in the 786-O lines were used separately for the binding region inputs.

There were no H3K4Me3 lost peaks, so BETA could not be run for this condition.

Page 96: Dissertation Cover pages v1.0 EDITS 122815

83

Figure 4.15 – BETA

analysis of H3K27Ac,

H3K27Me3, and

H3K9Ac changes and

differential expression.

BETA analysis as in

Figure 4.11 for changes

in the histone marks

H3K27Ac (A),

H3K27Me3 (B), and

H3K9Ac (C). The

gained or lost peak files

generated from the

bdgdiff analysis of

histone mark binding in

the 786-O lines were

used separately for the

binding region inputs.

There was only 1 gained

H3K9Ac peaks, so BETA

could not be run for this

condition.

Page 97: Dissertation Cover pages v1.0 EDITS 122815

84

Figure 4.16 – BETA-identified direct gene targets with histone mark changes

in H3K4Me3 and H3K4Me1 and differential gene expression. The top 40

BETA-identified direct gene targets that gain H3K4Me3 peaks and have

upregulated or downregulated expression or lose H3K4Me1 and have

downregulated expression. The genes are ordered by the rank product, which

BETA uses as a p-value.

Page 98: Dissertation Cover pages v1.0 EDITS 122815

85

A.

D.

C.

B.

Figure 4.17 – H3K4Me3 binding patterns at specific loci. H3K4Me3 binding enrichment levels (compared to

input DNA) around the TSS of specific genes, as visualized on the UCSC genome browser. For a specific gene, the

scales of binding enrichment have been set equal to each other across all lines. H3K4Me3 binding around the

following genes are shown (A-D): ALDH1A1, CA2, ID1, and JAG1.

Page 99: Dissertation Cover pages v1.0 EDITS 122815

86

ARID2 is required for tumorsphere formation but not cell viability

We next tested whether ARID2 knockdown had any functional consequences. We knocked down

ARID2 in the 786-O control and PBRM1 shRNA #2 cells, and then checked their tumorsphere-forming

capacity (Figure 4.19). As seen previously, the PBRM1 shRNA cells formed significantly more

tumorspheres than the controls cells when transfected with control siRNA. With ARID2 knockdown, the

tumorsphere-forming capacity significantly declined for both cells lines. The results resembled the effects

of knocking down or inhibiting ALDH1A1.

We wondered whether ARID2 knockdown inhibited tumorsphere formation due to reduced cell

viability or growth rates. Synthetic lethal relationships have been found between some SWI/SNF

components, such as BRG1 and BRM, or ARID1A and ARID1B. However, that does not appear to be the

Figure 4.18 - ALDH1A1 upregulation with PBRM1 deficiency requires ARID2. A.) Western blot

analysis exploring effects of knocking down ARID2 in the 786-O control or PBRM1 shRNA #1 lines. A non-

targeting siRNA was used as a control (C); siRNAs #6, #7, and #9 targeted ARID2. B.) qRT-PCR analysis

of ALDH1A1 mRNA levels (relative to GAPDH) in the indicated cell lines with ARID2 knockdown. Error

bars represent standard deviations for n=3. The delta-delta-Ct method was used to analyze the data, with

786-O control shRNA and C siRNA set to 1. An ordinary one-way ANOVA correcting for multiple

comparisons comparing the C siRNA to the ARID2 siRNAs in each line were performed. C.) Western blot

analysis exploring effects of knocking down ARID2 in the 786-O control or PBRM1 shRNA #2 lines. D.)

Western blot analysis exploring effects of knocking down ARID2 in the A-704 EV and WT lines.

Page 100: Dissertation Cover pages v1.0 EDITS 122815

87

case for PBRM1 and ARID2, although ARID2 knockdown did slow growth and reduce cell viability

somewhat (Figure 4.20A and B). However, even with ARID2 knockdown, the cells still grew, and the

large majority were viable, with only a 10-20% reduction in viability compared to the control siRNA

transfected cells. These differences are unlikely to account for the large drop in tumorsphere-forming

capacity.

SNF5, BRG1, and BRM appear dispensable for ALDH1A1 upregulation with PBRM1 deficiency

We next tested whether other SWI/SNF subunits found in the PBAF complex were required for

ALDH1A1 upregulation with PBRM1 deficiency. Employing the same strategy as we did for ARID2, we

knocked down SNF5 or BRG1 with siRNA in 786-O control and PBRM1 shRNA #1 cells (Figure 4.21A

and B, respectively). It appeared that we achieved good levels of knockdown of both proteins. However,

unlike for ARID2, SNF5 and BRG1 seemed dispensable for the high levels of ALDH1A1 expression in the

PBRM1 knockdown line. In the control cells, the low levels of ALDH1A1 were still reduced with SNF5 or

BRG1 knockdown. Additionally, PBRM1 stability did not seem as dependent on SNF5 or BRG1. With

SNF5 knockdown, PBRM1 levels were reduced in the control cells, but not to the same degree as seen

with ARID2 knockdown. BRG1 knockdown did not impact PBRM1 levels in the control cells.

Figure 4.19 – ARID2 is required for tumorsphere formation. A.) Tumorsphere assays

in 786-O control and PBRM1 shRNA #2 lines. Cells were transfected with non-targeting

siRNA (C) or ARID2-targting siRNAs (#7 or #9) 24hrs before plating for tumorsphere

assay. n=3-6/condition; unpaired t-tests were used for statistical testing. B.) Western blot

analysis of ARID2 expression levels in same cells used for tumorsphere assay with ARID2

siRNA knockdown at time of plating.

Page 101: Dissertation Cover pages v1.0 EDITS 122815

88

Figure 4.20 – ARID2 knockdown has only minor effects on cell growth and viability. 786-O

control and PBRM1 shRNA #2 cells were plated on wells of 6-well plates, and transfected with the

indicated siRNA (NC=non-targeting siRNA; and “mock” transfected cells received no siRNA). At the

indicated timepoints, cells were trypsinized, resuspended in the presence of trypan blue, and then

viable and non-viable (trypan blue positive) cells were counted. The cell counts were used to

generate the total number of live cells (A) and cell viability (B) measures.

Page 102: Dissertation Cover pages v1.0 EDITS 122815

89

We also performed the SNF5 knockdown in the A-704 EV and WT lines (Figure 4.21E). As

before, we achieved good levels of knockdown in both lines. In the A-704 EV cells, SNF5 knockdown led

to an increase in ALDH1A1 expression. In the WT cells, no ALDH1A1 expression was detectable, and

SNF5 knockdown slightly reduced PBRM1 levels.

Although the ATPase subunit BRM has not previously been reported to bind in a complex with

ARID2, in light of the seeming dispensability of BRG1, we wondered whether a subunit switch of BRM for

BRG1 was occurring with PBRM1 deficiency. We tested this by knocking down BRM in the 786-O lines,

but again saw no changes in ALDH1A1 expression levels in the PBRM1 shRNA cells (Figure 4.21C). To

see if either ATPase subunit was required, we also performed the double-knockdown of BRM and BRG1,

but still saw no changes in ALDH1A1 expression in the PBRM1 knockdown line (Figure 4.21D).

As BRG1 knockdown did not affect ALDH1A1 protein levels in the PBRM1 shRNA cells, we next

tested how BRG1 knockdown impacted tumorsphere-forming capacity. Mirroring the effects on ALDH1A1

protein levels, BRG1 knockdown did not significantly alter tumorsphere formation in the PBRM1 shRNA

cells (Figure 4.22A). In the control cells, BRG1 siRNA #1 also had no impact, although siRNA #5 caused

increased tumorsphere formation.

In total, these findings suggest a specific requirement for ARID2 for increased ALDH1A1

expression in the PBRM1 deficient setting, perhaps acting outside of a traditional SWI/SNF complex or

activity. More work is needed to fully elucidate ARID2’s role in this setting.

ARID2 is also required for increased EGFR protein levels, but not for decreased IFI27 mRNA levels, seen

with PBRM1 knockdown

To see if the requirement for ARID2 extended beyond ALDH1A1 upregulation, we also tested the

effects of ARID2 knockdown on total EGFR protein levels, which were increased with PBRM1 deficiency,

and IFI27 mRNA levels, which were decreased. Like we saw for ALDH1A1, siRNA knockdown of ARID2

in the PBRM1 shRNA line led to reduced total EGFR protein, down to a level comparable to the control

cells (Figure 4.23A). Additionally, SNF5 or BRG1 knockdown in the PBRM1 shRNA cells had very little

impact on EGFR levels, again suggesting their dispensability to some of the pro-tumorigenic changes

associated with PBRM1 deficiency (Figure 4.23B and C).

Page 103: Dissertation Cover pages v1.0 EDITS 122815

90

However, ARID2 knockdown did not affect the decreased IFI27 mRNA levels in the PBRM1

shRNA cells – they remained at similarly low levels (Figure 4.23D). In the control cells, ARID2

knockdown led to significant decreases in IFI27 levels, although not to levels comparable to those seen in

PBRM1 shRNA cells. This finding, along with the ARID2 ChIP-seq results, suggests that different

mechanisms may underlie PBRM1-associated transcriptional changes.

Figure 4.21 - SNF5, BRG1, and BRM appear dispensable for ALDH1A1 upregulation with PBRM1 deficiency.

A.) Western blot analysis exploring effects of knocking down SNF5 in the 786-O control or PBRM1 shRNA #1 lines.

A non-targeting siRNA was used as a control (“C”); siRNAs #1 and #2 targeted SNF5. B.) Western blot analysis

exploring effects of knocking down BRG1 in the 786-O control or PBRM1 shRNA #1 lines. siRNAs #1 and #5

targeted BRG1. C.) Western blot analysis exploring effects of knocking down BRM in the 786-O control or PBRM1

shRNA #1 lines. siRNAs #1, #2, and #5 targeted BRM. D.) Western blot analysis exploring effects of dual knock

down of BRM and BRG1 in the 786-O control or PBRM1 shRNA #1 lines. BRM1 siRNA #5 and BRG1 siRNA #5

were used. E.) Western blot analysis exploring effects of knocking down SNF5 in the A-704 EV and WT lines.

Page 104: Dissertation Cover pages v1.0 EDITS 122815

91

Figure 4.22 – BRG1 is not required for tumorsphere formation. A.) Tumorsphere assays in

786-O control and PBRM1 shRNA #2 lines. Cells were transfected with non-targeting siRNA (C) or

BRG1-targting siRNAs (#1 or #5) 24hrs before plating for tumorsphere assay. n=3/condition;

unpaired t-tests were used for statistical testing. B.) Western blot analysis of BRG1 expression

levels in same cells used for tumorsphere assay with BRG1 siRNA knockdown at time of plating.

Figure 4.23 - ARID2 is also required for increased EGFR

protein levels, but not for decreased IFI27 mRNA levels, seen

with PBRM1 knockdown. A.) Western blot analysis exploring

effects of knocking down ARID2 in the 786-O control or PBRM1

shRNA #1 lines. A non-targeting siRNA was used as a control

(“C”); siRNAs #7 and #9 targeted ARID2. B.) Western blot analysis

exploring effects of knocking down SNF5 in the 786-O control or

PBRM1 shRNA #1 lines. siRNAs #1 and #2 targeted SNF5. C.)

Western blot analysis exploring effects of knocking down BRG1 in

the 786-O control or PBRM1 shRNA #1 lines. siRNAs #1 and #5

targeted BRG1. D.) qRT-PCR analysis of IFI27 mRNA levels

(relative to GAPDH) in the indicated cell lines with ARID2

knockdown. Error bars represent standard deviations for n=3. The

delta-delta-Ct method was used to analyze the data, with 786-O

control shRNA and C siRNA set to 1. An ordinary one-way

ANOVA correcting for multiple comparisons comparing the C

siRNA to the ARID2 siRNAs in each line were performed.

Page 105: Dissertation Cover pages v1.0 EDITS 122815

92

DISCUSSION

In this chapter, we explored the mechanisms by which PBRM1 deficiency leads to pro-

tumorigenic transcriptional changes. We first considered how PBRM1 deficiency would affect the larger

PBAF complex. We found that with PBRM1 knockdown in the 786-O and ACHN lines, ARID2 protein

levels increased, while expression of WT, but not cancer-associated mutant, PBRM1 in the A-704 line

lowered ARID2 levels (Figure 4.1). These changes were at least partly due to increased transcription of

ARID2 in the 786-O and A-704 cells, and PBRM1 mutant primary tumors also had increased ARID2

mRNA (Figure 4.2).

Immunoprecipitation experiments revealed that ARID2 was still capable of binding to other

SWI/SNF components (BRG1 and SNF5) without PBRM1 (Figure 4.3), confirming previous reports (Yan

et al., 2005). As this did not preclude the possibility that some ARID2 was binding outside of SWI/SNF

with PBRM1 deficiency, we also performed glycerol gradient fractionation of the 786-O lines (Figure 4.4).

In the control cells, the majority of ARID2 and PBRM1 eluted in the heaviest fractions, separate from

BRG1 and SNF5 elution, and thus presumably outside of the SWI/SNF complex. This is in line with a

previous study of SWI/SNF fractionation patterns in mouse brain nuclear extracts that also found most of

the PBRM1 eluting apart in the heaviest fractions (Lessard et al., 2007). The authors conjectured that the

PBRM1 in these fractions could be part of the mitotic machinery, as PBRM1 had previously been found to

localize to the kinetochore during mitosis (Xue et al., 2000). In our cells, with PBRM1 knockdown, we

saw a shift in the ARID2 elution pattern away from these heavy fractions and more into lighter fractions

also containing BRG1 and SNF5, but not into other fractions free of BRG1 and SNF5. The findings thus

far suggest that PBRM1 deficiency is compensated by increased ARID2 expression and possibly

increased ARID2 association with SWI/SNF components.

