dna damage response suppresses epstein-barr virus-driven

131
DNA Damage Response Suppresses Epstein-Barr Virus-Driven Proliferation of Primary Human B Cells by Pavel Nikitin Department of Molecular Genetics and Microbiology Duke University Date: Approved: ___________________________ Micah Luftig, Supervisor ___________________________ Sandeep Dave ___________________________ Thomas Petes ___________________________ David Pickup ___________________________ Nancy Raab-Traub Dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Department of Molecular Genetics and Microbiology in the Graduate School of Duke University 2012

Upload: others

Post on 07-Nov-2021

1 views

Category:

Documents


0 download

TRANSCRIPT

i

v

i

v

DNA Damage Response Suppresses Epstein-Barr Virus-Driven Proliferation

of Primary Human B Cells

by

Pavel Nikitin

Department of Molecular Genetics and Microbiology

Duke University

Date:

Approved:

___________________________

Micah Luftig, Supervisor

___________________________

Sandeep Dave

___________________________

Thomas Petes

___________________________

David Pickup

___________________________

Nancy Raab-Traub

Dissertation submitted in partial fulfillment of the requirements for the degree of Doctor

of Philosophy in the Department of Molecular Genetics and Microbiology in the

Graduate School of Duke University

2012

ABSTRACT

DNA Damage Response Suppresses Epstein-Barr Virus-Driven Proliferation

of Primary Human B Cells

by

Pavel Nikitin

Department of Molecular Genetics and Microbiology

Duke University

Date:

Approved:

___________________________

Micah Luftig, Supervisor

___________________________

Sandeep Dave

___________________________

Thomas Petes

___________________________

David Pickup

___________________________

Nancy Raab-Traub

An abstract of a dissertation submitted in partial fulfillment of the requirements for the

degree of Doctor of Philosophy in the Department of Molecular Genetics and

Microbiology in the Graduate School of Duke University

2012

Copyright by

Pavel A. Nikitin

Павел А. Никитин

2012

iv

Abstract

The interaction of human tumor viruses with host growth suppressive pathways

is a fine balance between controlled latent infection and virus-induced oncogenesis. This

dissertation elucidates how Epstein-Barr virus interacts with the host growth

suppressive DNA damage response signaling pathways (DDR) in order to transform

infected human B lymphocytes.

Here I report that the activation of the ATM/Chk2 branch of the DDR in hyper-

proliferating infected B cells results in G1/S cell cycle arrest and limits viral-mediated

transformation. Similar growth arrest was found in mitogen-driven proliferating of B

cells that sets the DDR as a default growth suppressive mechanism in human B cells.

Hence, the viral protein EBNA3C functions to attenuate the host DDR and to promote

immortalization of a small portion of infected B cells. Additionally, the pharmacological

inhibition of the DDR in vitro increases viral immortalization of memory B cells that

facilitates the isolation of broadly neutralizing antibodies to various infectious agents.

Overall, this work defines early EBV-infected hyper-proliferating B cells as a new stage

in viral infection that determines subsequent viral-mediated tumorigenesis.

v

Dedication

Моей маме To my mom

vi

Contents

Abstract ......................................................................................................................................... iv

List of Tables .................................................................................................................................. x

List of Figures ............................................................................................................................... xi

Acknowledgements .................................................................................................................. xiii

1. Introduction ............................................................................................................................... 1

1.1. Human tumor viruses associate with multiple malignancies ................................... 1

1.2. Epstein-Barr virus infection cycle determines viral latency programs .................... 1

1.2.1. EBV infection in vitro results in poor transformation efficiency .......................... 7

1.3. The tumor suppressive DNA damage responsive signaling pathway .................... 8

1.3.1. Activated oncogenes induce replicative stress ....................................................... 8

1.3.2. Ataxia telangiectasia mutated (ATM) regulates a response to DNA double-

stranded breaks ..................................................................................................................... 9

1.3.3. Innate tumor suppression limits oncogene-induced replicative stress ............ 10

1.3.4. Chronic DNA damage foci form DNA segments with chromatin alterations

reinforcing senescence (DNA-SCARS) ............................................................................ 11

1.4. The DNA damage response in viral-induced cellular transformation .................. 12

1.4.1. Viral gene expression provokes the tumor suppressive DDR ........................... 13

1.4.1.1. Viral oncoproteins drive cellular hyper-proliferation and thus, induce a

tumor suppressive DDR ............................................................................................... 14

1.4.1.2. Viral proteins directly induce a beneficial DDR, including the DNA repair

and activation of checkpoints ....................................................................................... 18

1.4.1.3. DDR activation through viral oncoprotein-mediated mitotic effects ........ 19

vii

1.4.1.4. Tumor viruses activate the DDR through induction of reactive oxygen

species .............................................................................................................................. 20

1.4.2. Tumor viruses modulate the activated DDR to promote tumorigenesis ......... 21

1.4.2.1. Tumor virus suppression of downstream DDR signaling components ... 21

1.4.2.2. Viral oncoproteins directly target DDR checkpoint kinases ....................... 23

1.4.2.3. Viral oncoproteins perturb mitotic checkpoint signaling ........................... 24

2. Results ....................................................................................................................................... 27

2.1. An ATM/Chk2-Mediated DNA Damage-Responsive Signaling Pathway

Suppresses Epstein - Barr virus Driven Proliferation of Primary B cells ..................... 27

2.1.1. Contributions ............................................................................................................ 27

2.1.2. Introduction ............................................................................................................... 28

2.1.3. Results ........................................................................................................................ 29

2.1.3.1. Epstein-Barr virus infection of primary B cells activates a cellular DNA

damage response ............................................................................................................ 29

2.1.3.2. The EBV-induced DNA damage response in primary B cell infection is

not associated with viral episomes or lytic replication ............................................ 33

2.1.3.3. The EBV-induced DNA damage response is associated with a transient

period of hyper-proliferation ....................................................................................... 34

2.1.3.4. Proliferation and DNA damage responsive genes are highly induced

early after EBV infection, then attenuated during LCL outgrowth ........................ 39

2.1.3.5. The EBV-induced hyper-proliferation associated DNA damage response

is growth suppressive.................................................................................................... 43

2.1.3.6. ATM and Chk2 kinases suppress EBV-mediated transformation and

initial B cell proliferation .............................................................................................. 45

2.1.3.7. ATM and Chk2 suppress B cell growth 4-8 days after EBV infection ....... 47

viii

2.1.3.8. EBV latent gene expression changes and consequences in early infected

cell divisions ................................................................................................................... 48

2.1.3.9. EBNA3C is required to attenuate the EBV-induced DNA damage

response ........................................................................................................................... 51

2.1.4. Discussion .................................................................................................................. 57

2.2. Mitogen-Induced B Cell Proliferation Activates Chk2-dependent G1/S Cell Cycle

Arrest ...................................................................................................................................... 58

2.2.1. Contributions ............................................................................................................ 58

2.2.2. Introduction ............................................................................................................... 58

2.2.3. Results ........................................................................................................................ 60

2.2.3.1. Mitogen stimulation of primary B cells results in a period of robust

proliferation .................................................................................................................... 60

2.2.3.2. Mitogen stimulation activates the ATM signaling pathway in hyper-

proliferating cells ........................................................................................................... 61

2.2.3.3. EBV and mitogen-induced B-cell proliferation is suppressed by Chk2 .... 63

2.2.3.4. B-cell mitogens induce caspase 3/7-dependent apoptosis independent of

Chk2 ................................................................................................................................. 65

2.2.3.6. Mitogen-induced hyper-proliferation of human B cells induced a Chk2-

dependent G1/S cell cycle arrest .................................................................................. 66

2.2.3.7. Activated Chk2 induces expression of the CDK inhibitor p21 ................... 69

2.2.4. Discussion .................................................................................................................. 69

2.3. Enhanced Method of Epstein-Barr Virus Mediated Transformation of B Cells

Used for Generation of Human Monoclonal Antibodies ............................................... 73

2.3.1. Contributions ............................................................................................................ 73

2.3.2. Introduction ............................................................................................................... 73

ix

2.3.3. Results ........................................................................................................................ 75

2.3.3.1. Pharmacological inhibition of ATM or Chk2 increases EBV-mediated

transformation of low numbers of B cells ................................................................... 75

2.3.3.2. Inhibition of Chk2 coupled with TLR9 stimulation additively enhances

EBV-mediated transformation of memory B cells from normal donors ................ 77

2.3.3.3. Combined treatment with Chk2i and TLR9 ligand increases EBV-

mediated transformation of memory B cells from a chronic HIV-infected patient

.......................................................................................................................................... 78

2.3.4. Discussion .................................................................................................................. 80

3. Conclusions and future directions ........................................................................................ 81

3.1. The source of DNA damage ......................................................................................... 82

3.2. DDR signaling in transformed cells ............................................................................ 85

3.3. Implication in vivo ........................................................................................................ 86

3.4. Distinct expression of viral latency proteins in EBV infected primary B cells ...... 88

3.4.1. How is EBNA3C activity regulated in infected cells? ......................................... 88

3.4.2. What is the role of LMP1 in restricting the DDR and why is its expression is

delayed? ............................................................................................................................... 89

3.4.3. How EBNA-LP length and expression control EBV-driven oncogenesis? ...... 89

3.5. A proposed model of EBV infection of primary B cells ........................................... 90

References .................................................................................................................................... 92

Biography ................................................................................................................................... 118

x

List of Tables

Table 1: Human oncogenic viruses and their interactions with the host DNA damage

response (adapted from (Nikitin and Luftig, 2012)). ............................................................. 13

xi

List of Figures

Figure 1: The model of EBV initial and persistent infection (from (Thorley-Lawson and

Allday, 2008)). ................................................................................................................................ 4

Figure 2: Interplay between viral oncoproteins and the host DNA damage response

(from (Nikitin and Luftig, 2012)). ............................................................................................. 16

Figure 3: Nuclear genome deposition and the analysis of viral and host gene expression

following primary B cell infection with EBV B95-8, UV-inactivated B95-8, P3-HR1 (A-C).

EBV genomes in infected primary B cells are independent of -H2AX foci (D-G). ........... 30

Figure 4: EBV induced a DNA damage response in primary B cells. .................................. 32

Figure 5: EBV induced a period of hyper-proliferative early after infection that was

associated with activation of the DNA damage response..................................................... 35

Figure 6: CFSE-based kinetic analysis of EBV-induced proliferation. ................................. 37

Figure 7: Transcriptional changes correlate with an EBV-induced early period of hyper-

proliferation and DNA damage response followed by attenuation upon LCL outgrowth.

....................................................................................................................................................... 40

Figure 8: Schematic diagram of GSEA analysis performed on B cells, Proliferating cells,

and LCL microarray data with ATM/p53 target genes. ........................................................ 42

Figure 9: Growth suppression and DNA damage enrichment in early cell divisions. ..... 44

Figure 10: Inhibition of ATM and Chk2 kinases increased EBV transformation efficiency

and proliferation of B cells during a critical period 4-8 days post infection. ...................... 46

Figure 11: EBV latency and consequential host gene expression changes from initial B

cell proliferation through LCL outgrowth. ............................................................................. 50

Figure 12: Expression of viral latency genes during early cell divisions through LCL

outgrowth. .................................................................................................................................... 52

Figure 13: EBNA3C attenuates the EBV-induced DNA damage response. ....................... 54

Figure 14: Characterization of EBNA-3A KO and EBNA-3C KO viruses. ......................... 56

xii

Figure 15: Multiple B cell mitogens induce hyper-proliferation. ......................................... 60

Figure 16: B cell mitogens induce the ATM signaling pathways in hyper-proliferating

cells. ............................................................................................................................................... 62

Figure 17: Chk2 inhibition increase proliferation of human B cells. .................................... 64

Figure 18: B-cell mitogens, but not EBV induces caspases 3/7-dependent apoptosis. ...... 66

Figure 19: Mitogen stimulation or EBV infection of human B cells activates Chk2-

dependent cell cycle arrest. ........................................................................................................ 68

Figure 20: Pharmacological inhibition of DDR kinases increases EBV transformation of

low numbers of total B cells derived from a normal donor. ................................................. 76

Figure 21: Chk2 inhibitor and TLR ligand CpG additively increase EBV-mediated

transformation of human memory B cells. .............................................................................. 77

Figure 22: Enhanced EBV transformation of memory B cells derived from a chronic HIV

patient. .......................................................................................................................................... 79

xiii

Acknowledgements

First of all, I would like to thank my PhD advisor, Dr. Micah Luftig, for his

incredible support during the time spent in graduate school. His invaluable guidance

shaped me as a scientist. This thesis work would not have succeeded without the help of

my committee members, Drs. Raab-Traub, Pickup, Petes and Dave whose critical

reviews facilitated my professional growth. Further, I would like to specially thank all

the present and past members of Luftig lab.

I would like to acknowledge our collaborators from Imperial College London,

Rob White and Martin Allday for their expertise and critical revision of our work and for

EBV viral mutants they kindly provided. I would like to thank Duke’s Dave lab,

Crawford lab, Hauser lab and Human Vaccine Institute members for our productive

collaborations. In addition, I would like to thank Cullen lab members for their advices

and for reagents they shared. I would like to acknowledge Lynn Martinek, Nancy

Martin and Mike Cook from DCI Flow Cytometry core facility for their enormous

support. Finally, I would like to thank my family, including my wife Mayya Shveygert

and my little daughter Masha, for their constant support.

1

1. Introduction

1.1. Human tumor viruses associate with multiple malignancies

Approximately 20% of all cases of human cancer have an infectious etiology,

with ~80% of those being viral (Bouvard et al., 2009) (Table 1). To date there are five

bone-fide human DNA tumor viruses and at least one RNA tumor virus that cause

various malignancies (discussed in details below). A unique aspect of tumor virus

infection is the interaction with the host immune system and cell-intrinsic tumor

suppressive mechanisms. Recent studies identified the tumor suppressive DNA damage

responsive signaling pathway (DDR) as one of the major barriers to viral-driven

tumorigenesis, reviewed below. In this dissertation I focus on Epstein-Barr virus (EBV)-

driven tumorigenesis and report ATM/Chk2-mediated DNA damage response restricts

viral immortalization of primary human B lymphocytes in-vitro.

1.2. Epstein-Barr virus infection cycle determines viral latency programs

EBV is a large double stranded DNA γ-herpesvirus that establishes a latent

infection in more than 95% of the adult population worldwide (Rickinson and Kieff,

2006). EBV infection is associated with various tumors of lymphoid and epithelial origin

including Burkitt’s lymphomas and gastric carcinoma, Hodgkin’s disease and

nasopharyngeal carcinoma, and AIDS-associated lymphomas and post-transplant

lymphomas (Kieff and Rickinson, 2006; Raab-Traub, 2002).

2

Histologically and molecularly diverse EBV-associated malignancies arise from

different stages of viral infectious cycle and reflect dynamic viral latent gene expression.

The initial asymptomatic EBV infection occurs in the oral-pharyngeal epithelium during

the childhood period (Figure 1 and (Thorley-Lawson and Allday, 2008)). In epithelial

cells, EBV undergoes lytic replication and amplification resulting in secretion of new

virions that infect B cells and establish a latent infection.

Viral expression in primary B cells is well studied in a convenient in vitro system

(Alfieri et al., 1991). After infection of B cells, the EBV genome enters the nucleus and

circularizes into an episome (Hurley and Thorley-Lawson, 1988) which allows viral

expression from the W-promoter ((Arrand and Rymo, 1982)) (Woisetschlaeger et al.,

1990). The initial Wp-dependent transcript includes two viral proteins, EBV nuclear

antigen – leader protein (EBNA-LP) and EBNA2 (Alfieri et al., 1991). EBNA2 binds to

host transcription factors, such as RBP-Jk, and upregulates cellular expression of growth

promoting genes (Henkel et al., 1994; Johannsen et al., 1996; Robertson et al., 1996; Wang

et al., 1991). Next, EBNA2 initiates viral transcription of the remaining EBV nuclear

antigens, including EBNA1, that maintains viral episome, EBN3A, 3B and 3C (Zimber-

Strobl et al., 1993) from the C-promoter (Cp), and latent membrane proteins 1 (LMP1),

LMP2A and LMP2B (Kieff and Rickinson, 2006; Wang et al., 1990). In addition to viral

proteins, latently infected cells display a distinct set of viral non-coding RNAs, including

abundant EBV-encoded small RNAs (EBERS) (Arrand and Rymo, 1982; Pathmanathan

3

et al., 1995; Swaminathan et al., 1991), and viral microRNAs encoded within BART and

BHRF regions (Cai et al., 2006; Forte and Luftig, 2011; Grundhoff et al., 2006; Pfeffer et

al., 2004; Skalsky et al., 2012). Intriguingly, expression of LMP-1, a viral protein required

for immortalization of B cells (Kaye et al., 1993; Kulwichit et al., 1998), is regulated by a

separate promoter (Chang et al., 1997; Fennewald et al., 1984; Sadler and Raab-Traub,

1995) and temporally separated in vitro (Price et al., 2012). Expression of all nine latent

proteins and non-coding RNAs leads to immortalization of infected B cells into

lymphoblasts and is termed latency III program.

4

Figure 1: The model of EBV initial and persistent infection (from (Thorley-

Lawson and Allday, 2008)).

5

In vivo, there are at least three possible destinies for the infected proliferating B

cell to progress. The first and the most common pathway includes the elimination of the

infected cell by cytotoxic T and NK-cells. Particularly, EBNA-3 viral proteins and, to a

lesser extent, EBNA-2 induce the potent cytotoxic cell-mediated immune response

(Khanna et al., 1992; Murray et al., 1992). Interestingly, EBV-infected cells expressing the

complete set of nine viral latent proteins are predominantly found in

immunosuppressed individuals, such as transplant or AIDS-infected patients (Raab-

Traub, 2007).

However, EBNA3s binding to RBP-Jk modulates the expression of EBNA2 and

Cp-driven transcripts (Henkel et al., 1994; Robertson et al., 1996) and restricts viral

expression, hence escaping from the immune control and providing the alternative way

to progress. Consistently, in immunocompetent individuals EBV-associated tumors of

epithelial and lymphoid origins express only EBNA-1 and viral non-coding RNAs or

additional latent membrane proteins LMP1 and LMP2. The former expression program

is termed latency I and found in Burkitt’s lymphoma and gastric carcinoma (Rowe et al.,

1987) while the latter is termed latency II and found in germinal center-derived

Hodgkin’s lymphoma and epithelial nasopharyngeal carcinoma (Brooks et al., 1992;

Rowe et al., 1992). While latency III tumors arise in tumor suppression, latency II and I

tumors are most likely promoted by environmental and genetic factors (Raab-Traub,

2012). Further, as latency II tumors were revealed in germinal-center derived B cells, the

6

initial Cp expression in activated lymphoblasts is likely silenced in infected cells

migrated to germinal center. In germinal center infected B cells mimic the normal B-cell

maturation process and differentiate into memory-like B cells (Babcock et al., 1998).