As the IP and glycerol gradient fractionation experiments included only the soluble part of nuclear

extracts, we also conducted cellular fractionation experiments to see if ARID2 cellular localization was

altered with PBRM1 knockdown (Figure 4.5). We saw no ARID2, or other SWI/SNF components, in the

cytosol in any line. With PBRM1 knockdown, we saw ARID2 levels increase in both the soluble and

insoluble nuclear fractions. However, we were not able to detect BRG1 in the insoluble nuclear fraction in

any cell line, and SNF5 localization here seemed to decrease with PBRM1 knockdown (more so in the

Page 106: Dissertation Cover pages v1.0 EDITS 122815

93

PBRM1 shRNA #2 line). These findings do not invalidate the IP and glycerol gradient results, but suggest

that more ARID2 may also transit to the insoluble nuclear fraction, perhaps outside of the SWI/SNF

complex, in the absence of PBRM1.

Based on its domain structure, and because it is one of the defining subunits of the PBAF

complex, PBRM1 is thought to help “target” the complex in the genome. This theory, however, has never

been tested. Because ARID2 is also a PBAF-specific subunit, and at least some fraction of it still binds to

SWI/SNF components after PBRM1 knockdown, ARID2 ChIP-seq with or without PBRM1 would help

determine PBRM1’s role in targeting. Our ARID2 ChIP-seq results suggest that PBRM1 does indeed

help target PBAF binding in the genome. With PBRM1 knockdown, we saw a general decline in ARID2

binding to the genome (Figures 4.6 and 4.7). Moreover, clustering analysis indicated broad shifts in

ARID2 binding patterns without PBRM1 (Figure 4.8). We also saw very little overlap in the binding

patterns between the two PBRM1 shRNA lines, suggesting some degree of randomness to ARID2

binding without PBRM1 (Figures 4.7, 4.9). These findings may also be explained by technical failures of

the ARID2 ChIP-seq in the PBRM1 shRNA lines, but we do not believe this to be the case. The shifts in

binding patterns compared to the control were present in both shRNA lines, de novo clustering analysis in

both these lines displayed patterns of binding, and gain or loss of ARID2 peaks in both shRNA lines

significantly correlated with gene expression changes (Figure 4.11).

We found that ARID2 binding changes with PBRM1 knockdown were related to gene expression

changes from our earlier RNA-seq analysis. For the top upregulated genes, the average ARID2 binding

profile revealed a peak immediately upstream of the TSS in the control cells that was diminished or lost

with PBRM1 knockdown (Figure 4.10). For the most downregulated genes, there was an ARID2 peak

immediately upstream of the TSS and a wider plateau of binding downstream of the TES in the control

cells; both these features were largely lost in the PBRM1 knockdown lines. We also saw an association

between ARID2 binding changes and differential gene expression using the program BETA. BETA

analysis indicated that both loss and gain of ARID2 binding altered gene transcription, and that ARID2

binding could both activate and repress genes (Figure 4.11). BETA analysis identified ALDH1A1 as a

direct gene target with loss of ARID2 binding and increased gene expression with PBRM1 knockdown,

although it was not among the top 50 genes identified. Among the top genes inferred as direct targets,

Page 107: Dissertation Cover pages v1.0 EDITS 122815

94

we saw EGFL7, CA2, ID1, and JAG1, again implicating PBRM1 in the regulation of various cancer-

related genes and pathways (Figure 4.12).

We also explored how PBRM1 knockdown altered the histone landscape. We saw a widespread

increase in H3K4Me3 peaks with PBRM1 knockdown in the 786-O cells (Table 4.2). BETA analysis

indicated that these gains were highly associated with differential gene expression, and identified

numerous cancer-associated genes that gained H3K4Me3 peaks and were upregulated, including CA2,

ID1, JAG1, and ALDH1A1 (Figure 4.14-4.17). Further studies are needed to determine whether this a

general mechanism of tumor promotion associated with PBRM1 deficiency. For example,

methyltransferases such as MLL1 that methylate H3K4 could be perturbed in the PBRM1 deficient setting

and changes in growth or tumorsphere formation could be investigated. Alternatively, one could perturb

the H3K4Me3 demethylases (KDM5A-C) and observe the resultant changes in growth and gene

expression. It is interesting to note that KDM5C (also known as JARID1C) is mutated in ~5% of ccRCC

cases, and its knockdown in 786-O has been associated with increased tumorigenic potential (Dalgliesh

et al., 2010) (Niu et al., 2012). Confirmation of these findings in other cells lines is also needed.

Interestingly, we saw very few changes in H3K27Me3 peaks and no association with differential

gene expression. This suggests that the antagonism seen between EZH2 and SWI/SNF subunits in other

cancer types may not extend to PBRM1 in ccRCC.

In the last section of this chapter, we found that ARID2 was required for some of the pro-

tumorigenic changes we previously associated with PBRM1 deficiency, such as the upregulation of

ALDH1A1 and EGFR (Figures 4.18 and 4.23), while it wasn’t needed for IFI27 downregulation. These

data, along with the ARID2 ChIP-seq results, suggests that different mechanisms may underlie PBRM1-

associated transcriptional changes. For some genes, such as ALDH1A1 and EGFR, in the presence of

PBRM1, ARID2 binding in the vicinity of the gene locus is inhibitory. With the loss of PBRM1, ARID2

binding around the gene is altered, and is now activating. In this setting, ARID2 is now required for

continued upregulation. For other genes with differential expression, such as ARID2 and IFI27, ARID2

doesn’t seem to bind in the vicinity with or without PBRM1. Additionally, ARID2 is not required for

differential expression, at least for IFI27 downregulation. These findings could be explained by an indirect

method of transcriptional regulation. For yet other genes, such as ID1, JAG1, and EGFL7, the ARID2

Page 108: Dissertation Cover pages v1.0 EDITS 122815

95

ChIP-seq suggests yet another mode of regulation. When PBRM1 is present, ARID2 binds at the TSS

and gene expression is repressed. When PBRM1 is lost, ARID2 binding is also lost, and expression

increases. It will be interesting to see whether expression of these genes is ARID2 dependent.

Unlike ARID2, SNF5 and BRG1 seemed dispensable for the ALDH1A1 and EGFR upregulation

seen with PBRM1 deficiency (Figure 4.18 and 4.23). There are a few possible explanations for why other

SWI/SNF subunits may not be required for differential gene expression of genes that require ARID2. For

example, after PBAF-complex relocation after PBRM1 loss, nucleosome remodeling could initially be

required, but after establishing a new context for transcription, could then become dispensable, except for

continued ARID2 binding. Alternatively, ARID2 could be acting in a more novel, uncharacterized manner.

In light of our histone ChIP-seq results, one novel function could be ARID2-specific recruitment or

exclusion of histone modifying enzymes, outside of nucleosome remodeling. These results could also be

explained by ineffective knockdown of SNF5 and BRG1 that we were unable to detect by Western Blot –

using alternative means of gene ablation, such as CRISPR, would help clarify the findings.

Page 109: Dissertation Cover pages v1.0 EDITS 122815

96

CHAPTER 5: Conclusions

Page 110: Dissertation Cover pages v1.0 EDITS 122815

97

In this research study, we investigated the mechanisms of tumor suppression by PBRM1 in clear

cell renal cell carcinoma (ccRCC). PBRM1, a defining subunit of the PBAF type of SWI/SNF complex, is

mutated in a variety of cancers, including in ~40% of ccRCCs, making it the second most highly mutated

gene in ccRCC (behind VHL). Although many recent studies have looked at how other SWI/SNF

components function in cancer control, relatively little is known about the tumor suppressive mechanisms

of PBRM1 in ccRCC.

We first found that PBRM1 deficiency enhanced the tumorigenic potential of cells. These growth

advantages, however, were only clearly apparent when the cells were grown under stressful conditions,

such as reduced serum or a 3-D culture environment. Future studies of PBRM1 tumor suppressor

functions, and the evaluation of therapies for targeting PBRM1-mutant cells, should bear this in mind.

By comparing gene expression differences in ccRCC cell lines and murine embryonic fibroblasts

(MEFs) with or without PBRM1, we discovered that PBRM1 regulated numerous cancer-related genes

and pathways. Recent studies of other SWI/SNF subunits and other classes of epigenetic modifiers have

come to similar conclusions. PBRM1 mutation in cancer would thus be expected to have wide-ranging

effects on a cell, and the targeting of any one specific downstream pathway might have limited efficacy.

We decided to further investigate PBRM1 regulation of ALDH1A1 expression. Numerous studies

have identified ALDH1A1 as a marker of tumor-initiating cells, although it is unclear if ALDH1A1 actually

promotes tumorigenicity and how it does so. Not much has been known about the regulation of

ALDH1A1 expression in cancer, although recent studies have posited upregulation by β-catenin or

smad4-mediated repression in prostate cancer cells (Cojoc et al., 2015) (Hoshino et al., 2015). Another

study has described inhibition of ALDH1A1 by post-translational acetylation in breast cancer cells, which

is reversed by SIRT2 action downstream of NOTCH signaling (Zhao et al., 2014). Our results indicate a

novel method of regulation. We found that PBRM1 deficiency led to ALDH1A1 upregulation in numerous

ccRCC cell lines and MEFs, and PBRM1 mutation in primary tumors was associated with higher

ALDH1A1 levels. Additionally, our results suggest that ALDH1A1 is more than just a marker of tumor-

initiating cells, but can increase the tumorigenic potential of cells. Inhibiting ALDH1A1 or knocking it

down in the context of PBRM1 deficiency reduced anchorage-independent growth, while over-expressing

it in the PBRM1-normal setting increased tumorsphere-forming capacity.

Page 111: Dissertation Cover pages v1.0 EDITS 122815

98

We additionally explored PBRM1 regulation of the EGFR and IFN pathways. PBRM1 acted to

decrease total EGFR protein levels and dampen downstream signaling. These changes had functional

consequences, as PBRM1 deficient cells grew faster in response to EGF stimulation, but did not create a

setting of oncogenic addiction, as PBRM1 deficient cells were also more resistant to EGFR inhibition.

Alternatively, PBRM1 deficiency reduced basal and IFNα-induced levels of IFI27, a pro-apoptotic

interferon response gene, and made cells more resistant to growth inhibition by IFNα. These findings

suggest that EGFR inhibition, which has largely been abandoned as a therapeutic option for metastatic

ccRCC due to lack of superiority over IFNα treatment, and IFNα therapy itself might have better clinical

outcomes in non-PBRM1 mutant tumors. Previously, ccRCC patients have not been routinely classified

based on molecular genetic criteria, although recent efforts suggest that this might be where the field is

headed.

Finally, we investigated the molecular mechanisms of how PBRM1 deficiency could alter

transcription, keeping in mind that it is one subunit of the larger PBAF complex. In agreement with

previous reports, we found that ARID2 was expressed and still bound to other SWI/SNF components in

the absence of PBRM1. In fact, in our ccRCC cell lines, we found that ARID2 mRNA and protein levels

increased with PBRM1 deficiency, and that more ARID2 may associate with BRG1 and SNF after PBRM1

knockdown. ARID2 ChIP-seq in these cell lines suggested that the remnant PBAF-like complex was

bound to fewer locations in the genome, and was broadly redistributed where it did bind. As has been

previously speculated, PBRM1 did indeed seem to function as a “targeting” subunit of PBAF. Both

increased and decreased ARID2 binding was associated with differential gene expression, of both

upregulated and downregulated genes, suggesting that the genomic context influences whether PBAF-

binding is activating or repressive. Interestingly, we also found that ARID2 was required for some of the

pro-tumorigenic changes associated with PBRM1 deficiency, such as upregulated ALDH1A1 and EGFR

levels, but not others, such as decreased IFI27 levels. Further investigation is needed to determine which

gene expression changes are ARID2-dependent, and what may be determining this dependency.

Based on these findings, and looking at patterns of ARID2 binding at specific loci, we propose

two models by which PBRM1 deficiency could lead to transcriptional changes (Figure 5.1). For some

genes, such as ALDH1A1 and EGFR, when PBRM1 is present, PBAF binds in the vicinity of the gene

Page 112: Dissertation Cover pages v1.0 EDITS 122815

99

and dampens transcription (Figure 5.1A). With PBRM1 deficiency, ARID2 still binds in a PBAF-like

complex, but binding around the gene is altered, and is now activating. In this setting, ARID2 is now

required for continued upregulation, potentially acting through novel mechanisms, such as the recruitment

of activating co-factors. For other genes, such as ID1, JAG1, and EGFL7, we propose another mode of

regulation (Figure 5.1B). When PBRM1 is present, PBAF binds at the TSS and gene expression is

repressed. When PBRM1 is lost, ARID2 binding around the gene is also lost, and expression

subsequently increases. It will be interesting to see if differential expression of genes fitting this model of

regulation is ARID2 dependent. For yet other genes with differential expression, such as ARID2 and

IFI27, ARID2 doesn’t seem to bind in the vicinity with or without PBRM1. Additionally, ARID2 is not

required for differential expression, at least for IFI27 downregulation. These findings could be explained

by an indirect method of transcriptional regulation.

ChIP-seq of various histone marks also indicated that the active promoter mark H3K4Me3 is

increasing across the genome with PBRM1 deficiency, and that these increases are highly associated

with differential gene expression. Further studies are needed to determine whether this a general

mechanism of tumor promotion associated with PBRM1 deficiency, and also how these gains are

associated with ARID2 differential binding.

Figure 5.1 – Proposed models of how PBRM1 deficiency leads to transcriptional changes. The PBAF

complex is illustrated as follows: PBAF-specific components PBRM1, ARID2, and BRD7 are colored red; core

SWI/SNF components BRG1, SNF5, BAF155, and BAF170 are colored blue; and the rest of the complex is

colored grey. PBAF components whose association in the complex were inferred from prior reports but were not

directly verified by co-IP are shaded translucently.

Page 113: Dissertation Cover pages v1.0 EDITS 122815

100

While we have identified some novel cancer-related genes and pathways regulated by PBRM1,

we believe PBRM1 exerts wide effects on transcription and other important PBRM1-regulated genes have

yet to be characterized. Moreover, much future work can be done to verify and expand upon the models

of transcriptional control we are proposing. We were unable to successfully perform ChIP-seq analysis of

PBRM1 – doing so will greatly aid in the discovery and verification of genes directly regulated by PBRM1.

This analysis could also be compared to our ARID2 ChIP-seq results. ChIP-seq analysis of other

SWI/SNF components, such as BRG1 or SNF5, can also help determine if a PBAF-like complex is truly

being redirected with PBRM1 deficiency, or if it is just ARID2. Lastly, verification of our findings in other

ccRCC cell lines, and potentially other lineages of cancer, will help establish the veracity of our proposed

models of PBRM1 transcriptional control. Ultimately, we hope that a better understanding of the

mechanisms of PBRM1 tumor suppression in ccRCC will assist in the discovery of new treatment

paradigms.

Page 114: Dissertation Cover pages v1.0 EDITS 122815

101

MATERIALS AND METHODS

Cell lines

786-O, A-704, and ACHN cell lines were purchased from the ATCC. Murine embryonic

fibroblasts (MEFs) were generated from PBRM1 fl/fl mice using standard protocols. Normal culturing

conditions for each cell line were as follows: 786-O - RPMI 1640 medium supplemented with 10% (v/v)

fetal bovine serum (FBS), 100 IU penicillin, and 100ug/mL streptomycin (Cellgro); A-704 and ACHN –

MEM (Cellgro) supplemented with 10% FBS, 100 IU penicillin, and 100ug/mL streptomycin; MEFs –

DMEM (Cellgro) supplemented with 10% FBS, 1mM glutamine, 100 IU penicillin, and 100ug/mL

streptomycin. For reduced serum growth, the cells were washed 2x with PBS, and then switched to the

appropriate culture media with reduced FBS % (v/v). When treated with EGF (Peprotech), the cells were

starved for 16 hours (hr) in the appropriate media without FBS before treatment. IFNα was purchased

from Sigma (#SRP4596), erlotinib was purchased from Fisher (#50-148-625), and DEAB (dissolved in

95% ethanol) provided in the ALDEFLUOR kit (STEMCELL) was used.

Generation of PBRM1 fl/fl mice

In collaboration with the Transgenic Mouse Core at Columbia University, we designed a bacterial

artificial chromosome in which a LoxP site and a Frt-Neo-Frt-LoxP cassette were engineered to flank

exon 2 of the PBRM1 allele, such that Cre-mediated recombination would result in a frame-shifting

mutation. A gene-targeting vector was then constructed with a 2kb short homology arm, a 5kb long

homology arm, and a DTA negative selection marker. This vector was transfected into KV1 (129B6

hybrid) ES cells, which were then selected for recombination with G418. Two of the positively selected

ES cells were then injected into C57BL/6 blastocysts to generate chimeric male mice. Mice that were

positive for germline transmission of the modified allele were bred to homozygous ACTB (Flpe/Flpe)

females (in a C57BL/6 background) to remove the Neo cassette. Heterozygous mice (fl/+) derived from

the same initial chimeric mouse were bred to generate homozygous PBRM1 floxed mice (fl/fl).

Page 115: Dissertation Cover pages v1.0 EDITS 122815

102

Plasmids and constructs

Full-length PBRM1 was cloned into a pBABEpuro vector as previously described (Xia et al.,

2008). A C-terminal V5 tag was added to this plasmid using site-directed mutagenesis, and this plasmid

was then used as a template to make the PBRM1 cancer mutants. The ALDH1A1-HA vector in pcDNA

was purchased from Addgene (#11610) and was produced by the Steven Johnson lab.

Mutagenesis

The QuikChange II XL Site-Directed Mutagenesis Kit (Agilent) was used. Primers were designed

using the Agilent website for site-directed mutagenesis and are listed below. Clones were sequenced to

confirm the presence of the engineered mutations and the absence of secondary mutations.

Table 6.1: PCR Site Directed Mutagenesis Primers:

Mutant Primer

PBRM1 T232P Forward: 5'-cctatagatctcaagcccattgcccagaggata-3'

Reverse: 5'-tatcctctgggcaatgggcttgagatctataggct-3'

PBRM1 A597D Forward: 5'-gctgatgttccggaatgacaggcactataatgagg-3'

Reverse: 5'-cctcattatagtgcctgtcattccggaacatcagc-3'

PBRM1 H1204P Forward: 5'-acatttttgtgggctcaggctctgtttcttcaggg-3'

Reverse: 5'-ccctgaagaaacagagcctgagcccacaaaaatgt-3'

PBRM1 Ile57del Forward: 5'-gccatgaactctataatacccgagactataaggatgaaca-3'

Reverse: 5'-tgttcatccttatagtctcgggtattatagagttcatggc-3'

PBRM1 1204 6AA del Forward: 5'-cagagcatgagcccacaaaagtatttctgagtaatctgga-3'

Reverse: 5'-tccagattactcagaaatacttttgtgggctcatgctctg-3'

Transfection and infection

Transfections were done using Lipofectamine 2000 (Invitrogen). Retrovirus was produced by

transfection of pBABE constructs into the Phoenix HEK-293 packaging cell line (Swift et al., 2001).

Transduced A-704 cells were selected in 2ug/mL puromycin for 10-14 days.

Page 116: Dissertation Cover pages v1.0 EDITS 122815

103

For PBRM1 knockdown experiments, MISSION Lentiviral Transduction Particles were purchased

from Sigma-Aldrich (clone IDs: TRCN0000235890 and TRCN0000015994) and used according to the

manufacturer’s instructions. Briefly, 1x105 (786-O) or 2x105 (ACHN) cells were transduced with the viral

particles at a MOI = 3, along with a final concentration of 12 μg/mL polybrene. 786-O cells were selected

with 3 μg/mL puromycin, and ACHN cells with 1 μg/mL puromycin, for 10-14 days. Non-targeting control

shRNA (Sigma, SHC016) lentiviruses were produced in HEK-293T cells (Lois et al., 2002).

At passage 2, MEFs were infected with either control GFP adenovirus (Vector Biolabs, #1060) or

Cre adenovirus (Vector Biolabs, #1045) in 6-well plates. MEFs were passaged 3 times, and then mRNA

was collected for microarray and qRT-PCR analysis, protein was collected, and growth curve analysis

was performed.

For siRNA knockdown experiments, siRNA sets were purchased from Qiagen: ALDH1A1 –

GS216; ARID2 - GS196528; BRG1 - GS6597; SNF5 - GS6598; BRM - GS6595. The indicated siRNAs

from each set were used for experiments. Sigma MISSION siRNA Universal Negative Control #1

(SIC001) was used as negative control siRNA.

Immunoblotting

Protein lysates from cell lines was were collected by scraping the tissue culture plate with 2X

Sample Buffer (125mMTris-HCl pH 6.8, 4% SDS, 20% glycerol, 0.05% Bromophenol Blue, 8M urea, and

10% BME added fresh), subjecting the lysates to 5x 1 second bursts of sonication, boiling for 5 minutes

(min), and then centrifuging at 14,000 rpm for 5 min at 4⁰C. Samples were separated by SDS-

polyacrylamide gel electrophoresis (SDS-PAGE) on Tris-Glycine precast gels (Invitrogen) and then

transferred to PVDF membrane using wet transfer methods. After probing with primary and secondary

antibodies, proteins were detected using enhanced chemiluminescence (Thermo Scientific, #34080)

Page 117: Dissertation Cover pages v1.0 EDITS 122815

104

Antibodies

The following antibodies were used (Table 6.2):

Protein Target Purpose Company Catalogue # Type

Concentration

PBRM1* WB in-house -- rabbit polyclonal 1:5000

ARID2 WB/ChIP-seq

Santa Cruz Biotechnology

sc-1666117 (E-3)

mouse monoclonal

1:500 for WB; 5ug/ChIP

BRG1 WB Santa Cruz Biotechnology sc-8749 (N-15) goat polyclonal 1:500

BRG1 IP Santa Cruz Biotechnology sc-17796 (G-7)

mouse monoclonal 3ug per IP

SNF5 WB Bethyl Laboratories A301-087A-1 rabbit polyclonal 1:5000

BRM WB Santa Cruz Biotechnology sc-6450 (N-19) goat polyclonal 1:500

ALDH1A1 WB Abcam ab52492 (EP1933Y)

rabbit monoclonal 1:1000

H3 WB Abcam ab1791 rabbit polyclonal 1:10000

MYC WB Cell Signaling Technology 5605 (D84C12)

rabbit monoclonal 1:1000

Lamin B1 WB ThermoFisher Scientific 33-2000 (L-5)

mouse monoclonal 1:1000

Actin WB Sigma-Aldrich A4700 mouse monoclonal 1:40000

Tubulin WB Covance MMS-410P (Tu27)

mouse monoclonal 1:50000

Vinculin WB Sigma-Aldrich V9131 mouse monoclonal 1:50000

p-EGFR (Y1173) WB

Cell Signaling Technology 4407

rabbit monoclonal 1:1000

EGFR WB Cell Signaling Technology 4276 (D38B1)

rabbit monoclonal 1:1000

p-AKT (T308) WB

Cell Signaling Technology 4056 (244F9)

rabbit monoclonal 1:1000

p-AKT (S473) WB

Cell Signaling Technology 3787

rabbit monoclonal 1:1000

AKT WB Cell Signaling Technology 9272

rabbit polyclonal 1:1000

p-EKR WB Cell Signaling Technology 9101

rabbit polyclonal 1:1000

ERK WB Cell Signaling Technology 9102

rabbit polyclonal 1:1000

V5 agarose affinity gel IP Sigma-Aldrich A7345

mouse monoclonal 50ul per IP

H3K4Me3 ChIP-seq Abcam ab8580 rabbit polyclonal 3ug/ChIP

H3K4Me1 ChIP-seq Abcam ab8895 rabbit polyclonal 3ug/ChIP

H3K9Ac ChIP-seq Abcam ab10812 rabbit polyclonal 3ug/ChIP

Page 118: Dissertation Cover pages v1.0 EDITS 122815

105

H3K27Ac ChIP-seq Millipore 07-360 rabbit polyclonal 3ug/ChIP

H3K27Me3 ChIP-seq Millipore 07-449 rabbit polyclonal 3ug/ChIP

* PBRM1 polyclonal antibodies were generated against a glutathione S-transferase fusion protein of a C-

terminal PBRM1 fragment (amino acids 736–1,475).

Quantitative reverse transcription polymerase chain reaction (qRT-PCR)

Total RNA was prepared using the Qiagen RNeasy kit. Approximately 1 ug of RNA was used to

generate cDNA using Superscript Reverse Transcriptase II (Invitrogen). qRT-PCR was performed in an

ABI 7500 machine, using Fast SYBR Green (Invitrogen) according to the manufacturer’s instructions. The

primer sequences were as follows: PBRM1 forward, 5′- TGGAACCTGCAGAGGCCAACC -3′, and

reverse, 5′- TTTTCACCAGCTGAATCCTCCCACA -3′; ALDH1A1 forward, 5′-

TTGGAAGATAGGGCCTGCAC -3′, and reverse, 5′- GGAGGAAACCCTGCCTCTTTT -3′; IFI27 forward,

5′- ATCAGCAGTGACCAGTGTGG -3′, and reverse, 5′- GGCCACAACTCCTCCAATCA -3′; ARID2

forward, 5′- TACTTGCTAATGCCGGGGTG -3′, and reverse, 5′- ACGATGTCTTTCCAAAACTTAACGA -3′;

GAPDH forward, 5′-GAAGGTGAAGGTCGGAGTCAAC-3′, and reverse, 5′-

CAGAGTTAAAAGCAGCCCTGGT-3′. Samples were run in triplicate. The efficiencies of the primers were

measured using standard curves and were calculated to be: PBRM1 – 2.00; ALDH1A1 – 2.03; IFI27 –

1.99; ARID2 – 1.98; and GAPDH – 2.00. Quantitative analysis was performed on the basis of the ΔΔCt

method using the housekeeping gene GAPDH.