EBNA1 expressing memory-like B cells that persist in peripheral blood are found in

normal individuals and therefore thought to be a viral reservoir (Thorley-Lawson and

Allday, 2008).

Finally, the third way for an infected naïve B-cell to turn into a memory-like B

cell was recently proposed by the Rickinson and Bell group. According to this report,

EBV-driven expression of activation-induced cytidine deaminase (AID) results in

mutagenesis within Ig locus and thus drives the infected cell towards non-switched

memory-like phenotype. Moreover, infected cells with edited Ig loci have an advantage

during in vitro outgrowth (Heath et al., 2012). Therefore, if the proposed mechanism

takes place in vivo, EBV-infected naïve lymphocytes may bypass the germinal center on

their path to a memory-like phenotype. The resulted infected memory B cell circulates in

the peripheral blood and upon stress undergoes differentiation into a plasma cell

driving EBV lytic reactivation (Thorley-Lawson and Allday, 2008).

Overall, a distinct pathway of the host B cell to the final differentiated state

determines dynamic changes in viral gene expression that, in turn, regulates the cellular

growth.

7

1.2.1. EBV infection in vitro results in poor transformation efficiency

As mentioned above, our understanding of EBV latent gene expression changes

predominantly comes from viral infection of primary human B cells in vitro (reviewed in

(Kieff and Rickinson, 2006)). While having limitations (Thorley-Lawson and Allday,

2008), in vitro system allows investigating the EBV latency III program in primary cells at

physiological concentrations of viral proteins, non-coding viral RNAs and miRNAs. In

this system peripheral blood mononuclear cells (PBMC) are latently infected with EBV

in the presence of T-cell suppressants, such as cyclosporine A (Borel et al., 1977).

Infected B cells are induced to proliferate and undergo a growth transformation into

lymphoblastoid cells (LCL). However, only a small percentage of infected cells become

indefinitely proliferating lymphoblasts (Henderson et al., 1977; Sugden and Mark, 1977).

Therefore, there may be intrinsic tumor suppression at play that prevents cellular

proliferation and/or suppresses the immortalization of proliferating B cells.

The study of EBV-induced innate tumor suppressor pathways has been limited.

In immortalized lymphoblasts EBNA-3A and 3C epigenetically silence p16 and prevent

activation of Rb that promotes G1/S transition (Maruo et al., 2011; Skalska et al., 2010).

However, it remains unclear what the role of p16 is in primary infection of lymphocytes.

Aside from p16, EBV infection of primary B cells induces the p53 protein concomitant

with EBNA-LP expression early after infection (Szekely et al., 1995). However, it remains

unclear whether this innate response to EBV-induced proliferation has any long-term

8

functional consequence or what pathways activate p53. Therefore, investigation of cell-

intrinsic mechanisms to suppress tumorigenesis will inform about barriers the virus has

to overcome and allow us to better understand interactions between the virus and the

host.

1.3. The tumor suppressive DNA damage responsive signaling pathway

In the last decade the DNA damage responsive signaling pathway (DDR) was

recognized as a barrier to tumorigenesis in oncogene-expressing cells and in

precancerous lesions. The Chapter 1.3 briefly overviews activation signals, components

and consequences of tumor suppressive DDR.

1.3.1. Activated oncogenes induce replicative stress

Replicative stress is a condition describing the presence of stalled replicative

forks, whether due to a replication block or an impetuous increase in a number of new

origins, resulting in inhibition of DNA synthesis (reviewed by (Halazonetis et al., 2008;

Osborn et al., 2002)). Such collapse of replicative forks often occurs at common places in

the genome, termed fragile sites, and requires activation of recombination and formation

of DNA double-stranded breaks (DSB) to complete the replication.

Activated oncogenes in mammalian cells increase the number of origins of

replication and presumably collapse of replication forks, resulting in the formation of

aberrant DNA structures, such as stretches of single stranded DNA and formation of the

DNA DSB (Bartkova et al., 2006; Di Micco et al., 2006). Upon formation of DSBs, a

9

cascade of events is initiated to activate checkpoints and initiate repair or apoptosis

(Khanna and Jackson, 2001). Therefore, oncogene-induced DSBs initiate a tumor

suppressive cellular response (Bartkova et al., 2005; Gorgoulis et al., 2005).

1.3.2. Ataxia telangiectasia mutated (ATM) regulates a response to DNA double-stranded breaks

ATM serves as the key regulator of the cellular response to DNA double-

stranded breaks and upregulates cellular checkpoints to promote DNA repair or initiate

programmed cell death. The function of ATM and its yeast homolog Tel1 in maintaining

the chromosome stability was established in two initial works (Greenwell et al., 1995;

Savitsky et al., 1995). While mutations in Tel1 led to telomere shortening (Greenwell et

al., 1995) and thus to chromosomal instability, human cells derived from patients with

ataxia telangiectasia (AT), a recessive autosomal disorder characterized by

predisposition to cancer and checkpoints abnormalities, carried a mutant gene named

AT-mutated or ATM (Savitsky et al., 1995). ATM was found to be a serine/threonine

protein kinase with hydrophobic-X-hydrophobic-[S/T]-Q consensus motif (Kim et al.,

1999), and is a member of the phosphoinositide 3-kinase-related protein kinase (PIKK)

family. Later works identified ATM and its downstream kinase Chk2 as regulating

replication checkpoints in response to ionizing irradiation-induced DNA DSBs (Falck et

al., 2001; Matsuoka et al., 1998). Currently, ATM is believed to induce G1/S, intra-S-

phase and G2/M cell cycle checkpoints through the following mechanisms: ATM

phosphorylation of p53 upregulates p21 expression resulting in inhibition of Cyclin-

10

E/CDK2 complex and blocking G1/ S transition. Additionally, an intra-S-phase

checkpoint is regulated by ATM through activation of NBS1, BRCA1, SMC1, FANCD2

and Chk2 kinase. Activated Chk2 kinase induces phosphorylation and a subsequent

degradation of CDC25A. As CDC25A normally activates Cyclin-E/CDK2 complex, its

degradation blocks G1/S transition. Furthermore, CDC25A degradation results in

elevated level of CDK2 phosphorylation and its destabilization slows down the

progression through S-phase. Finally, ATM and Chk2 play a role in the G2/M checkpoint

to promote repair through Rad17 and Artemis (reviewed in (Derheimer and Kastan,

2010; Harper and Elledge, 2007; Shiloh, 2003)).

1.3.3. Innate tumor suppression limits oncogene-induced replicative stress

Innate tumor suppression in response to oncogenic stress includes the well-

characterized alternative reading frame to 16 (ARF) tumor suppressor, also known as

p14, CDKN2A)-mediated activation and stabilization of p53 (Christophorou et al., 2006;

Efeyan et al., 2006; Zindy et al., 2003) and the cellular DNA damage response (DDR) that

is activated following oncogene-induced replicative stress (Halazonetis et al., 2008). As

first recognized by Halazonetis, Bartek and colleagues, tumor cells often display an

activated DDR as evidenced by foci of DDR signaling proteins such as 53BP1 and

activated ATM (DiTullio et al., 2002). Subsequent works demonstrated that acute over-

expression of oncogenes caused replicative stress sensed by the ATM and Rad3-related

kinase (ATR) signaling pathway as well as double-stranded breaks recognized by the

11

ATM pathway (Bartkova et al., 2005; Gorgoulis et al., 2005). Not long after the initial

characterization of these pathways, the functional significance of the DDR activation

was revealed by genetic studies indicating that ATM and Chk2 were critical tumor

suppressors downstream of oncogenes including H-RasV12, Mos, Cdc6, and cyclin E

(Bartkova et al., 2006; Di Micco et al., 2006; Hong et al., 2006; Stracker et al., 2008).

Mechanistically, these data linked the well-studied DDR response to DNA double

stranded breaks and known tumor suppressor functions of its components, including

activation of checkpoints and p53-mediated apoptosis and senescence, to an oncogene-

induced replicative stress.

1.3.4. Chronic DNA damage foci form DNA segments with chromatin alterations reinforcing senescence (DNA-SCARS)

As discussed above, activated oncogenes in cell lines or in precancerous lesions

were found to activate ATM/Chk2-dependent signaling. However, the question remains

whether the activation signal comes from continuous DNA damage or may persist in

proliferating cells after the damage is repaired. Recent works elucidate the difference

between acute DDR signaling and chronic chromatin changes induced by initial

aberrations during DNA synthesis (Di Micco et al., 2011; Rodier et al., 2009). The

Campisi group found chronic DDR signaling foci that lacked the DNA repair proteins

replication protein A (RPA) and RAD51 as well as active DNA replication, but instead

contained activated ATM-downstream components, such as localized Chk2 kinase and

phospho p53-Ser15. Such foci were termed “DNA segments with chromatin alterations

12

reinforcing senescence” or DNA-SCARS. Notably, normally senescent DNA-SCARS

positive cells continued to proliferate after experimental inactivation of p16 (Rodier et

al., 2011) or knockdown of ATM (Rodier et al., 2009). Consistently, d’Adda di Fagagna

group found senescence-associated heterochromatin foci (SAHF) associated with

senescence that required activated p16 (Di Micco et al., 2011).

DNA-SCARS and SAHF presumably mark separate oncogene-induced changes

of the chromatin, as the former co-localizes with activated ATM-signaling molecules and

γH2AX, while the latter excludes γH2AX and depends on ATR. However, both

chromatin alterations occur upon oncogene-induced senescence and persist in

proliferating cells with abrogated checkpoints.

1.4. The DNA damage response in viral-induced cellular transformation

In the last decade, multiple studies have found the tumor suppressive role of the

DDR in response to viral oncoproteins (Table 1). A unique aspect of these interactions is

the interplay between the virus and the host with respect to virus replication versus

aberrant induction of growth control genes and inhibition of apoptosis. Chapter 1.4 will

focus on complex interactions between tumor viruses and the host DNA damage

response and outcomes that promote or prevent virus-induced tumorigenesis.

13

Table 1: Human oncogenic viruses and their interactions with the host DNA

damage response (adapted from (Nikitin and Luftig, 2012)).

Oncogenic

Virus

Tumors associated with

virus infection

Oncoproteins involved

in DDR

Reference(s)

EBV Burkitt’s lymphoma,

Post-transplant lymphoma,

Non-hodgkin’s/Diffuse

large B cell lymphomas,

Nasopharyngeal carcinoma,

Gastric carcinoma

EBNA1 ROS DDR

EBNA3C Chk2, p53

EBNA3C G2/M

checkpoint

LMP1 ATM

(Gruhne et al., 2009a)

(Choudhuri et al.,

2007; Yi et al., 2009)

(Gruhne et al., 2009b;

Parker et al., 2000)

(Gruhne et al., 2009b)

KSHV Kaposi’s sarcoma, primary

effusion lymphoma

v-cyclin ATM

v-cyclinOIS

LANA myc DDR

LANA:p53

v-FLIP OIS

(Koopal et al., 2007)

(Liu et al., 2007)

(Chen et al., 2010;

Friborg et al., 1999;

Leidal et al., 2012)

HPV Cervical cancer, ovarian

cancer

E6,E7 repl stress

DDR E7:pATM

E6 p53

(Bester et al., 2011)

(Moody and Laimins,

2009)

(Scheffner et al.,

1990)

HBV Hepatocellular carcinoma HBV ATR

HBx Ras DDR

HBx p53

(Wang et al., 2008;

Zhao et al., 2008a)

(Klein and Schneider,

1997)

(Wang et al., 1994)

HTLV I Adult T cell leukemia (ATL) Tax DNA-PK

Tax Chk1/Chk2

Tax p53

(Durkin et al., 2008)

(Park et al., 2004;

Park et al., 2006)

(Ariumi et al., 2000;

Pise-Masison et al.,

2000)

1.4.1. Viral gene expression provokes the tumor suppressive DDR

The replication of tumor viruses is intrinsically linked to their ability to drive cell

proliferation. Most of these viruses infect quiescent cells driving re-entry into the cell

14

cycle to promote an environment conducive for viral nucleic acid replication. The

consequences of such aberrant induction of cell proliferation include increased

replicative stress, similar to that of cellular oncogene activation, leading to induction of

the DNA damage response. However, direct viral oncoprotein activation of the DDR

also occurs through multiple mechanisms discussed below.

1.4.1.1. Viral oncoproteins drive cellular hyper-proliferation and thus, induce a tumor

suppressive DDR

Small DNA tumor viruses antagonize the transcriptionally repressive Rb family

of proteins to promote E2F-driven cellular proliferation. Uncontrolled E2F activity has

been shown to activate an ATM-dependent growth-suppressive DDR (Powers et al.,

2004; Rogoff et al., 2004). HPV E7 and SV40 large T antigen are classic examples of viral

oncoproteins targeting Rb by direct disruption of the interaction with E2F thereby

increasing S-phase- promoting E2F family members to drive cellular DNA replication

(Cheng et al., 1995; DeCaprio et al., 1988; Dyson et al., 1989; Munger et al., 1989; Zalvide

and DeCaprio, 1995). In recent work, E6 and E7 over-expression has been shown to

induce replicative stress in primary keratinocytes suggesting that potent loss of growth

control through E7- mediated Rb antagonism drives uncontrolled origin firing leading to

damaged DNA that may contribute to cervical cancer pathogenesis (Bester et al., 2011).

Similarly, SV40 large T antigen is sufficient to activate an ATM-induced DDR (Figure 1)

(Boichuk et al., 2010). However, as described later, SV40 large T antigen activates the

DDR through multiple mechanisms including those independent of Rb interaction

15

(Boichuk et al., 2010). While the polyomavirus SV40 does not cause human cancer,

Merkel cell carcinoma polyomavirus (MCV) expresses a truncated large T antigen in

these tumors lacking the capacity to replicate viral DNA (Shuda et al., 2008). These

mutant large T antigens still retain the ability to perturb cell growth through Rb

antagonism and likely activate the DDR providing selective pressure for mutations in

DDR genes and downstream signaling leading to MCC.

16

Figure 2: Interplay between viral oncoproteins and the host DNA damage

response (from (Nikitin and Luftig, 2012)).

Viral oncoproteins activate cellular oncogenes (green arrows top level) in order

to enter or re-enter the cell cycle thereby inducing replicative stress and causing DNA

single-strande breaks (ssDNA). ssDNA and DNA double-stranded breaks (DSB)

generated during repair of single stranded DNA recognized by ATR and ATM kinases

respectively which master regulate downstream signaling (all targets not shown),

including activation of Chk2 and p53. Tumor virus oncoproteins modulate the function

of DDR components by activating (green arrows), suppressing (shown in red) or directly

interacting with ATR, ATM, Chk2 and p53 proteins. From (Nikitin and Luftig, 2012).

17

Latent infection of large DNA tumor viruses drives robust proliferation of

infected cells and thus, activates a tumor suppressive DDR. -herpesvirus KSHV

encodes a host cyclin-D homolog, viral cyclin (v-cyclin), that binds to CDK6 and drives

cellular proliferation (Verschuren et al., 2004). Furthermore, v-cyclin ectopic expression

alone is sufficient to induce the DNA damage response (Koopal et al., 2007; Leidal et al.,

2012). Moreover, KSHV infection of immortalized endothelial cells in vitro induces the

ATM signaling pathway (Koopal et al., 2007). Consistently, latently infected primary

human foreskin fibroblasts display elevated levels of γ-H2AX and 53BP1 foci (Leidal et

al., 2012). Furthermore, investigation of KS tumors revealed activation of the DDR in

early (patch), but not late (nodular), KS lesions (Koopal et al., 2007). Finally, the

consequences of the activated DDR include the induction of oncogene-induced

senescence and autophagy (Leidal et al., 2012).

Hepatitis B virus (HBV) promotes cellular proliferation and the DDR through the

pleiotropic oncoprotein HBx. Heterologous expression of HBx increases cytosolic Ca2+

levels leading to activation of Pyk2 and c-Src kinases (Klein and Schneider, 1997) and,

ultimately, activation of Ras/Raf/MEK/ERK pathways. HBx expression can also promote

p38MAPK pathway activation which up-regulates E2F-dependent gene expression

(Wang et al., 2008). Constitutive activation of these signaling pathways leads to

activation of the ATR arm of the DDR pathway (Wang et al., 2008).The consequences of

18

this activation are actually beneficial for virus replication despite being tumor

suppressive (Zheng et al., 2011).

1.4.1.2. Viral proteins directly induce a beneficial DDR, including the DNA repair and

activation of checkpoints

Beyond the growth suppressive functions of the DDR, DNA repair and

activation of checkpoints may be beneficial for the replication of tumor viral genomes.

Oncogenic viruses have therefore developed mechanisms to activate specific

components of the DDR pathway, while strictly preventing downstream induction of

apoptosis. Recent work indicates that SV40 large T antigen can serve as both a substrate

for the ATM kinase as well as its direct upstream activator through binding the Nbs1

component of the ATM-activating Mre11/Rad50/Nbs1 complex (Boichuk et al., 2010; Wu

et al., 2004). ATM activation is actually necessary for viral DNA replication (Zhao et al.,

2008b). However, as discussed below, the growth-suppressive consequences of ATM

activation are attenuated by large T antigen downstream enabling SV40-infected cell

survival.

HPV-infected cells display increased, but non-canonical ATM pathway

activation. In particular, HPV oncoprotein-expressing undifferentiated keratinocytes

display an activated DDR characterized by ATM, Chk1, Chk2, and H2AX

phosphorylation (Moody and Laimins, 2009). However, upon differentiation of these

cells, which increases viral genome replication, an additional set of ATM targets is

19

phosphorylated including Nbs1 (Moody and Laimins, 2009). Interestingly, E7 was

demonstrated to associate with the activated Ser1981-phoshorylated form of ATM

independent of differentiation or other viral proteins (Moody and Laimins, 2009).

Therefore, direct association between E7 and phospho-ATM, HPV episome

amplification, and viral-induced replicative stress are all capable of activating the DDR

and it remains unclear which of these activities is critical in regulating HPV

pathogenesis (Moody and Laimins, 2009).