Nuclear extraction

Nuclear extraction was performed on 786-O and A-704 cells prior to the immunoprecipitation and

glycerol gradient experiments. For each cell line, 5 15cm plates (80-90% confluent) were collected at

once. Each plate was washed 2x in cold PBS, and then was collected by scraping into cold PBS

containing protease inhibitors (Sigma, P8340). Cells were spun at 700xg at 4⁰C for 5 min. The pellet was

then resuspended by pipetting and vortexing in 3.5mL of cold Buffer A (10mM HEPES-KOH pH 7.9,

10mM KCl, 0.1mM EDTA, 0.1mM EGTA), supplemented with fresh dithiothreitol (DTT) at 1mM and

protease inhibitors. After 5 min, 10% Triton X-100 in Buffer A (pre-made) was added 1:20 to a final

Page 119: Dissertation Cover pages v1.0 EDITS 122815

106

concentration of 0.5%, and the sample was vortexed again. After another 5 min, the sample was

centrifuged at 1000xg at 4⁰C for 5 min. The pellet (nuclei) was then washed once by resuspending in

1mL of cold Buffer D (20mM HEPES-KOH pH 7.9, 1mM EDTA, 1mM EGTA), supplemented with fresh

1mM DTT and protease inhibitors. The sample was centrifuged again at 1000xg at 4⁰C for 5 min, and the

pellet was now resuspended in 0.5mL of cold Buffer C (20mM HEPES-KOH pH 7.9, 400mM NaCl, 1mM

EDTA, 1mM EGTA), supplemented with fresh 1mM DTT, protease inhibitors, and 1mM sodium

orthovanadate. The sample was vortexed, passed numerous times through a 18.5G syringe, and then

incubated on ice for 5 min. 10% Triton X-100 in Buffer C (pre-made) was added 1:20 to a final

concentration of 0.5%, and the sample was again vortexed, passed numerous times through a 18.5G

syringe, and then incubated on ice for 5 min. The sample was then centrifuged at 14,000 rpm at 4⁰C for

10 min. The supernatant (nuclear extract) was removed, and the extraction was repeated on the pellet,

beginning with resuspension in 0.5mL of Buffer C. The second nuclear fraction was then combined with

the first, and then 0.6 volumes of cold Buffer D, supplemented with fresh 1mM DTT and protease

inhibitors, was added to lower the solute concentration. The nuclear extract was then filtered through a

0.22um filter using a 1mL syringe, a small aliquot was set aside to quantify the protein concentration

using the Bradford protein assay, and the nuclear extract was snap frozen and stored at -80⁰C.

Immunoprecipitation

Immunoprecipitation (IP) experiments were performed on isolated nuclear extracts. 0.5mg of

total protein was diluted to 1mL total volume using cold Buffer C/D (1.0:0.6 ratio), supplemented with

fresh 1mM DTT, protease inhibitors, and 1mM sodium orthovanadate. For each BRG1 IP, 3ug of BRG1

antibody (G-7) or mouse IgG (as a control) (Santa Cruz, #sc-2025) was prebound to 30ul Protein G

Dynabeads (ThermoFisher) by nutating for 2-4 hr at 4⁰C in PBS plus 0.02% Tween-20. The beads were

then washed once in 200ul PBS plus 0.02% Tween-20, and then resuspended in 30ul cold Buffer C/D,

supplemented with fresh 1mM DTT, protease inhibitors, and 1mM sodium orthovanadate for use in the IP

reaction.

The nuclear extract was pre-cleared using 30ul Protein G Dynabeads and nutating for 1hr at 4⁰C.

An aliquot to serve as the input control was removed (10ul), and then the sample was divided in two for

Page 120: Dissertation Cover pages v1.0 EDITS 122815

107

incubation with BRG1-bound beads or control IgG-bound beads. The samples were nutated overnight at

4⁰C, and then using a magnet the magnetic beads were washed 4x with 1mL cold Buffer C/D,

supplemented with fresh 1mM DTT, protease inhibitors, and 1mM sodium orthovanadate. For the last

wash, the resuspended beads were moved to a fresh tube. After the last wash, the beads were

resuspended in 60ul of 2X sample buffer, boiled for 5min, and then the sample was placed back on the

magnet and the sample buffer supernatant was transferred to a fresh tube and subjected to Western blot

analysis.

The V5 IP experiments in the A-704 lines were performed similarly, except 50ul V5-agarose

affinity gel was used for the IP, and 50ul of protein A/G agarose beads (Santa Cruz, #sc-2003)) and 4ug

mouse IgG was used for the pre-clear. Beads were isolated during the wash steps by centrifuging at

200xg for 3 min at 4⁰C.

Growth curve analysis

Experiments were carried out in 48-well plates at least in triplicate (see figure legends for n-sizes

for individual experiments). For 786-O and ACHN, 4 × 103 cells were plated per well, for A-704, 6 × 103

cells were plated per well, and for MEFs, 2 × 103 cells were plated per well. At the indicated time

intervals, cells were washed once with PBS, stained with 0.05% crystal violet in formalin at room

temperature for 20–30 min, and washed again twice with PBS before extracting the dye with 10% acetic

acid. Plates were shaken on a microplate shaker at medium speed for at least 1hr, and the absorbance

(O.D.), corresponding to cell numbers attached on the plates, was measured at 595 nm on a multi-well

plate reader (Bio-Rad).

Alternatively, cell growth in some experiments was monitored using the IncuCyte live cell imaging

system (Essen BioScience), which was placed in a cell culture incubator operated at 37°C and 5% CO2.

Cell confluence was determined using calculations derived from phase-contrast images.

Colony formation in soft-agar

1 × 104 cells were used per plate, and experiments were performed on all samples in triplicate.

Agar (Difco) was resuspended to 3% (w/v) in PBS and sterilized. 1mL of agar diluted to 0.5% in the

Page 121: Dissertation Cover pages v1.0 EDITS 122815

108

appropriate culture media (warm) was aliquoted into a 35mm plate as a support layer, which was then

allowed to dry in the 37°C incubator. Healthy-looking cells were then trypsinized and counted,

appropriate cell numbers were spun down, and then resuspended in warm media to a single-cell

suspension. When drug treatments were used (e.g., DEAB, erlotinib), they were mixed into the media at

this stage. The liquid 3% agar (kept warm at 60⁰C in a heat block) was added moderately quickly to the

cell mixture to a final concentration of 0.3%, the solution was thoroughly mixed, and then 1mL of the

cell:agar solution was layered onto the support layer (creating a single, uniform layer without agar

chunks). The agar plates were placed into larger plates containing some sterile water to prevent the

plates from drying out, and then were placed in the 37°C incubator to allow colonies to form for 23 days,

with careful monitoring every 3-4 days.

To quantify colony formation, the cells were stained with 1mL of 0.005% crystal violet solution in

formalin for 1 hr. The staining solution was carefully removed, and then the plates were scanned and

colonies quantified using ImageJ software (with a minimum colony size cut-off of 10 pixels).

Xenografts

Cells used for xenograft experiments had been split a few days prior to use and were healthy

looking and still growing. 2 hr prior to injection, the cells were re-fed. They were then trypsinized,

counted, washed in media, resuspended in cold media, and then combined 1:1 with cold Matrigel

(Trevigen) to a concentration of 1x106 per 0.2mL and kept on ice. 6 week-old female athymic nude mice

were subcutaneously injected on their ventral flanks with 1x106 control and PBRM1 shRNA cells (opposite

flanks of the same mouse – total of 4 mice). Tumor xenograft growth was monitored, and after the tumors

were palpable (10 days), size was measured using calipers (length and width measurements were taken,

and volume calculated using the formula: (length x width2)/2). When tumor size exceeded the limitations

specified by IACUC (>1000mm3), the mice were euthanized. All mice were treated humanely according

to the guidelines established by IACUC.

After euthanasia, tumors were harvested and weighed. For Western blot analysis, small sections

were excised and placed in 500ul of cold RIPA buffer (50 mM Tris-HCl pH 7.4, 1% NP-40, 0.25%

NaDeoxycholate, 150 mM NaCl, 1 mM EDTA), supplemented with fresh protease inhibitors, in pre-chilled

Page 122: Dissertation Cover pages v1.0 EDITS 122815

109

12x75mm borosilicate glass tubes. The excised xenografts were then homogenized for ~10 seconds,

until no chunks were visible. The mixture was then sonicated 5x with 1 second bursts, and was allowed

to sit on ice for 30 min, vortexing every 5 min. The solution was transferred to Eppendorf tubes, spun at

4⁰C for 20 min at maximum speed, the supernatant was transferred to a new tube, and then the spin step

was repeated. The supernatant was again transferred to a fresh tube, and was subjected to the Bradford

protein assay. 20ug of protein was then combined with 2X sample buffer, and Western blot analysis was

performed.

ALDEFLUOR assay

The ALDEFLUOR assay kit was purchased from STEMCELL Technologies, and the

manufacturer’s instructions were followed. Briefly, cells that were ~80% confluent were trypsinized,

washed in PBS, and resuspended in the provided Assay Buffer, and then counted. 2x105 cells in 0.5mL

Assay Buffer were used per reaction, and 2.5ul of substrate per reaction was used for the 786-O cells,

and 5ul per reaction for the A-704 cells. The chemical inhibitor DEAB was added to the recommended

concentration of 15uM to the negative control sample before adding the substrate to set the negative

baseline of no enzyme activity. The cells were incubated with substrate for 40 min at 37⁰C, were

centrifuged and then resuspended in 0.5mL cold Assay Buffer, and kept on ice in the dark until

fluorescence was measured using a BD LSRFortessa cell analyzer and FACSDiva software. ALDH1-

activity was calculated by gating on live, single cells, establishing a negative baseline using the DEAB-

treated sample, and then measuring the % of cells that were positive for ALDH1-activity by green

fluorescence levels on the x-axis. This analysis was done using Cytobank.

Tumorsphere Assay

The tumorsphere assay described herein was derived from published protocols (Shaw et al.,

2012) (Ueda et al., 2013). Healthy-looking, growing cells were trypsinized, counted, and washed in

tumorsphere media (appropriate cell culture medium, penicillin, streptomycin, 1x B-27 Supplement

(Gibco, #17504-044), 20ng/mL EGF, and 20ng/mL bFGF (R&D, #233-FB-025)). Cells were then

resuspended to single-cell suspensions in tumorsphere media, and 4 x103 cells in 3mL were plated into

Page 123: Dissertation Cover pages v1.0 EDITS 122815

110

wells of ultra-low attachment 6-well plates (Costar, #3741). When drug treatments were added, they were

added to the single-cell suspensions with mixing before plating. Tumorsphere formation was quantified

after 3.5 days using phase contrast images at 100x of all cells in a well, and then analyzing the images

with ImageJ software. A grouping of cells was counted as a tumorsphere if it was >50um in diameter,

and represented a solid mass of cells, with indistinguishable cell borders.

To measure tumorsphere self-renewal, the tumorspheres were passaged at this point. All the

media was collected from a well, the cells were briefly spun down, and then resuspended in tumorsphere

media and counted. Cells were re-plated as before at roughly the same starting density (4 x103 cells in

3mL) – if the total cell count for a well was less than 6 x103 cells, all the cells went back into a single well,

but if the cell count was higher, the cells were divided into 2 wells. When tumorsphere formation was

quantified for these cells, tumorspheres from cells that had been divided between two wells were added

together (see (Shaw et al., 2012)).

For siRNA experiments, cells were transfected with the indicated siRNA for 24 hr before they

were plated for the tumorsphere assay. For the ALDH1A1 overexpression experiment, the cells were

transfected for 36 hr prior to plating. Extra cells that were not plated were spun down, combined with 2X

sample buffer, and then analyzed by Western Blot analysis to check for successful knockdown or

overexpression.

Subcellular fractionation

Cells were collected from a 10cm plate at ~80% confluence by scraping into cold PBS

supplemented with fresh protease inhibitors. The cells were pelleted by centrifuging at 700xg at 4⁰C for 5

min, and were washed twice with cold PBS plus protease inhibitors. The cells were lysed by adding

300ul of Buffer A (10 mM HEPES, pH 7.4, 10mM KCl, 100uM EDTA), supplemented with fresh protease

inhibitors and 1mM DTT. The cells were placed on ice for 15 min, and then 10% NP40 in Buffer A (pre-

made) was added 1:20 to a final concentration of 0.5%, the sample was vortexed for 5 seconds, and then

nutated at 4⁰C for 2 min. The sample was then centrifuged at 1000xg at 4⁰C for 5 min, and 200ul of the

supernatant was carefully taken from the top and stored as the cytoplasmic fraction. The rest of the

supernatant was removed, and the pellet consisting of the nuclei was resuspended in 100ul Buffer C (20

Page 124: Dissertation Cover pages v1.0 EDITS 122815

111

mM HEPES, pH 7.4, 400mM NaCl, 1mM EDTA), supplemented with fresh protease inhibitors and 1mM

DTT. The sample was taped to a vortexer and vortexed at moderate speed (setting just below 6) for 15

min at 4⁰C. The sample was then centrifuged at top speed at 4⁰C for 15 min to collect the supernatant as

the soluble nuclear fraction. The pellet was the insoluble nuclear fraction. To resuspend it, 40ul of 2X

sample buffer was added, the sample was vortexed vigorously, sonicated 5x with 1 second bursts, boiled

for 15 min, and then centrifuged again at top speed at 4⁰C for 5 min. The supernatant was run on a gel

as the insoluble nuclear fraction. For the other fractions, 2X sample buffer was added 1:1, the samples

were boiled for 5 min, and then were ready for SDS-PAGE.

Glycerol gradient fractionation

Glycerol gradients were made using 10% and 30% glycerol solutions, to a total volume of 11.5

mL per gradient, using a dual piston gradient maker (Jule, J17) and 13.2mL thin-wall polypropylene tubes

(Beckman Coulter, 14x89mm, #331372). Glycerol solutions were made by diluting glycerol (v/v) into

Buffer C/D (1.0:0.6), supplemented with fresh 1mM DTT, protease inhibitors, and 1mM sodium

orthovanadate. The fractionation was performed on isolated nuclear extracts diluted to a final

concentration of 1.7mg/mL in cold Buffer C/D (1.0:0.6), supplemented with fresh 1mM DTT, protease

inhibitors, and 1mM sodium orthovanadate. 0.5 mL of the diluted nuclear extract was carefully layered on

top of the glycerol gradient. The gradients were spun for 18 hr in a TH-640 swinging bucket rotor at

40,700 rpm at 4⁰C in an ultracentrifuge. ~0.5mL fractions (9 drops) were collected from the bottom

(heavier fractions) of the tube (24 fractions total). Aliquots of the fractions were combined with 2X sample

buffer and subjected to Western blot analysis.

RNA-seq

Before RNA was collected, cells were passaged and grown at the specified serum levels for 48

hr. When RNA was collected, the cells were ~70% confluent and healthy-looking. Total RNA was

collected using the Qiagen RNeasy kit. Poly-A selection of mRNA, RNA-seq library preparation, and

100bp single-end sequencing using an Illumina HiSeq machine was performed by the Genomics Core

Facility at the Icahn Institute and Department of Genetics and Genomic Sciences. FastQC checks were

Page 125: Dissertation Cover pages v1.0 EDITS 122815

112

performed on the raw data, and then the Tuxedo suite of tools was used to align the reads to

GRCh37/hg19 and check for differential expression following the protocol outlined in Trapnell et al., 2012

(Trapnell et al., 2012). Downstream processing and analysis was performed as described in the text.

Microarray expression analysis

After infection with adenovirus, MEFs were passaged 3 times, and then total RNA was collected

for microarray analysis using the Qiagen RNeasy kit. The Ambion WT Expression Kit (#4411973) was

used to generate amplified sense-strand cDNA, and fragmentation and labeling was performed with the

GeneChip WT Terminal Labeling and Controls Kit (Affymetrix, #901525). Microarray expression analysis

was performed using GeneChip Mouse Gene 2.0 ST Arrays (Affymetrix, #902118) following the

manufacturer’s instructions.

ARID2 ChIP-seq

Cell fixation and sonication

786-O cells were grown to ~80% confluency on 15cm plates, the media was replaced with 10mL

cold PBS, and the cells were cross-linked with 1% formaldehyde (Pierce, #28908) for 5 min at 4⁰C with

gentle rocking. The fixation was stopped by the addition of glycine, the plates were washed 2x with cold

PBS, and then the fixed cells were collected into cold PBS supplemented with fresh protease inhibitors.

The output from 3-5 plates were pooled (enough to generate >18x106 fixed cells – 1 extra plate was set

aside to count and passage the cells). The cells were collected by centrifuging at 700xg for 5 min at 4⁰C,

and the pellets were snap frozen and stored at -80⁰C for less than two weeks.

To sonicate, the cell pellets were thawed on ice, and then were prepared for sonication using the

TruChIP Chromatin Shearing Reagant Kit (Covaris, # PN 520154) following the manufacturer’s

instructions. 3x106 cells in 130ul of D3 buffer (Covaris kit) were loaded into Covaris AFA microtubes, and

the cells were sonicated for 3.5 min on a Covaris M220 focused ultrasonicator (5% duty factor, 75W peak

incident power, 200 cycles/burst, 7°C). This level of sonication preserved the ARID2 epitope (as shown

by Western blot), and yielded chromatin ranging in size from 100-1000bp, with a peak around 500-700bp

Page 126: Dissertation Cover pages v1.0 EDITS 122815

113

(evaluated using Agilent Bioanalyzer 2100). For each cell line, 6 aliquots of cells were sonicated, and the

sonicated chromatin was pooled.

ChIP

Each sample was centrifuged at maximum speed for 20 min at 4⁰C, and a volume of supernatant

equivalent to 15x106 cells was transferred to a new tube. The total volume was raised to 1mL using cold

D3 buffer, and NaCl was added to raise the total salt concentration to 140mM. The sample was pre-

cleared by adding 30ul of Magna ChIP Protein A+G Magnetic Beads (Millipore, #16-663) and nutating at

4⁰C for 20 min. The beads were removed, and 10ul (1%) of the sample was removed and stored at -80⁰C

as the input DNA. To the rest of the sample, 5ug of ARID2 (E-3) antibody was added, and the

immunoprecipitation was carried out for 4 hr at 4⁰C. 50ul of Magna beads were then added, and a further

2 hr incubation at 4⁰C was carried out.

The magnetic beads were then isolated using a magnet, the unbound chromatin was removed,

and the beads were washed for 5 min in 1mL of each of the following buffers: Low salt wash buffer (0.1%

SDS, 1% Triton X-100, 2mM EDTA, 20mM Tris-HCl, pH 8.1, 150mM NaCl); High salt wash buffer (0.1%

SDS, 1% Triton X-100, 2mM EDTA, 20mM Tris-HCl, pH 8.1, 500mM NaCl); LiCl wash buffer (0.25M LiCl,

1% deoxycholate, 1% NP40, 1mM EDTA, 10mM Tris-HCl, pH 8.1); and two washes in TE buffer (10mM

Tris-HCl, pH 8.1, 1mM EDTA). For the last wash, the beads were moved to a new tube. After the last

wash, the wash buffer was removed and the beads were stored at -80⁰C overnight.

Beads were thawed, and precipitated chromatin was eluted in 150ul of elution buffer (1% SDS,

0.1M sodium bicarbonate) by shaking on a Thermomixer for 15 min at 37⁰C. This process was repeated

on the beads, and the eluted chromatin was pooled to 300ul. At this point, input DNA was thawed, and

the total volume was also brought up to 300ul using elution buffer. To reverse crosslinks, 12ul of 5M

NaCl was added, and the sample was incubated for 4 hr at 65°C. The sample was then treated with

RNase for 30 min at 37⁰C, and proteinase K for 2 hr at 45°C. DNA was extracted via phenol/chloroform

followed by DNA purification using Qiagen MiniElute PCR purification tubes. The amount and size of the

precipitated DNA was evaluated using the Bioanalyzer.

Page 127: Dissertation Cover pages v1.0 EDITS 122815

114

To generate enough DNA to make ChIP-seq libraries for sequencing, we repeated the entire

process on a different batch of cells for each cell line. The precipitated DNA was then pooled for each

cell line (ARID2-associated DNA from 30x106 cells total).

ChIP-seq library preparation and sequencing

ChIP-seq libraries were made following well-established protocols (see Hasson et al., 2013)

(Hasson et al., 2013). Briefly, 2-10ng of DNA was end-repaired, an A-overhang was added, Illumina

Truseq adapters were ligated, DNA was run on an agarose gel and size-selected at 350-500bp using the

Qiagen Gel Extraction Kit, and finally libraries were PCR amplified for 10-14 cycles using KAPA

Biosystems HiFi PCR Master Mix. Where not indicated, all library preparation steps involved New

England Biolabs enzymes. The quality and concentration of the prepared libraries were assessed on the

Bioanalyzer - the libraries produced U-shaped DNA profiles, with an average peak around 400bp. 75bp

single-end sequencing was performed on an Illumina NextSeq 500 machine.

ChIP-seq data processing and analysis

FastQC was run on the ChIP-seq raw data for quality control. Bowtie 2 was used to align reads

to the genome (GRCh37/hg19). The following settings were used (if not specified, the default settings

were applied): a seed length of 75 was used, a maximum of 2 mismatches were allowed, only uniquely

aligning reads were used, and the --best option was selected. After aligning, ENCODE-defined blacklisted

regions of the genome were removed.

SICER-df was used for ARID2 peak-calling and differential peak identification with the following

options: genome build – hg19; effective genome size – 0.793; redundancy threshold – 2; window size –

200; fragment size – 400; gap size – 600; and an FDR threshold of less than 0.05 for both peak and

differential peak identification. Input DNA was used as the control sample for each cell line. ARID2

enrichment levels were also generated using SICER, comparing the ARID2 signal to the input DNA.

HOMER’s annotatePeaks was used for genomic annotation. Galaxy’s deepTools was used to look at the

average enrichment profiles at specified regions and for clustering analysis. The genomic location of

Page 128: Dissertation Cover pages v1.0 EDITS 122815

115

specific genes was defined according to the human hg19 (GRCh37.p13) gene annotation of Ensembl

(gene database version 75), and was provided by the lab of Emily Bernstein.

Native ChIP-seq of histone marks

Native ChIP-seq was performed according to the detailed procedures outlined in Hasson et al.,

2013 (Hasson et al., 2013). Briefly, 20-50x106 cells were swelled and lysed using 0.1% IGEPAL, nuclei

were collected by centrifugation through a sucrose gradient, and micrococcal nuclease digestion was

optimized for each cell collection to primarily generate mononucleosomes, without overdigesting.

For the ChIP step, 3ug of each histone mark antibody was used, and 50ug of chromatin was used

for methyl marks, and 100ug was used for acetyl marks. The cells were precleared as for ARID2 (and

input DNA was collected), and the antibody incubation was performed overnight. 50ul of Magna beads

were then added, and a further 3 hr incubation at 4⁰C was carried out. Bead washing, processing of input

DNA, and extraction and purification of DNA was performed as in Hasson et al., 2013.

ChIP-seq libraries were prepared as described above, with the following changes: DNA was size

selected from between 300-400bp on the agarose gel to exclude polynucleosomes, which produced

amplified libraries with sharp peaks of ~280bp, as seen on the Bioanalyzer. Sequencing and ChIP-seq

data processing was performed as before. MACS2 was used for peak-calling, with the options --nomodel

--extsize 150, and with default q-value cutoffs (<0.01), except for the H3K27Me3, where the --broad

option was selected and the default q-value threshold is 0.1. For use in bdgdiff, the SPMR option was not

selected, but to generate pileups, it was turned on. The MACS2 tool bdgdiff was used with default

parameters. Pileups were generated using MACS2 for the default enrichment value (histone mark signal

over input DNA).

Statistical analysis

All statistical analysis was performed on GraphPad Prism 6 software. Statistical tests used for specific

experiments are described in the figure legends. In general, P<0.05 was considered significant.

Page 129: Dissertation Cover pages v1.0 EDITS 122815

116

REFERENCES

Albini, S., and Puri, P.L. (2010). SWI/SNF complexes, chromatin remodeling and skeletal myogenesis: it's time to exchange! Exp Cell Res 316, 3073-3080.

Armache, K.J., Garlick, J.D., Canzio, D., Narlikar, G.J., and Kingston, R.E. (2011). Structural basis of silencing: Sir3 BAH domain in complex with a nucleosome at 3.0 A resolution. Science 334, 977-982.

Barnes, D.W. (1982). Epidermal growth factor inhibits growth of A431 human epidermoid carcinoma in serum-free cell culture. J Cell Biol 93, 1-4.

Biankin, A.V., Waddell, N., Kassahn, K.S., Gingras, M.C., Muthuswamy, L.B., Johns, A.L., Miller, D.K., Wilson, P.J., Patch, A.M., Wu, J., et al. (2012). Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 491, 399-405.

Bitler, B.G., Aird, K.M., Garipov, A., Li, H., Amatangelo, M., Kossenkov, A.V., Schultz, D.C., Liu, Q., Shih Ie, M., Conejo-Garcia, J.R., et al. (2015). Synthetic lethality by targeting EZH2 methyltransferase activity in ARID1A-mutated cancers. Nat Med 21, 231-238.

Bourgo, R.J., Siddiqui, H., Fox, S., Solomon, D., Sansam, C.G., Yaniv, M., Muchardt, C., Metzger, D., Chambon, P., Roberts, C.W., et al. (2009). SWI/SNF deficiency results in aberrant chromatin organization, mitotic failure, and diminished proliferative capacity. Mol Biol Cell 20, 3192-3199.

Breeden, L., and Nasmyth, K. (1987). Cell cycle control of the yeast HO gene: cis- and trans-acting regulators. Cell 48, 389-397.

Brownlee, P.M., Chambers, A.L., Cloney, R., Bianchi, A., and Downs, J.A. (2014). BAF180 promotes cohesion and prevents genome instability and aneuploidy. Cell Rep 6, 973-981.

Burrows, A.E., Smogorzewska, A., and Elledge, S.J. (2010). Polybromo-associated BRG1-associated factor components BRD7 and BAF180 are critical regulators of p53 required for induction of replicative senescence. Proc Natl Acad Sci U S A 107, 14280-14285.

Cairns, B.R., Kim, Y.J., Sayre, M.H., Laurent, B.C., and Kornberg, R.D. (1994). A multisubunit complex containing the SWI1/ADR6, SWI2/SNF2, SWI3, SNF5, and SNF6 gene products isolated from yeast. Proc Natl Acad Sci U S A 91, 1950-1954.

Cairns, B.R., Lorch, Y., Li, Y., Zhang, M., Lacomis, L., Erdjument-Bromage, H., Tempst, P., Du, J., Laurent, B., and Kornberg, R.D. (1996). RSC, an essential, abundant chromatin-remodeling complex. Cell 87, 1249-1260.

Cancer Genome Atlas Research, N. (2013). Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature 499, 43-49.

Cancer Genome Atlas Research, N. (2014a). Comprehensive molecular characterization of gastric adenocarcinoma. Nature 513, 202-209.

Cancer Genome Atlas Research, N. (2014b). Comprehensive molecular characterization of urothelial bladder carcinoma. Nature 507, 315-322.

Carlson, M., and Laurent, B.C. (1994). The SNF/SWI family of global transcriptional activators. Curr Opin Cell Biol 6, 396-402.

Carrera, I., Zavadil, J., and Treisman, J.E. (2008). Two subunits specific to the PBAP chromatin remodeling complex have distinct and redundant functions during drosophila development. Mol Cell Biol 28, 5238-5250.

Page 130: Dissertation Cover pages v1.0 EDITS 122815

117

Cerami, E., Gao, J., Dogrusoz, U., Gross, B.E., Sumer, S.O., Aksoy, B.A., Jacobsen, A., Byrne, C.J., Heuer, M.L., Larsson, E., et al. (2012). The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov 2, 401-404.

Chanda, B., Ditadi, A., Iscove, N.N., and Keller, G. (2013). Retinoic acid signaling is essential for embryonic hematopoietic stem cell development. Cell 155, 215-227.

Chandrasekaran, R., and Thompson, M. (2007). Polybromo-1-bromodomains bind histone H3 at specific acetyl-lysine positions. Biochem Biophys Res Commun 355, 661-666.

Chen, T., and Dent, S.Y. (2014). Chromatin modifiers and remodellers: regulators of cellular differentiation. Nat Rev Genet 15, 93-106.