1.4.1.3. DDR activation through viral oncoprotein-mediated mitotic effects

Tumor viruses perturb normal cell cycle control in order to establish a

constitutive S phase-like environment in which cellular factors present are required for

viral replication. One consequence of this constitutive S-phase induction is inappropriate

entry into mitosis which activates DDR checkpoints including those triggered by Chk2

(Sato et al., 2010; Stolz et al., 2010). It was previously shown that KSHV v-cyclin

expression promotes polyploidy and cytokinesis defects (Verschuren et al., 2002) and it

was confirmed later by Ojala and colleagues that v-cyclin expression promotes

amplification of centrosomes and intra-S-phase growth arrest (Koopal et al., 2007).

Moreover, chemical inhibition of ATM/Chk2 led to aberrant mitoses and mitotic

catastrophe in v-cyclin-expressing cells (Koopal et al., 2007).

20

In order to successfully transform cells, SV40 large T antigen targets the spindle

assembly checkpoint component Bub1 leading to ATM/ATR activation (Cotsiki et al.,

2004; Hein et al., 2009). Similarly, the high-risk HPV16 E6 and E7 proteins have been

well documented to increase genomic instability by deregulating mitosis through the

induction of multipolar spindles and centrosome duplication (Duensing et al., 2000).

Specifically, E7 binding to nuclear mitotic apparatus protein 1 appears to deregulate

normal chromosome alignment during prometaphase (Nguyen and Munger, 2009).

More recently, E7 was observed to up-regulate Polo-like kinase 4 (PLK4) expression

leading to centriole multiplication (Korzeniewski et al., 2011). Therefore, multiple viral

oncoproteins perturb mitosis through diverse mechanisms leading to an activated DNA

damage response.

1.4.1.4. Tumor viruses activate the DDR through induction of reactive oxygen species

Elevated levels of reactive oxygen species (ROS) can activate DDR pathways and

may result in mutagenesis during oncogenic virus infection promoting tumorigenesis.

Several tumor virus oncoproteins have been shown to increase ROS levels. For example,

HTLV-1 Tax expression in fibroblasts or T cells induced a ROS-dependent DDR,

although the mechanism by which ROS was induced remains unknown (Kinjo et al.,

2010). Recently, Masucci and colleagues identified the EBV protein EBNA1, which is

required for viral episome maintenance and therefore expressed in every EBV-positive

tumor, as an inducer of ROS and consequent ATM-dependent DDR activation and

21

ultimately chromosomal aberrations (Gruhne et al., 2009a). Interestingly, EBNA1

induced ROS through up-regulation of the mRNA encoding the catalytic subunit of the

leukocyte NADPH oxidase NOX2, which directly promotes ROS accumulation (Gruhne

et al., 2009a). A more recent study suggests that this EBNA1-driven ROS accumulation

may promote telomere dysfunction, another known molecular signal for DDR activation

(Kamranvar and Masucci, 2011).

1.4.2. Tumor viruses modulate the activated DDR to promote tumorigenesis

With the explicit purpose of providing an environment for virus replication,

several tumor virus oncoproteins mitigate the growth suppressive function of the DNA

damage response through altering downstream signaling events. However, the

consequences of suppressing the DDR include aneuploidy and increased mutagenesis

which are major drivers of tumorigenesis. Tumor viruses have been well characterized

to antagonize the function of the p53 tumor suppressor and more recently several

viruses have been shown to target upstream checkpoint kinases as well.

1.4.2.1. Tumor virus suppression of downstream DDR signaling components

The small DNA tumor viruses SV40 and HPV have been well characterized for

their ability to transform cells through perturbing activation of the DDR downstream

target p53 (Kress et al., 1979; Lane and Crawford, 1979; Scheffner et al., 1993; Scheffner et

al., 1990). This activity is thought to be a requirement for cell survival following aberrant

22

S phase induction due to Rb antagonism by T Ag and E7 as described above. While large

DNA tumor viruses generally do not directly promote p53 degradation or abolish its

function, the KSHV latent protein LANA and EBV latent protein EBNA3C have been

shown to modulate p53 activity through direct association (Chen et al., 2010; Friborg et

al., 1999; Yi et al., 2009). Other tumor viruses also directly antagonize p53 function

including the HBV oncoprotein HBx, which both inhibits p53 DNA binding activity and

sequesters p53 in the cytoplasm thereby suppressing apoptosis (Elmore et al., 1997;

Takada et al., 1997; Wang et al., 1994; Wang et al., 1995). HTLV-1 Tax suppresses p53 by

directly antagonizing its trans-activating function through both NF B-dependent and

NF B-independent pathways (Ariumi et al., 2000; Miyazato et al., 2005; Pise-Masison et

al., 2000). While many tumor virus oncoproteins have been shown to associate with p53,

the extents to which these activities contribute to pathogenesis remain unclear.

In addition to targeting p53-downstream signaling, γ-herpseviruses evolved

different strategies to escape from the consequence of the activated DDR. EBV EBNA3C

latent protein downregulates the speed of proliferation and thus attenuates the extent of

the activated DDR, as demonstrated by our group and is discussed below. Unlike EBV,

closely-related KSHV rather actively targets the DDR downstream signaling.

Specifically, KSHV encoded v-FLIP suppresses v-cyclin-driven oncogene-induced

senescence, presumably via blocking autophagy (Leidal et al., 2012). Therefore, the

23

remaining DNA damage promotes mutagenesis and selection for mutations in the DDR

pathway in advanced KS lesions allowing tumor cell survival.

1.4.2.2. Viral oncoproteins directly target DDR checkpoint kinases

Upstream of p53 and cell cycle checkpoints are a series of DNA damage sensing

and signal relaying kinases (Figure 1). Several viral oncoproteins directly target these

upstream kinases through a number of mechanisms ultimately attenuating their

function. For example, the HTLV-1 Tax oncoprotein directly binds to and inhibits

signaling downstream of both Chk1 and Chk2 checkpoint kinases (Gupta et al., 2007;

Park et al., 2004; Park et al., 2006) as well as the upstream DNA damage sensing DNA-

PK (Durkin et al., 2008). Interestingly, Tax was also demonstrated to sequester the DDR

components MDC1, DNA-PK and BRCA1 at artificial Tax-induced foci of pseudo-DNA

damage as a unique mechanism to perturb endogenous DDR signaling pathways

(Belgnaoui et al., 2010). Not unexpectedly, Tax expression attenuated ATM-downstream

signaling leading to faster release of the G1/S checkpoint in response to ionizing

radiation (Chandhasin et al., 2008).

Under circumstances where EBV oncoproteins are aberrantly expressed, as

evidenced in heterologous expression studies in EBV-negative B cells, the DDR

pathways can be directly attenuated. Specifically, Robertson and colleagues have

observed a direct interaction between EBNA3C and Chk2 leading to decreased Chk2

24

activity, which may also contribute to DDR attenuation during primary B cell outgrowth

(Choudhuri et al., 2007). Another study identified the latent membrane protein LMP1 as

an inhibitor of ATM signaling due to transcriptional down-regulation of ATM upon

LMP1 over-expression (Gruhne et al., 2009b). Under certain circumstances, such as in

Hodgkin’s lymphoma or nasopharyngeal carcinoma where LMP1 is expressed at high

levels and may be important for cell survival, this activity may contribute to

tumorigenesis due to the inability of ATM to trigger checkpoints and mediate efficient

DNA repair.

1.4.2.3. Viral oncoproteins perturb mitotic checkpoint signaling

Mitotic checkpoints are often provoked by viral oncoprotein promotion of cell

cycle progression as discussed above. Therefore, in order for these viruses to replicate in

the infected cell, signaling downstream of the G2/M checkpoint must be attenuated.

Several oncogenic viruses encode proteins that precisely target this checkpoint with

potentially catastrophic consequences on the karyotype of surviving cells. HTLV-1 Tax

expression abolishes cellular mitotic checkpoints through directly targeting and

prematurely activating the anaphase promoting complex (Liu et al., 2005) as well as

suppressing the spindle assembly checkpoint protein Mad 1 (Jin et al., 1998) resulting in

highly aneuploid ATL cells. Similarly, the EBV EBNA3 proteins are capable of inhibiting

the canonical G2/M checkpoint through suppression of p27 levels or activity depending

on the cell type (Knight and Robertson, 2004; Parker et al., 2000; Wade and Allday, 2000).

25

Specifically, EBNA3C is capable of suppressing the effects of mitotic poisons in part

through decreasing the levels of the spindle assembly checkpoint protein BubR1

(Gruhne et al., 2009b; Leao et al., 2007). The consequence of bypassing the mitotic

checkpoint and DDR signaling downstream is the accumulation of aneuploid cells that

can promote tumorigenesis through copy number amplification of oncogenes or loss of

tumor suppressors.

In summary, the DDR can be activated directly by aberrant expression of

oncoproteins, cellular or viral, or as a consequence of cellular proliferation-induced

replicative stress. DNA tumor virus-driven cellular transformation occurs as a by-

product of the virus promoting the cell cycle in order to establish an appropriate

environment with the requisite DNA replication machinery and repair factors necessary

for viral DNA replication. Similarly, viruses such as HTLV-1 must activate the infected T

cell in order to promote a favorable environment for proviral DNA integration.

However, in the inadvertent setting such as following aberrant integration of viral

genomes where loss of normal viral replication function occurs or other changes lead to

increased viral oncoprotein expression, a constitutively activated DDR is triggered. DDR

signaling typically limits viral oncogenesis, but also provides selective pressure for

mutations in DDR signaling components that promote tumorigenesis. The delicate

balance between virus replication, latency, and the extent of activation of the DDR

26

ultimately dictates whether an infected cell will give rise to a productive cycle

generating progeny virions or a tumor.