Chen, Y.C., Chen, Y.W., Hsu, H.S., Tseng, L.M., Huang, P.I., Lu, K.H., Chen, D.T., Tai, L.K., Yung, M.C., Chang, S.C., et al. (2009). Aldehyde dehydrogenase 1 is a putative marker for cancer stem cells in head and neck squamous cancer. Biochem Biophys Res Commun 385, 307-313.

Cheriyath, V., Leaman, D.W., and Borden, E.C. (2011). Emerging roles of FAM14 family members (G1P3/ISG 6-16 and ISG12/IFI27) in innate immunity and cancer. J Interferon Cytokine Res 31, 173-181.

Chi, T.H., Wan, M., Zhao, K., Taniuchi, I., Chen, L., Littman, D.R., and Crabtree, G.R. (2002). Reciprocal regulation of CD4/CD8 expression by SWI/SNF-like BAF complexes. Nature 418, 195-199.

Chong, C.R., and Janne, P.A. (2013). The quest to overcome resistance to EGFR-targeted therapies in cancer. Nat Med 19, 1389-1400.

Clifford, S.C., Prowse, A.H., Affara, N.A., Buys, C.H., and Maher, E.R. (1998). Inactivation of the von Hippel-Lindau (VHL) tumour suppressor gene and allelic losses at chromosome arm 3p in primary renal cell carcinoma: evidence for a VHL-independent pathway in clear cell renal tumourigenesis. Genes Chromosomes Cancer 22, 200-209.

Cojoc, M., Peitzsch, C., Kurth, I., Trautmann, F., Kunz-Schughart, L.A., Telegeev, G.D., Stakhovsky, E.A., Walker, J.R., Simin, K., Lyle, S., et al. (2015). Aldehyde Dehydrogenase Is Regulated by beta-Catenin/TCF and Promotes Radioresistance in Prostate Cancer Progenitor Cells. Cancer Res 75, 1482-1494.

Corominas-Faja, B., Oliveras-Ferraros, C., Cuyas, E., Segura-Carretero, A., Joven, J., Martin-Castillo, B., Barrajon-Catalan, E., Micol, V., Bosch-Barrera, J., and Menendez, J.A. (2013). Stem cell-like ALDH(bright) cellular states in EGFR-mutant non-small cell lung cancer: a novel mechanism of acquired resistance to erlotinib targetable with the natural polyphenol silibinin. Cell Cycle 12, 3390-3404.

Corti, S., Locatelli, F., Papadimitriou, D., Donadoni, C., Salani, S., Del Bo, R., Strazzer, S., Bresolin, N., and Comi, G.P. (2006). Identification of a primitive brain-derived neural stem cell population based on aldehyde dehydrogenase activity. Stem Cells 24, 975-985.

Cote, J., Quinn, J., Workman, J.L., and Peterson, C.L. (1994). Stimulation of GAL4 derivative binding to nucleosomal DNA by the yeast SWI/SNF complex. Science 265, 53-60.

Cuddapah, S., Barski, A., Cui, K., Schones, D.E., Wang, Z., Wei, G., and Zhao, K. (2009). Native chromatin preparation and Illumina/Solexa library construction. Cold Spring Harb Protoc 2009, pdb prot5237.

Curtis, B.J., Zraly, C.B., Marenda, D.R., and Dingwall, A.K. (2011). Histone lysine demethylases function as co-repressors of SWI/SNF remodeling activities during Drosophila wing development. Dev Biol 350, 534-547.

Page 131: Dissertation Cover pages v1.0 EDITS 122815

118

Dalgliesh, G.L., Furge, K., Greenman, C., Chen, L., Bignell, G., Butler, A., Davies, H., Edkins, S., Hardy, C., Latimer, C., et al. (2010). Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes. Nature 463, 360-363.

Desai, P., Guha, N., Galdieri, L., Hadi, S., and Vancura, A. (2009). Plc1p is required for proper chromatin structure and activity of the kinetochore in Saccharomyces cerevisiae by facilitating recruitment of the RSC complex. Mol Genet Genomics 281, 511-523.

Dontu, G., Abdallah, W.M., Foley, J.M., Jackson, K.W., Clarke, M.F., Kawamura, M.J., and Wicha, M.S. (2003). In vitro propagation and transcriptional profiling of human mammary stem/progenitor cells. Genes Dev 17, 1253-1270.

Drucker, B., Bacik, J., Ginsberg, M., Marion, S., Russo, P., Mazumdar, M., and Motzer, R. (2003). Phase II trial of ZD1839 (IRESSA) in patients with advanced renal cell carcinoma. Invest New Drugs 21, 341-345.

Dulak, A.M., Stojanov, P., Peng, S., Lawrence, M.S., Fox, C., Stewart, C., Bandla, S., Imamura, Y., Schumacher, S.E., Shefler, E., et al. (2013). Exome and whole-genome sequencing of esophageal adenocarcinoma identifies recurrent driver events and mutational complexity. Nat Genet 45, 478-486.

Dunaief, J.L., Strober, B.E., Guha, S., Khavari, P.A., Alin, K., Luban, J., Begemann, M., Crabtree, G.R., and Goff, S.P. (1994). The retinoblastoma protein and BRG1 form a complex and cooperate to induce cell cycle arrest. Cell 79, 119-130.

Dykhuizen, E.C., Hargreaves, D.C., Miller, E.L., Cui, K., Korshunov, A., Kool, M., Pfister, S., Cho, Y.J., Zhao, K., and Crabtree, G.R. (2013). BAF complexes facilitate decatenation of DNA by topoisomerase IIalpha. Nature 497, 624-627.

Euskirchen, G.M., Auerbach, R.K., Davidov, E., Gianoulis, T.A., Zhong, G., Rozowsky, J., Bhardwaj, N., Gerstein, M.B., and Snyder, M. (2011). Diverse roles and interactions of the SWI/SNF chromatin remodeling complex revealed using global approaches. PLoS Genet 7, e1002008.

Farnie, G., Clarke, R.B., Spence, K., Pinnock, N., Brennan, K., Anderson, N.G., and Bundred, N.J. (2007). Novel cell culture technique for primary ductal carcinoma in situ: role of Notch and epidermal growth factor receptor signaling pathways. J Natl Cancer Inst 99, 616-627.

Fillmore, C.M., Xu, C., Desai, P.T., Berry, J.M., Rowbotham, S.P., Lin, Y.J., Zhang, H., Marquez, V.E., Hammerman, P.S., Wong, K.K., et al. (2015). EZH2 inhibition sensitizes BRG1 and EGFR mutant lung tumours to TopoII inhibitors. Nature 520, 239-242.

Freedman, V.H., and Shin, S.I. (1974). Cellular tumorigenicity in nude mice: correlation with cell growth in semi-solid medium. Cell 3, 355-359.

Gao, J., Aksoy, B.A., Dogrusoz, U., Dresdner, G., Gross, B., Sumer, S.O., Sun, Y., Jacobsen, A., Sinha, R., Larsson, E., et al. (2013). Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 6, pl1.

Gartenberg, M. (2009). Heterochromatin and the cohesion of sister chromatids. Chromosome Res 17, 229-238.

Gerlinger, M., Horswell, S., Larkin, J., Rowan, A.J., Salm, M.P., Varela, I., Fisher, R., McGranahan, N., Matthews, N., Santos, C.R., et al. (2014). Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat Genet 46, 225-233.

Page 132: Dissertation Cover pages v1.0 EDITS 122815

119

Gerlinger, M., Rowan, A.J., Horswell, S., Larkin, J., Endesfelder, D., Gronroos, E., Martinez, P., Matthews, N., Stewart, A., Tarpey, P., et al. (2012). Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med 366, 883-892.

Ghiaur, G., Yegnasubramanian, S., Perkins, B., Gucwa, J.L., Gerber, J.M., and Jones, R.J. (2013). Regulation of human hematopoietic stem cell self-renewal by the microenvironment's control of retinoic acid signaling. Proc Natl Acad Sci U S A 110, 16121-16126.

Ginestier, C., Hur, M.H., Charafe-Jauffret, E., Monville, F., Dutcher, J., Brown, M., Jacquemier, J., Viens, P., Kleer, C.G., Liu, S., et al. (2007). ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell 1, 555-567.

Hasson, D., Panchenko, T., Salimian, K.J., Salman, M.U., Sekulic, N., Alonso, A., Warburton, P.E., and Black, B.E. (2013). The octamer is the major form of CENP-A nucleosomes at human centromeres. Nat Struct Mol Biol 20, 687-695.

Heinz, S., Benner, C., Spann, N., Bertolino, E., Lin, Y.C., Laslo, P., Cheng, J.X., Murre, C., Singh, H., and Glass, C.K. (2010). Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol Cell 38, 576-589.

Helming, K.C., Wang, X., and Roberts, C.W. (2014a). Vulnerabilities of mutant SWI/SNF complexes in cancer. Cancer Cell 26, 309-317.

Helming, K.C., Wang, X., Wilson, B.G., Vazquez, F., Haswell, J.R., Manchester, H.E., Kim, Y., Kryukov, G.V., Ghandi, M., Aguirre, A.J., et al. (2014b). ARID1B is a specific vulnerability in ARID1A-mutant cancers. Nat Med 20, 251-254.

Ho, L., Jothi, R., Ronan, J.L., Cui, K., Zhao, K., and Crabtree, G.R. (2009). An embryonic stem cell chromatin remodeling complex, esBAF, is an essential component of the core pluripotency transcriptional network. Proc Natl Acad Sci U S A 106, 5187-5191.

Hodis, E., Watson, I.R., Kryukov, G.V., Arold, S.T., Imielinski, M., Theurillat, J.P., Nickerson, E., Auclair, D., Li, L., Place, C., et al. (2012). A landscape of driver mutations in melanoma. Cell 150, 251-263.

Hoffman, G.R., Rahal, R., Buxton, F., Xiang, K., McAllister, G., Frias, E., Bagdasarian, L., Huber, J., Lindeman, A., Chen, D., et al. (2014). Functional epigenetics approach identifies BRM/SMARCA2 as a critical synthetic lethal target in BRG1-deficient cancers. Proc Natl Acad Sci U S A 111, 3128-3133.

Hoshino, Y., Nishida, J., Katsuno, Y., Koinuma, D., Aoki, T., Kokudo, N., Miyazono, K., and Ehata, S. (2015). Smad4 Decreases the Population of Pancreatic Cancer-Initiating Cells through Transcriptional Repression of ALDH1A1. Am J Pathol 185, 1457-1470.

Huang, C.P., Tsai, M.F., Chang, T.H., Tang, W.C., Chen, S.Y., Lai, H.H., Lin, T.Y., Yang, J.C., Yang, P.C., Shih, J.Y., et al. (2013). ALDH-positive lung cancer stem cells confer resistance to epidermal growth factor receptor tyrosine kinase inhibitors. Cancer Lett 328, 144-151.

Huang, M., Qian, F., Hu, Y., Ang, C., Li, Z., and Wen, Z. (2002). Chromatin-remodelling factor BRG1 selectively activates a subset of interferon-alpha-inducible genes. Nat Cell Biol 4, 774-781.

Iliopoulos, O., Kibel, A., Gray, S., and Kaelin, W.G., Jr. (1995). Tumour suppression by the human von Hippel-Lindau gene product. Nat Med 1, 822-826.

Iliopoulos, O., Levy, A.P., Jiang, C., Kaelin, W.G., Jr., and Goldberg, M.A. (1996). Negative regulation of hypoxia-inducible genes by the von Hippel-Lindau protein. Proc Natl Acad Sci U S A 93, 10595-10599.

Institute, N.C. (2015). Renal Cell Cancer Treatment - for health professionals (PDQ).

Page 133: Dissertation Cover pages v1.0 EDITS 122815

120

Isakoff, M.S., Sansam, C.G., Tamayo, P., Subramanian, A., Evans, J.A., Fillmore, C.M., Wang, X., Biegel, J.A., Pomeroy, S.L., Mesirov, J.P., et al. (2005). Inactivation of the Snf5 tumor suppressor stimulates cell cycle progression and cooperates with p53 loss in oncogenic transformation. Proc Natl Acad Sci U S A 102, 17745-17750.

Jelinic, P., Mueller, J.J., Olvera, N., Dao, F., Scott, S.N., Shah, R., Gao, J., Schultz, N., Gonen, M., Soslow, R.A., et al. (2014). Recurrent SMARCA4 mutations in small cell carcinoma of the ovary. Nat Genet 46, 424-426.

Jiao, Y., Pawlik, T.M., Anders, R.A., Selaru, F.M., Streppel, M.M., Lucas, D.J., Niknafs, N., Guthrie, V.B., Maitra, A., Argani, P., et al. (2013). Exome sequencing identifies frequent inactivating mutations in BAP1, ARID1A and PBRM1 in intrahepatic cholangiocarcinomas. Nat Genet 45, 1470-1473.

Jones, S., Wang, T.L., Shih Ie, M., Mao, T.L., Nakayama, K., Roden, R., Glas, R., Slamon, D., Diaz, L.A., Jr., Vogelstein, B., et al. (2010). Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science 330, 228-231.

Joseph, R.W., Kapur, P., Serie, D.J., Parasramka, M., Ho, T.H., Cheville, J.C., Frenkel, E., Parker, A.S., and Brugarolas, J. (2015). Clear Cell Renal Cell Carcinoma Subtypes Identified by BAP1 and PBRM1 Expression. J Urol.

Kadoch, C., and Crabtree, G.R. (2013). Reversible disruption of mSWI/SNF (BAF) complexes by the SS18-SSX oncogenic fusion in synovial sarcoma. Cell 153, 71-85.