27

2. Results

2.1. An ATM/Chk2-Mediated DNA Damage-Responsive Signaling Pathway Suppresses Epstein - Barr virus Driven Proliferation of Primary B cells

2.1.1. Contributions

The following Chapter 2.1, including text and figures, is adapted from (Nikitin et

al., 2010) with permission from Cell Press. This paper was published in co-first

authorship between Pavel Nikitin and Christopher Yan. Micah Luftig, Pavel Nikitin and

Christopher Yan designed and performed experiments with significant help from Jason

Tourigny, Eleonora Forte and Sandeep Dave. P.N. designed the method to sort

populations of EBV infected B cells and discovered the early proliferating cells

phenotype. C.Y. demonstrated the activation of the DDR by immunofluorescent

microscopy and revealed the role of viral DNA replication by fluorescent in situ

hybridization. E.F. revealed the initial activation of the DDR by Western and prepared

samples for microarray. M.L., J.T. and S.D. analyzed and confirmed the expression

changes. Martin Allday and Rob White kindly shared viral mutants and critically

discussed the manuscript. P.N. and C.Y. partially wrote the manuscript, and M.L. wrote

the final version of the paper. Finally, I would like to thank our co-authors Alessio

Bocedi, Amee Patel, William Kim, Katherine Hu, Jing Guo, David Tainter and Elena

Rusyn for their invaluable help in performing experiments, analyzing expression data

and running the lab in 24/7 schedule.

28

2.1.2. Introduction

As mentioned above, a convenient in vitro system allows identifying early event

upon EBV infection of primary human B cells. Precluding observation that only few

proliferating cells are transformed into indefinitely proliferating lymphoblasts

(Henderson et al., 1977; Sugden and Mark, 1977) led us to hypothesize that an active

host tumor suppressive mechanism prevents the EBV tumorigenesis.

Innate tumor suppressor responses have been better characterized in other

systems. The DNA damage response, overviewed in Chapter 1.3., is triggered by

aberrant replication structures generated by activated oncogenes attempting to

constitutively fire new origins and inappropriately enter S phase (Halazonetis et al.,

2008). The DDR limits aberrant proliferation by mediating oncogene-induced senescence

and apoptosis (Bartkova et al., 2006; Di Micco et al., 2006). Signaling downstream of

oncogenic stress involves activation of the single-stranded DNA-dependent ATR

pathway and the double-stranded break-induced ATM pathway. These DDR kinases

relay downstream signals to critical repair factors and other checkpoint kinases

including Chk1 and Chk2 with extensive cross-talk ultimately resulting in suppression

of oncogene-induced proliferation (Halazonetis et al., 2008; Stiff et al., 2006). Genetic

experiments have identified critical roles for ATM and Chk2 in mediating oncogene-

induced senescence and tumor suppression (Bartkova et al., 2006; Pusapati et al., 2006;

Stracker et al., 2008). Given these observations and the low efficiency of EBV

29

transformation, the intriguing question remains as to whether the host DNA damage

response senses EBV-induced oncogenic stress and, importantly, if this is responsible for

the block to long-term outgrowth of the majority of infected cells.

2.1.3. Results

2.1.3.1. Epstein-Barr virus infection of primary B cells activates a cellular DNA

damage response

We first sought to determine whether EBV infection of primary B cells might

drive an oncogenic stress leading to the activation of the DNA damage response.

Purified CD19+ B cells were infected with the prototypical transforming EBV strain B95-

8 at a multiplicity of infection (MOI) of ~5. Nearly all cells were EBV genome positive as

determined by fluorescence in situ hybridization (FISH) (Fig. 3A).

30

Figure 3: Nuclear genome deposition and the analysis of viral and host gene

expression following primary B cell infection with EBV B95-8, UV-inactivated B95-8,

P3-HR1 (A-C). EBV genomes in infected primary B cells are independent of -H2AX

foci (D-G).

(A) Fluorescence in situ hybridization (FISH) of viral genomes 2 days post B cell

infection with EBV B95-8, UV-inactivated B95-8 (UV B95-8), and P3HR1 (P3) at an MOI

of ~5. (B) Quantitative RT-PCR of the interferon responsive mRNAs ISG15 and IFIT4 24h

following PBMC infection with B95-8 and UV-inactivated B95-8. mRNA levels were

normalized to GAPDH and primers were as described previously (Martin et al., 2007)

(C) Quantitative RT-PCR for Wp-initiated mRNAs 48h after PBMC infection with B95-8,

UV-B95-8, or P3 virus strains. Analysis of the relative levels of W0 – W1/W2 containing

mRNAs indicate that infection was equivalent between B95-8 and P3, while UV-

inactivated virus was essentially unable to produce Wp-initiated transcripts. mRNAs

were normalized to GAPDH and primers for these experiments were previously

described (Bell et al., 2006). (D) Fluorescence in situ hybridization (FISH) of EBV

genomes (green) in B95-8 Z-HT cells (Johannsen et al., 2004). Uninduced cells (-HT)

31

mostly contained latent episomes (left). A representative cell undergoing lytic

replication following exposure to 4-hydroxytamoxifen (+HT) is shown on the right. (E)

EBV FISH in B cells 5 days and 14 days after infection. Intense lytic staining was rarely

observed 5 days after infection (left). Approximately 1-5% of cells were undergoing lytic

DNA replication by FISH at 14 days (right). (F) Left, Representative images of EBV

genomic FISH in primary CD19+ B cells at 1 and 5 days post infection with B95-8 (MOI

~5), steady state 8-10 genomes in a LCL, and 50 genomes per Raji cell. Right, Episome

number as determined by FISH at different times after infection. The y-axis represents

the average number of episomes per cell, the x-axis represents days after infection when

cells were collected and fixed for hybridization (5, 7, 10, 14, 21, and 35 days). Vertical

dashed lines show the beginning and end of the period in which the DNA damage

response (DDR) was observed. Error bars represent SEM. These data were collected

from greater than 50 nuclei at each time point for three independent normal donors. (G)

Left, IF/FISH of H2AX (red) and EBV genomes (green) in B cells 7 days post infection.

Left cell shows IF/FISH, right cell shows only IF for H2AX with white arrows

representing EBV genome location. The right panels are additional representative

IF/FISH and IF images.

Infected cells were initially assayed for the expression of the earliest viral latency

gene product, EBNA-LP, and the DNA damage marker, -H2AX, at different times post

infection. -H2AX activation was not evident prior to 4 days post infection, was robust

from 4 to 7 days post infection, and declined after 7 days to the low levels observed in

LCLs (Fig. 4A and data not shown). Approximately 60% of the infected cells were -

H2AX positive at 7 days post infection. Corroborating our findings of -H2AX

activation, EBV infection induced additional hallmarks of the DDR including auto-

phosphorylation of the H2AX kinase ATM (pATM Ser1981), and punctate localization of

the damage adaptor 53BP1 (Fig. 4B and 4C).

32

Figure 4: EBV induced a DNA damage response in primary B cells.

(A) Indirect immunofluorescence (IF) images of EBNA-LP (green) and -H2AX

(red) in uninfected B cells, B cells 4 and 7 days after infection with EBV B95-8 (MOI ~5),

the recently derived LCL EF3D, and uninfected -irradiated B cells (0.2, 1, and 5 Gy, 1h).

DNA is stained with DAPI. These images are representative of infections in five

different normal donors. (B) Ser1981 phosphorylated ATM (pATM, red) in uninfected B

cells and B cells 7 days after EBV B95-8 infection. EBNA-LP or other EBV latent antigen

staining was not possible in these samples due to antibody source, however we know

that ~50% of these infected cells are EBNA-LP-positive. (C) EBNA-LP (green) and 53BP1

(red) in uninfected B cells and B cells 7 days after infection. (D) EBNA-LP (green) and -

H2AX (red) in B cells 4 days after infection with UV-inactivated EBV B95-8 (UV EBV) or

the non-transforming EBV strain P3HR1 (P3).

33

EBV gene expression was important for virus-induced DDR activation. Cells

infected with UV-inactivated B95-8 virus did not show -H2AX staining at any point

within the first week after infection (Fig. 4D and data not shown). Importantly, UV-

inactivated EBV B95-8 genomes reached the nucleus and these infections induced

interferon-responsive genes (Fig. 3A and B). EBNA2 and latency III gene expression was

specifically necessary to induce the DDR as B lymphocytes infected with the EBNA2

deleted, transformation-incompetent P3HR1 strain of EBV did not contain -H2AX foci

(Fig. 4D) despite similar levels of infection compared to B95-8 (Fig. 3A-C). These data

collectively demonstrate that EBV latent gene expression rather than simply virion

binding or nucleic acid deposition into the nucleus was required to induce -H2AX

activation.

2.1.3.2. The EBV-induced DNA damage response in primary B cell infection is not

associated with viral episomes or lytic replication

We reasoned that either viral or cellular DNA may activate the DNA damage

response. Since evidence in the literature suggested that either viral lytic DNA

replication (Kudoh et al., 2005) or latent viral episome replication (Dheekollu et al., 2007)

may be capable of inducing a DDR, we first assayed viral DNA as a possible source of

the damage. Incoming linear viral DNA was not the source of the damage since UV-

irradiated and EBNA2-deleted P3HR1 virus infections did not induce the DDR (Fig. 4).