Kadoch, C., Hargreaves, D.C., Hodges, C., Elias, L., Ho, L., Ranish, J., and Crabtree, G.R. (2013). Proteomic and bioinformatic analysis of mammalian SWI/SNF complexes identifies extensive roles in human malignancy. Nat Genet 45, 592-601.

Kakarougkas, A., Ismail, A., Chambers, A.L., Riballo, E., Herbert, A.D., Kunzel, J., Lobrich, M., Jeggo, P.A., and Downs, J.A. (2014). Requirement for PBAF in transcriptional repression and repair at DNA breaks in actively transcribed regions of chromatin. Mol Cell 55, 723-732.

Kal, A.J., Mahmoudi, T., Zak, N.B., and Verrijzer, C.P. (2000). The Drosophila brahma complex is an essential coactivator for the trithorax group protein zeste. Genes Dev 14, 1058-1071.

Kapur, P., Pena-Llopis, S., Christie, A., Zhrebker, L., Pavia-Jimenez, A., Rathmell, W.K., Xie, X.J., and Brugarolas, J. (2013). Effects on survival of BAP1 and PBRM1 mutations in sporadic clear-cell renal-cell carcinoma: a retrospective analysis with independent validation. Lancet Oncol 14, 159-167.

Kent, N.A., Chambers, A.L., and Downs, J.A. (2007). Dual chromatin remodeling roles for RSC during DNA double strand break induction and repair at the yeast MAT locus. J Biol Chem 282, 27693-27701.

Kia, S.K., Gorski, M.M., Giannakopoulos, S., and Verrijzer, C.P. (2008). SWI/SNF mediates polycomb eviction and epigenetic reprogramming of the INK4b-ARF-INK4a locus. Mol Cell Biol 28, 3457-3464.

Kiefer, F.W., Orasanu, G., Nallamshetty, S., Brown, J.D., Wang, H., Luger, P., Qi, N.R., Burant, C.F., Duester, G., and Plutzky, J. (2012a). Retinaldehyde dehydrogenase 1 coordinates hepatic gluconeogenesis and lipid metabolism. Endocrinology 153, 3089-3099.

Kiefer, F.W., Vernochet, C., O'Brien, P., Spoerl, S., Brown, J.D., Nallamshetty, S., Zeyda, M., Stulnig, T.M., Cohen, D.E., Kahn, C.R., et al. (2012b). Retinaldehyde dehydrogenase 1 regulates a thermogenic program in white adipose tissue. Nat Med 18, 918-925.

Kim, S., and Alexander, C.M. (2014). Tumorsphere assay provides more accurate prediction of in vivo responses to chemotherapeutics. Biotechnol Lett 36, 481-488.

Page 134: Dissertation Cover pages v1.0 EDITS 122815

121

Langmead, B., and Salzberg, S.L. (2012). Fast gapped-read alignment with Bowtie 2. Nat Methods 9, 357-359.

Larkin, J., Goh, X.Y., Vetter, M., Pickering, L., and Swanton, C. (2012). Epigenetic regulation in RCC: opportunities for therapeutic intervention? Nat Rev Urol 9, 147-155.

Latif, F., Tory, K., Gnarra, J., Yao, M., Duh, F.M., Orcutt, M.L., Stackhouse, T., Kuzmin, I., Modi, W., Geil, L., et al. (1993). Identification of the von Hippel-Lindau disease tumor suppressor gene. Science 260, 1317-1320.

Lee, R.S., Stewart, C., Carter, S.L., Ambrogio, L., Cibulskis, K., Sougnez, C., Lawrence, M.S., Auclair, D., Mora, J., Golub, T.R., et al. (2012). A remarkably simple genome underlies highly malignant pediatric rhabdoid cancers. J Clin Invest 122, 2983-2988.

Lemon, B., Inouye, C., King, D.S., and Tjian, R. (2001). Selectivity of chromatin-remodelling cofactors for ligand-activated transcription. Nature 414, 924-928.

Lessard, J., Wu, J.I., Ranish, J.A., Wan, M., Winslow, M.M., Staahl, B.T., Wu, H., Aebersold, R., Graef, I.A., and Crabtree, G.R. (2007). An essential switch in subunit composition of a chromatin remodeling complex during neural development. Neuron 55, 201-215.

Li, M., Zhao, H., Zhang, X., Wood, L.D., Anders, R.A., Choti, M.A., Pawlik, T.M., Daniel, H.D., Kannangai, R., Offerhaus, G.J., et al. (2011). Inactivating mutations of the chromatin remodeling gene ARID2 in hepatocellular carcinoma. Nat Genet 43, 828-829.

Liu, N., Long, Y., Liu, B., Yang, D., Li, C., Chen, T., Wang, X., Liu, C., and Zhu, H. (2014a). ISG12a mediates cell response to Newcastle disease viral infection. Virology 462-463, 283-294.

Liu, N., Zuo, C., Wang, X., Chen, T., Yang, D., Wang, J., and Zhu, H. (2014b). miR-942 decreases TRAIL-induced apoptosis through ISG12a downregulation and is regulated by AKT. Oncotarget 5, 4959-4971.

Lois, C., Hong, E.J., Pease, S., Brown, E.J., and Baltimore, D. (2002). Germline transmission and tissue-specific expression of transgenes delivered by lentiviral vectors. Science 295, 868-872.

Luger, K., Dechassa, M.L., and Tremethick, D.J. (2012). New insights into nucleosome and chromatin structure: an ordered state or a disordered affair? Nat Rev Mol Cell Biol 13, 436-447.

Ma, W., Tessarollo, L., Hong, S.B., Baba, M., Southon, E., Back, T.C., Spence, S., Lobe, C.G., Sharma, N., Maher, G.W., et al. (2003). Hepatic vascular tumors, angiectasis in multiple organs, and impaired spermatogenesis in mice with conditional inactivation of the VHL gene. Cancer Res 63, 5320-5328.

Maxwell, P.H., Wiesener, M.S., Chang, G.W., Clifford, S.C., Vaux, E.C., Cockman, M.E., Wykoff, C.C., Pugh, C.W., Maher, E.R., and Ratcliffe, P.J. (1999). The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 399, 271-275.

McKenna, E.S., Sansam, C.G., Cho, Y.J., Greulich, H., Evans, J.A., Thom, C.S., Moreau, L.A., Biegel, J.A., Pomeroy, S.L., and Roberts, C.W. (2008). Loss of the epigenetic tumor suppressor SNF5 leads to cancer without genomic instability. Mol Cell Biol 28, 6223-6233.

Medina, P.P., Romero, O.A., Kohno, T., Montuenga, L.M., Pio, R., Yokota, J., and Sanchez-Cespedes, M. (2008). Frequent BRG1/SMARCA4-inactivating mutations in human lung cancer cell lines. Hum Mutat 29, 617-622.

Mihalich, A., Vigano, P., Gentilini, D., Borghi, M.O., Vignali, M., Busacca, M., and Di Blasio, A. (2012). Interferon-inducible genes, TNF-related apoptosis-inducing ligand (TRAIL) and interferon inducible

Page 135: Dissertation Cover pages v1.0 EDITS 122815

122

protein 27 (IFI27) are negatively regulated in leiomyomas: implications for a role of the interferon pathway in leiomyoma development. Gynecol Endocrinol 28, 216-219.

Moshkin, Y.M., Mohrmann, L., van Ijcken, W.F., and Verrijzer, C.P. (2007). Functional differentiation of SWI/SNF remodelers in transcription and cell cycle control. Mol Cell Biol 27, 651-661.

Nakayama, T., Shimojima, T., and Hirose, S. (2012). The PBAP remodeling complex is required for histone H3.3 replacement at chromatin boundaries and for boundary functions. Development 139, 4582-4590.

Neigeborn, L., and Carlson, M. (1984). Genes affecting the regulation of SUC2 gene expression by glucose repression in Saccharomyces cerevisiae. Genetics 108, 845-858.

Nguyen, L.V., Vanner, R., Dirks, P., and Eaves, C.J. (2012). Cancer stem cells: an evolving concept. Nat Rev Cancer 12, 133-143.

Niu, X., Zhang, T., Liao, L., Zhou, L., Lindner, D.J., Zhou, M., Rini, B., Yan, Q., and Yang, H. (2012). The von Hippel-Lindau tumor suppressor protein regulates gene expression and tumor growth through histone demethylase JARID1C. Oncogene 31, 776-786.

Ohh, M., Park, C.W., Ivan, M., Hoffman, M.A., Kim, T.Y., Huang, L.E., Pavletich, N., Chau, V., and Kaelin, W.G. (2000). Ubiquitination of hypoxia-inducible factor requires direct binding to the beta-domain of the von Hippel-Lindau protein. Nat Cell Biol 2, 423-427.

Papoulas, O., Beek, S.J., Moseley, S.L., McCallum, C.M., Sarte, M., Shearn, A., and Tamkun, J.W. (1998). The Drosophila trithorax group proteins BRM, ASH1 and ASH2 are subunits of distinct protein complexes. Development 125, 3955-3966.

Parsons, D.W., Li, M., Zhang, X., Jones, S., Leary, R.J., Lin, J.C., Boca, S.M., Carter, H., Samayoa, J., Bettegowda, C., et al. (2011). The genetic landscape of the childhood cancer medulloblastoma. Science 331, 435-439.

Pearce, D.J., Taussig, D., Simpson, C., Allen, K., Rohatiner, A.Z., Lister, T.A., and Bonnet, D. (2005). Characterization of cells with a high aldehyde dehydrogenase activity from cord blood and acute myeloid leukemia samples. Stem Cells 23, 752-760.

Pena-Llopis, S., Vega-Rubin-de-Celis, S., Liao, A., Leng, N., Pavia-Jimenez, A., Wang, S., Yamasaki, T., Zhrebker, L., Sivanand, S., Spence, P., et al. (2012). BAP1 loss defines a new class of renal cell carcinoma. Nat Genet 44, 751-759.

Phelan, M., et al. (1999). Reconstitution of a Core Chromatin Remodeling Complex from SWI/SNF Subunits. Molecular Cell 3, 247-253.

Ponti, D., Costa, A., Zaffaroni, N., Pratesi, G., Petrangolini, G., Coradini, D., Pilotti, S., Pierotti, M.A., and Daidone, M.G. (2005). Isolation and in vitro propagation of tumorigenic breast cancer cells with stem/progenitor cell properties. Cancer Res 65, 5506-5511.

Qiu, X., Wang, Z., Li, Y., Miao, Y., Ren, Y., and Luan, Y. (2012). Characterization of sphere-forming cells with stem-like properties from the small cell lung cancer cell line H446. Cancer Lett 323, 161-170.

Raha, D., Wilson, T.R., Peng, J., Peterson, D., Yue, P., Evangelista, M., Wilson, C., Merchant, M., and Settleman, J. (2014). The cancer stem cell marker aldehyde dehydrogenase is required to maintain a drug-tolerant tumor cell subpopulation. Cancer Res 74, 3579-3590.

Ramos, P., Karnezis, A.N., Craig, D.W., Sekulic, A., Russell, M.L., Hendricks, W.P., Corneveaux, J.J., Barrett, M.T., Shumansky, K., Yang, Y., et al. (2014). Small cell carcinoma of the ovary, hypercalcemic

Page 136: Dissertation Cover pages v1.0 EDITS 122815

123

type, displays frequent inactivating germline and somatic mutations in SMARCA4. Nat Genet 46, 427-429.

Ramp, U., Jaquet, K., Reinecke, P., Schardt, C., Friebe, U., Nitsch, T., Marx, N., Gabbert, H.E., and Gerharz, C.D. (1997). Functional intactness of stimulatory and inhibitory autocrine loops in human renal carcinoma cell lines of the clear cell type. J Urol 157, 2345-2350.

Ravaud, A., Hawkins, R., Gardner, J.P., von der Maase, H., Zantl, N., Harper, P., Rolland, F., Audhuy, B., Machiels, J.P., Petavy, F., et al. (2008). Lapatinib versus hormone therapy in patients with advanced renal cell carcinoma: a randomized phase III clinical trial. J Clin Oncol 26, 2285-2291.

Reisman, D.N., Sciarrotta, J., Wang, W., Funkhouser, W.K., and Weissman, B.E. (2003). Loss of BRG1/BRM in human lung cancer cell lines and primary lung cancers: correlation with poor prognosis. Cancer Res 63, 560-566.

Rendina, R., Strangi, A., Avallone, B., and Giordano, E. (2010). Bap170, a subunit of the Drosophila PBAP chromatin remodeling complex, negatively regulates the EGFR signaling. Genetics 186, 167-181.

Reynolds, B.A., and Weiss, S. (1992). Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system. Science 255, 1707-1710.

Roberts, C.W., Leroux, M.M., Fleming, M.D., and Orkin, S.H. (2002). Highly penetrant, rapid tumorigenesis through conditional inversion of the tumor suppressor gene Snf5. Cancer Cell 2, 415-425.

Romero, O.A., Setien, F., John, S., Gimenez-Xavier, P., Gomez-Lopez, G., Pisano, D., Condom, E., Villanueva, A., Hager, G.L., and Sanchez-Cespedes, M. (2012). The tumour suppressor and chromatin-remodelling factor BRG1 antagonizes Myc activity and promotes cell differentiation in human cancer. EMBO Mol Med 4, 603-616.

Rosebeck, S., and Leaman, D.W. (2008). Mitochondrial localization and pro-apoptotic effects of the interferon-inducible protein ISG12a. Apoptosis 13, 562-572.

Sato, Y., Yoshizato, T., Shiraishi, Y., Maekawa, S., Okuno, Y., Kamura, T., Shimamura, T., Sato-Otsubo, A., Nagae, G., Suzuki, H., et al. (2013). Integrated molecular analysis of clear-cell renal cell carcinoma. Nat Genet 45, 860-867.

Schones, D.E., Cui, K., Cuddapah, S., Roh, T.Y., Barski, A., Wang, Z., Wei, G., and Zhao, K. (2008). Dynamic regulation of nucleosome positioning in the human genome. Cell 132, 887-898.

Schug, T.T., Berry, D.C., Shaw, N.S., Travis, S.N., and Noy, N. (2007). Opposing effects of retinoic acid on cell growth result from alternate activation of two different nuclear receptors. Cell 129, 723-733.

Shain, A.H., Giacomini, C.P., Matsukuma, K., Karikari, C.A., Bashyam, M.D., Hidalgo, M., Maitra, A., and Pollack, J.R. (2012). Convergent structural alterations define SWItch/Sucrose NonFermentable (SWI/SNF) chromatin remodeler as a central tumor suppressive complex in pancreatic cancer. Proc Natl Acad Sci U S A 109, E252-259.

Shain, A.H., and Pollack, J.R. (2013). The spectrum of SWI/SNF mutations, ubiquitous in human cancers. PLoS One 8, e55119.

Shaw, F.L., Harrison, H., Spence, K., Ablett, M.P., Simoes, B.M., Farnie, G., and Clarke, R.B. (2012). A detailed mammosphere assay protocol for the quantification of breast stem cell activity. J Mammary Gland Biol Neoplasia 17, 111-117.

Shim, E.Y., Hong, S.J., Oum, J.H., Yanez, Y., Zhang, Y., and Lee, S.E. (2007). RSC mobilizes nucleosomes to improve accessibility of repair machinery to the damaged chromatin. Mol Cell Biol 27, 1602-1613.

Page 137: Dissertation Cover pages v1.0 EDITS 122815

124

Sif, S., Saurin, A.J., Imbalzano, A.N., and Kingston, R.E. (2001). Purification and characterization of mSin3A-containing Brg1 and hBrm chromatin remodeling complexes. Genes Dev 15, 603-618.

Singh, S., Brocker, C., Koppaka, V., Chen, Y., Jackson, B.C., Matsumoto, A., Thompson, D.C., and Vasiliou, V. (2013). Aldehyde dehydrogenases in cellular responses to oxidative/electrophilic stress. Free Radic Biol Med 56, 89-101.

Society, A.C. (2015). Cancer Facts & Figures - 2015.

Stern, M., Jensen, R., and Herskowitz, I. (1984). Five SWI genes are required for expression of the HO gene in yeast. J Mol Biol 178, 853-868.

Su, Y., Qiu, Q., Zhang, X., Jiang, Z., Leng, Q., Liu, Z., Stass, S.A., and Jiang, F. (2010). Aldehyde dehydrogenase 1 A1-positive cell population is enriched in tumor-initiating cells and associated with progression of bladder cancer. Cancer Epidemiol Biomarkers Prev 19, 327-337.

Suomela, S., Cao, L., Bowcock, A., and Saarialho-Kere, U. (2004). Interferon alpha-inducible protein 27 (IFI27) is upregulated in psoriatic skin and certain epithelial cancers. J Invest Dermatol 122, 717-721.

Swift, S., Lorens, J., Achacoso, P., and Nolan, G.P. (2001). Rapid production of retroviruses for efficient gene delivery to mammalian cells using 293T cell-based systems. Curr Protoc Immunol Chapter 10, Unit 10 17C.

Tamkun, J.W., Deuring, R., Scott, M.P., Kissinger, M., Pattatucci, A.M., Kaufman, T.C., and Kennison, J.A. (1992). brahma: a regulator of Drosophila homeotic genes structurally related to the yeast transcriptional activator SNF2/SWI2. Cell 68, 561-572.

Trapnell, C., Roberts, A., Goff, L., Pertea, G., Kim, D., Kelley, D.R., Pimentel, H., Salzberg, S.L., Rinn, J.L., and Pachter, L. (2012). Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat Protoc 7, 562-578.

Ueda, K., Ogasawara, S., Akiba, J., Nakayama, M., Todoroki, K., Ueda, K., Sanada, S., Suekane, S., Noguchi, M., Matsuoka, K., et al. (2013). Aldehyde dehydrogenase 1 identifies cells with cancer stem cell-like properties in a human renal cell carcinoma cell line. PLoS One 8, e75463.

van den Berg, A., Hulsbeek, M.F., de Jong, D., Kok, K., Veldhuis, P.M., Roche, J., and Buys, C.H. (1996). Major role for a 3p21 region and lack of involvement of the t(3;8) breakpoint region in the development of renal cell carcinoma suggested by loss of heterozygosity analysis. Genes Chromosomes Cancer 15, 64-72.

van den Hoogen, C., van der Horst, G., Cheung, H., Buijs, J.T., Lippitt, J.M., Guzman-Ramirez, N., Hamdy, F.C., Eaton, C.L., Thalmann, G.N., Cecchini, M.G., et al. (2010). High aldehyde dehydrogenase activity identifies tumor-initiating and metastasis-initiating cells in human prostate cancer. Cancer Res 70, 5163-5173.

Varela, I., Tarpey, P., Raine, K., Huang, D., Ong, C.K., Stephens, P., Davies, H., Jones, D., Lin, M.L., Teague, J., et al. (2011). Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature 469, 539-542.

Versteege, I., Sevenet, N., Lange, J., Rousseau-Merck, M.F., Ambros, P., Handgretinger, R., Aurias, A., and Delattre, O. (1998). Truncating mutations of hSNF5/INI1 in aggressive paediatric cancer. Nature 394, 203-206.

Wang, S., Sun, H., Ma, J., Zang, C., Wang, C., Wang, J., Tang, Q., Meyer, C.A., Zhang, Y., and Liu, X.S. (2013). Target analysis by integration of transcriptome and ChIP-seq data with BETA. Nat Protoc 8, 2502-2515.

Page 138: Dissertation Cover pages v1.0 EDITS 122815

125

Wang, W., Cote, J., Xue, Y., Zhou, S., Khavari, P.A., Biggar, S.R., Muchardt, C., Kalpana, G.V., Goff, S.P., Yaniv, M., et al. (1996). Purification and biochemical heterogeneity of the mammalian SWI-SNF complex. EMBO J 15, 5370-5382.

Wang, X., Sansam, C.G., Thom, C.S., Metzger, D., Evans, J.A., Nguyen, P.T., and Roberts, C.W. (2009). Oncogenesis caused by loss of the SNF5 tumor suppressor is dependent on activity of BRG1, the ATPase of the SWI/SNF chromatin remodeling complex. Cancer Res 69, 8094-8101.

Wang, Z., Zhai, W., Richardson, J.A., Olson, E.N., Meneses, J.J., Firpo, M.T., Kang, C., Skarnes, W.C., and Tjian, R. (2004). Polybromo protein BAF180 functions in mammalian cardiac chamber maturation. Genes Dev 18, 3106-3116.

Wenzel, J., Tomiuk, S., Zahn, S., Kusters, D., Vahsen, A., Wiechert, A., Mikus, S., Birth, M., Scheler, M., von Bubnoff, D., et al. (2008). Transcriptional profiling identifies an interferon-associated host immune response in invasive squamous cell carcinoma of the skin. Int J Cancer 123, 2605-2615.

Wiegand, K.C., Shah, S.P., Al-Agha, O.M., Zhao, Y., Tse, K., Zeng, T., Senz, J., McConechy, M.K., Anglesio, M.S., Kalloger, S.E., et al. (2010). ARID1A mutations in endometriosis-associated ovarian carcinomas. N Engl J Med 363, 1532-1543.

Wilson, B.G., Helming, K.C., Wang, X., Kim, Y., Vazquez, F., Jagani, Z., Hahn, W.C., and Roberts, C.W. (2014). Residual complexes containing SMARCA2 (BRM) underlie the oncogenic drive of SMARCA4 (BRG1) mutation. Mol Cell Biol 34, 1136-1144.

Wilson, B.G., and Roberts, C.W. (2011). SWI/SNF nucleosome remodellers and cancer. Nat Rev Cancer 11, 481-492.

Wilson, B.G., Wang, X., Shen, X., McKenna, E.S., Lemieux, M.E., Cho, Y.J., Koellhoffer, E.C., Pomeroy, S.L., Orkin, S.H., and Roberts, C.W. (2010). Epigenetic antagonism between polycomb and SWI/SNF complexes during oncogenic transformation. Cancer Cell 18, 316-328.

Witkowski, L., Carrot-Zhang, J., Albrecht, S., Fahiminiya, S., Hamel, N., Tomiak, E., Grynspan, D., Saloustros, E., Nadaf, J., Rivera, B., et al. (2014). Germline and somatic SMARCA4 mutations characterize small cell carcinoma of the ovary, hypercalcemic type. Nat Genet 46, 438-443.

Wu, J.I., Lessard, J., and Crabtree, G.R. (2009). Understanding the words of chromatin regulation. Cell 136, 200-206.

Wurster, A.L., Precht, P., Becker, K.G., Wood, W.H., 3rd, Zhang, Y., Wang, Z., and Pazin, M.J. (2012). IL-10 transcription is negatively regulated by BAF180, a component of the SWI/SNF chromatin remodeling enzyme. BMC Immunol 13, 9.

Xi, Q., He, W., Zhang, X.H., Le, H.V., and Massague, J. (2008). Genome-wide impact of the BRG1 SWI/SNF chromatin remodeler on the transforming growth factor beta transcriptional program. J Biol Chem 283, 1146-1155.

Xia, W., Nagase, S., Montia, A.G., Kalachikov, S.M., Keniry, M., Su, T., Memeo, L., Hibshoosh, H., and Parsons, R. (2008). BAF180 is a critical regulator of p21 induction and a tumor suppressor mutated in breast cancer. Cancer Res 68, 1667-1674.

Xie, W., Su, K., Wang, D., Paterson, A.J., and Kudlow, J.E. (1997). MDA468 growth inhibition by EGF is associated with the induction of the cyclin-dependent kinase inhibitor p21WAF1. Anticancer Res 17, 2627-2633.

Xu, X., Chai, S., Wang, P., Zhang, C., Yang, Y., Yang, Y., and Wang, K. (2015). Aldehyde dehydrogenases and cancer stem cells. Cancer Lett 369, 50-57.

Page 139: Dissertation Cover pages v1.0 EDITS 122815

126

Xue, Y., Canman, J.C., Lee, C.S., Nie, Z., Yang, D., Moreno, G.T., Young, M.K., Salmon, E.D., and Wang, W. (2000). The human SWI/SNF-B chromatin-remodeling complex is related to yeast rsc and localizes at kinetochores of mitotic chromosomes. Proc Natl Acad Sci U S A 97, 13015-13020.

Yamasaki, K., Toriu, N., Hanakawa, Y., Shirakata, Y., Sayama, K., Takayanagi, A., Ohtsubo, M., Gamou, S., Shimizu, N., Fujii, M., et al. (2003). Keratinocyte growth inhibition by high-dose epidermal growth factor is mediated by transforming growth factor beta autoinduction: a negative feedback mechanism for keratinocyte growth. J Invest Dermatol 120, 1030-1037.

Yan, Z., Cui, K., Murray, D.M., Ling, C., Xue, Y., Gerstein, A., Parsons, R., Zhao, K., and Wang, W. (2005). PBAF chromatin-remodeling complex requires a novel specificity subunit, BAF200, to regulate expression of selective interferon-responsive genes. Genes Dev 19, 1662-1667.

Yen, W.C., Fischer, M.M., Hynes, M., Wu, J., Kim, E., Beviglia, L., Yeung, V.P., Song, X., Kapoun, A.M., Lewicki, J., et al. (2012). Anti-DLL4 has broad spectrum activity in pancreatic cancer dependent on targeting DLL4-Notch signaling in both tumor and vasculature cells. Clin Cancer Res 18, 5374-5386.

Young, A.P., Schlisio, S., Minamishima, Y.A., Zhang, Q., Li, L., Grisanzio, C., Signoretti, S., and Kaelin, W.G., Jr. (2008). VHL loss actuates a HIF-independent senescence programme mediated by Rb and p400. Nat Cell Biol 10, 361-369.

Zang, C., Schones, D.E., Zeng, C., Cui, K., Zhao, K., and Peng, W. (2009). A clustering approach for identification of enriched domains from histone modification ChIP-Seq data. Bioinformatics 25, 1952-1958.

Zang, Z.J., Cutcutache, I., Poon, S.L., Zhang, S.L., McPherson, J.R., Tao, J., Rajasegaran, V., Heng, H.L., Deng, N., Gan, A., et al. (2012). Exome sequencing of gastric adenocarcinoma identifies recurrent somatic mutations in cell adhesion and chromatin remodeling genes. Nat Genet 44, 570-574.

Zhang, L., Jiao, M., Li, L., Wu, D., Wu, K., Li, X., Zhu, G., Dang, Q., Wang, X., Hsieh, J.T., et al. (2012). Tumorspheres derived from prostate cancer cells possess chemoresistant and cancer stem cell properties. J Cancer Res Clin Oncol 138, 675-686.

Zhang, Y., Liu, T., Meyer, C.A., Eeckhoute, J., Johnson, D.S., Bernstein, B.E., Nusbaum, C., Myers, R.M., Brown, M., Li, W., et al. (2008). Model-based analysis of ChIP-Seq (MACS). Genome Biol 9, R137.

Zhao, D., Mo, Y., Li, M.T., Zou, S.W., Cheng, Z.L., Sun, Y.P., Xiong, Y., Guan, K.L., and Lei, Q.Y. (2014). NOTCH-induced aldehyde dehydrogenase 1A1 deacetylation promotes breast cancer stem cells. J Clin Invest 124, 5453-5465.