We next used a FISH based assay to assess the possible role of lytic DNA replication.

34

The B95-8 Z-HT cell line was used as a positive control where lytic EBV DNA was

recognized as a brightly staining FISH signal rather than the punctate foci of episomal

genomes (Fig. 3D). Less than 1% of EBV-infected cells contained evidence of lytic viral

DNA 5 days post infection, while approximately 1-5% of infected cells were

spontaneously undergoing lytic replication by 14 days similar to that found in LCLs

(Fig. 3E and (Kieff and Rickinson, 2006)). Since far greater than 1% of EBV-infected cells

were -H2AX positive early after infection, we conclude that viral lytic DNA replication

is not responsible for DDR activation.

Next we assessed the possibility that latent viral episomes activate the

DNA damage response. The mean episome number per cell as assessed by FISH did not

increase during the period when -H2AX activity was high early after infection (Fig. 3F).

Furthermore, we failed to observe significant co-localization of EBV episomes with -

H2AX foci in these cells (Fig. 3G). In fact, the number of -H2AX foci per cell was

consistently much greater than the number of EBV genomes (Fig. 3G). Therefore, our

data collectively suggest that the observed EBV-induced DDR is not activated by viral

DNA.

2.1.3.3. The EBV-induced DNA damage response is associated with a transient period

of hyper-proliferation

We next focused our studies on changes in cellular DNA that may induce a DDR.

The period of time post infection when the DDR was active correlates with the initiation

of B cell proliferation (Kieff and Rickinson, 2006). Analysis of CD19+ B cells using the

35

proliferation-tracking dye CFSE at different days after infection indicated that i)

proliferating cells appeared at day 3 (Fig. 5A), ii) between days 3 and 4 there were

always cells that had divided more than once or even twice in 24h, and iii) at later days

post infection cells appeared to proliferate at a slower rate as judged by the less

pronounced shift of the CFSE profile to the left.

Figure 5: EBV induced a period of hyper-proliferative early after infection that

was associated with activation of the DNA damage response.

36

(A) Histograms show CD19+ B cell division as measured by CFSE staining at

different days after EBV infection. Mock, mock infected cells. (B) The mean division

number based on precursor cohort analysis for EBV-infected B cells is plotted at

different times post infection. Vertical dashed lines estimate the hyper-proliferation

period. Data are presented from 5 normal donors. (C) IF of -H2AX (red) and EBNA-LP

(green) of uninfected cells, infected cells that have yet to divide (PD0), infected cells after

1 or 2 divisions (PD1-2), or 7 or more divisions (PD7+) and LCLs. DNA is stained with

DAPI. (D) The percentage of EBNA-LP positive cells with -H2AX signal >5X over

background is graphed from uninfected B cells, sorted PDs, and LCLs. Uninfected B

cells following 0.2, 1, and 5 Gy (1hr) -irradiation are also shown as a positive control.

These data are representative of similar experiments from three independent normal

donors. (E) Immunoblot of p-Chk2 Thr68 and Chk2 in sorted cells as in (D) including an

LCL following 5 Gy -irradiation (1hr). (F) The percentage of EBNA-LP positive cells

containing 4 or more 53BP1 foci per cell in sorted populations as in (D) are shown along

with uninfected irradiated B cell controls. PD3-4 contained significantly more 53BP1 foci

per cell than uninfected B cells (p<0.0001), PD0 (p<0.0001), PD7 (p<0.01), and LCL

(p<0.0001).

A more rigorous kinetic analysis of EBV-induced B cell expansion highlighted

the biphasic nature of the proliferation rate (Fig. 5B). Infected CD19+ B cell CFSE profiles

from five normal donors were analyzed at time points prior to and during the first seven

cell divisions. The mean division number (MDN) at each time point was determined by

fitting the precursor-normalized number of cells in each division to a Gaussian

distribution (Fig. 6A and (Hawkins et al., 2007a)).

37

Figure 6: CFSE-based kinetic analysis of EBV-induced proliferation.

(A) Analysis of EBV-infected PBMC stained by CFSE was performed by FACS at

different time of infection (141 hour post infection point is shown, left). CFSE-based

population doublings of CD19+ B cells were determined using the “Proliferation” tool in

the FlowJo program. Number of cells in each PD (N) was normalized to averaged

division number i (i=i+0.5) and fit to a Gaussian distribution for each time point. Mean

division number was determined as shown (MDN=3.09 for 141 hour p.i. for a given

donor, middle). Then, MDN over time post infection plot was generated for each time

point (right). (B) CD19+ cells from population doublings 0, 1, 2 and 3 were double-

sorted by flow-cytometry and incubated separately for 48 hours. Then, proliferative

potency of cells from each population doubling was measured by FACS as a percent of

cells divided more than once, twice or thrice. The data shown is the average for 2

donors. Error bars represent standard error.

The slope of the function relating MDN to time post infection inversely correlates

with the proliferation rate. Consistent with the data in Fig. 5A, we observed that EBV

induced an early phase of hyper-proliferation that was attenuated over time (Fig. 5B).

The proliferation rate of initially proliferating cells was approximately once per 8-12h

38

while later cycles were ~24-30h similar to the ~24-28h rate of LCLs. These findings were

corroborated by cell sorting experiments where cells from earlier divisions proliferated

more quickly than those in later divisions (Fig. 6B). Thus, EBV-mediated B cell

expansion proceeds through an initial period of hyper-proliferation followed by slower

cell divisions typical of emergent LCLs.

We next asked whether the DNA damage response was activated specifically

during the hyper-proliferative divisions independent of time post infection. EBV-

infected B cells sorted based on population doubling (PD) were subjected to

immunofluorescence for EBNA-LP and -H2AX (Fig. 5C). Sorted cells were >85% EBNA-

LP positive in cells not yet dividing (PD0) and >95% EBNA-LP positive in all later PDs.

We observed a robust increase in LP+/ -H2AX+ cells during the early PDs (1-2 and 3-4)

relative to uninfected cells or infected cells not yet proliferating (PD0) (Fig. 5C and 5D).

Importantly, this response was attenuated through later PDs and in LCLs. Moreover, -

H2AX intensity per cell was significantly higher in PD3-4 than PD0 (p<0.0001) and LCL

(p<0.0001). We also observed a transient activation and attenuation of the ATM-specific

phosphorylation of Chk2 on Thr68 (Fig. 5E) as well as accumulation of 53BP1 into DDR

foci (Fig. 5F). These data strongly support the notion that the EBV-induced DDR is

caused by an early period of hyper-proliferation and is attenuated during LCL

outgrowth.

39

2.1.3.4. Proliferation and DNA damage responsive genes are highly induced early

after EBV infection, then attenuated during LCL outgrowth

Our cell-based findings were corroborated by mRNA microarray studies of i)

uninfected B cells, ii) EBV-infected early proliferating cells (Prolif), and iii) monoclonal

LCLs from four normal donors (Fig. 7). We first asked in an unbiased manner which

genes were significantly changed upon proliferation and then, subsequently, during

LCL outgrowth (2-way ANOVA, p<0.01). As expected, the most enriched gene ontology

(GO) category for genes induced from resting B cells to EBV-infected, proliferating B

cells was ‘Cell Proliferation’ (Fig. 7A; GO:0008283, Bayes factor: 51, p<0.0001 (Chang and

Nevins, 2006)).

40

Figure 7: Transcriptional changes correlate with an EBV-induced early period

of hyper-proliferation and DNA damage response followed by attenuation upon LCL

outgrowth.

(A) Heatmap of average expression data across four normal donors for the gene

ontology (GO) category “Cell Proliferation” in uninfected resting B cells (B cell), EBV-

infected early proliferating B cells (Prolif), and monoclonal LCLs (LCL). The genes

presented were derived from GATHER analysis of all genes with significant expression

changes (2-way ANOVA, p<0.01) where the expression level increased from B cell to

Prolif at least 1.5-fold and decreased from Prolif to LCL at least 1.2-fold (left). Heatmap

of individual samples of top 20 “Cell Proliferation” genes (right). (B) Heatmap of

“Response to DNA Damage Stimulus” GO genes across individual samples. (C) Gene

Set Enrichment Analysis (GSEA) of known DNA damage induced ATM and p53-

depending genes in the context of B-Prolif-LCL expression data. The reference list of

41

ATM/p53 target genes was derived from Clusters 2 and 3 of (Elkon et al., 2005) and

compared with a pre-ranked list (by fold) of global average gene expression changes

from B cell to Prolif (left) and Prolif to LCL (right). Statistical scores are inset into the top

right of analysis images (NES: Normalized enrichment score and FWER: Familywise

error rate).

Genes associated with the ‘Response to DNA Damage Stimulus’ were also highly

induced (Fig. 7B; GO:0006974, Bayes factor: 17, p<0.0001). Notably, we observed that the

majority of genes involved in cell proliferation and the DNA damage response were

consistently repressed as cells transitioned from early proliferating to established LCLs

(Fig. 7A, Cell Proliferation, Bayes factor: 63, p<0.0001 and Fig. 7B, Response to DNA

Damage Stimulus, Bayes factor: 22, p<0.0001). Consistently, the expression of genes in an

independently derived set of DNA damage responsive and ATM-dependent p53 targets

(Elkon et al., 2005) was also increased in early proliferating cells and subsequently

attenuated during LCL outgrowth (Fig. 7C and 8).

42

Figure 8: Schematic diagram of GSEA analysis performed on B cells, Proliferating

cells, and LCL microarray data with ATM/p53 target genes.

We imported our average mRNA expression changes from either B to

Proliferating (Prolif) or Prolif to LCL as molecular profiles. Then we assigned the ATM

and p53 target genes as identified from (Elkon et al., 2005) as a gene set into GSEA. The

enrichment of this gene set relative to 1000 random permutations within the B to Prolif

samples was plotted in the data in Fig. 4C, which indicated that EBV-induced

proliferation was associated with an activation of the ATM/p53 gene expression

signature, while from proliferation through LCL outgrowth this gene set was depleted

indicating attenuation of ATM/p53 target gene expression.

Collectively, these global gene expression analyses corroborate our findings of a

period of hyper-proliferation and activation of an ATM-dependent DNA damage

response early after infection that is attenuated during LCL outgrowth.

43

2.1.3.5. The EBV-induced hyper-proliferation associated DNA damage response is

growth suppressive

To further analyze the consequences of the activated DDR in early rapidly

proliferating cells, we designed a sorting strategy to assess the relative growth potential

and DDR activation in cells derived from early or late divisions after infection (Fig. 9A).

We initially stained cells with the proliferation tracking dye PKH26 and sorted cells after

infection for PD1-4 and PD6+ populations (Fig. 9B).

44

Figure 9: Growth suppression and DNA damage enrichment in early cell divisions.

(A) A flowchart shows the separation of arrested and proliferating EBV-infected

CD19+ PBMC used for IF and FACS. PBMC were first infected with EBV and labeled

with the red fluorescent dye PKH26. (B) Then, 8 days post infection, proliferating CD19+

B cells were sorted for PD1-4 and PD6+ based on PKH26 intensity, labeled with CFSE,

and cultured for two days. (C) Sorted cells were then analyzed in a FACS-based growth

assay where cells in the CFSElow population were considered proliferating and cells in

the CFSEhi population were considered arrested. Forward scatter (FSC) low reflects

dying cells. Results are representative of three normal donors. (D) PKH26low (PD1-4)

cells were subsequently labeled with CFSE as above, sorted after 48h in culture into

CFSEhi (arrested) and CFSElow (proliferating) populations, and analyzed by IF for -

H2AX (red).

45

Subsequent staining with CFSE enabled the analysis of proliferation from these

populations. Supporting our hypothesis, the cells in early hyper-proliferating divisions

(PD1-4) were, in fact, more prone to growth arrest and cell death than those in later

divisions (PD6+) and LCLs (Fig. 9C). Consistently, arrested PD1-4 cells displayed

significantly more intense -H2AX staining than their proliferating counterparts (Fig.

9D).

2.1.3.6. ATM and Chk2 kinases suppress EBV-mediated transformation and initial B

cell proliferation

To determine if the activation of the DDR restricts EBV-mediated long-term

outgrowth, we simultaneously infected peripheral blood mononuclear cells (PBMC)

with EBV and treated them with an inhibitor of either ATM (ATMi (Hickson et al.,

2004)) or its downstream effector kinase Chk2 (Chk2i (Arienti et al., 2005)); both are

critical kinases in the DDR checkpoint responding to DNA double-stranded breaks and

oncogenic stress. EBV-mediated B cell transformation efficiency increased in a dose-

dependent manner in response to the inhibitors where 2 M ATMi increased efficiency

by approximately 2-fold and 5 M ATMi by 6-fold over DMSO control treated cells (Fig.

10A). Similarly, Chk2 inhibition increased EBV transformation efficiency approximately

3-fold for 2 M Chk2i and 9-fold for 5 M Chk2i (Fig. 10B). Therefore, an ATM and Chk2

dependent DDR restricts EBV transformation.

46

Figure 10: Inhibition of ATM and Chk2 kinases increased EBV transformation

efficiency and proliferation of B cells during a critical period 4-8 days post infection.

(A) Quantification of EBV-induced B cell outgrowth following PBMC infection in

the presence of 0.1% DMSO (black), 2 μM ATMi (green), or 5 μM ATMi (red). The

percentages of wells positive for LCLs at five weeks post infection are plotted relative to

the transforming units (TU) of B95-8 virus per well. Results shown are the average of

experiments with at least four independent normal donors. Error bars represent

standard error of the mean (SEM). (B) Similar experiments were performed using DMSO

(black), 2 µM Chk2i (green), or 5 µM Chk2i (red). (C) CFSE-stained PBMC were infected

with EBV in the presence of increasing amounts of ATMi or Chk2i (DMSO, 1 µM, 2 µM,

5 µM, and 10 µM). The percentage of CD19+/CFSElow cells of total PBMCs at 14 days post

infection are plotted. The data shown are the average values from two different donors

+/- SEM. These data are representative of more than five independent experiments. (D)

Dot plots show CFSE- and CD19-stained PBMC that were treated with DMSO, 5 µM

ATMi, 5 µM Chk2i, or infected with EBV for six days. (E) This table summarizes when

ATM and Chk2 suppressed EBV-mediated proliferation at different times following

infection. CFSE-stained PBMC were infected with EBV at day 0. ATMi or Chk2i (5 μM)

was added at different times after infection (top) or at day 0 and washed out at different

times after infection (bottom). EBV-mediated B cell proliferation was detected by FACS

at day 14 post infection using CD19-PE and CFSE as in (C). A more than two-fold

47

increase in treated cells versus DMSO is represented by a green plus; a less than two-

fold increase is represented by a yellow plus; and no increase is represented by a red

dash. The lines indicate the period of incubation and are colored with the proliferation

phenotype after ATMi and Chk2i treatment. Average values from two independent

donors are shown. (F) EBV-induced outgrowth following PBMC infection was measured

as in (A) in the presence of 5 μM Chk2i (blue), 5 μM ATMi (red), or DMSO (black) added

at day 0, day 4 or day 12 after EBV infection. Results shown are the average of four

independent normal donors +/- SEM. (G) Efficiency of EBV outgrowth from (F) was

calculated and the average ratio of inhibitor-treated to DMSO-treated infections +/- SEM

for four normal donors is plotted.

We next assessed whether ATM and Chk2-mediated suppression of EBV

transformation was due to limiting initial B cell proliferation. The continuous presence

of either ATM or Chk2 inhibitor led to a dose dependent increase in B cell number at

two weeks post infection (Fig. 10C). Importantly, ATM or Chk2 inhibitor did not induce

B cell proliferation in the absence of EBV suggesting that these compounds act to

alleviate a block to proliferation rather than stimulating B cells per se (Fig. 10D).

2.1.3.7. ATM and Chk2 suppress B cell growth 4-8 days after EBV infection

Since the DDR peaked during the first week after infection, we assessed when

ATM and Chk2 inhibition enhanced proliferation and transformation. To that end,

PBMC infected with EBV were transiently exposed to ATMi and Chk2i from the start of

infection or the compounds were added at different days post infection. EBV-induced B

cell proliferation was most sensitive to the inhibitors between 4 and 8 days after

infection when cells were present in the hyper-proliferative period (Fig. 10E). For

example, when either inhibitor was added within the first 4 days of infection we

48

observed as pronounced an effect on proliferation as if the inhibitor was added at day 0.

However, if we added inhibitors after day 8, there was no effect on proliferation.

Conversely, if the inhibitors were removed prior to 4 days after infection, then increased

proliferation was not observed.

Similar results were obtained in long-term transformation assays.

Addition of either compound within 4 days of infection increased transformation

efficiency, while adding the compounds at 12 days post infection had little effect (Fig.

10F and 10G). The inhibitors also did not increase LCL growth rates at normal or

limiting density (data not shown). Therefore, during a critical period approximately 4-8

days following infection, EBV induced an ATM and Chk2-dependent growth

suppressive signaling pathway that limited initial B cell proliferation and, consequently,

long-term outgrowth into lymphoblastoid cell lines.

2.1.3.8. EBV latent gene expression changes and consequences in early infected cell

divisions

The dynamic changes in proliferation and DDR associated gene expression

support our cell-based assays indicating an early period of ATM/Chk2-mediated growth

suppression that is attenuated in later divisions enabling long-term LCL outgrowth.

However, to determine whether these changes correlated with viral gene expression, we

queried viral transcripts and proteins associated with the latency III growth program in

sorted population doublings after infection (Fig. 11 and 12). Wp-associated transcripts

were expressed at a markedly higher level than Cp transcripts prior to the first infected

49

cell division (PD0) (Fig. 11A). However, this ratio shifted such that Cp levels were

greater after 3-4 cell divisions and through LCL outgrowth consistent with previous

observations (Fig. 11B and (Schlager et al., 1996; Woisetschlaeger et al., 1989;

Woisetschlaeger et al., 1990)). The consequence of the high Wp/Cp ratio was heightened

levels of EBNA-LP protein as well as a heightened EBNA-LP to EBNA3A and 3C protein

ratio in early divisions that waned through LCL outgrowth (Fig. 11C-D). Thus, the initial

cell divisions characterized by hyper-proliferation display a distinct EBNA gene

expression equilibrium that may affect target gene expression.

50

Figure 11: EBV latency and consequential host gene expression changes from

initial B cell proliferation through LCL outgrowth.

(A) Expression of Wp (filled triangles) and Cp (open circles) derived mRNAs in

EBV-infected cells sorted by population doubling (PD) and monoclonal LCLs. Relative

mRNA abundance normalized to a beta-actin control is plotted versus PD. These data

are representative of two normal donors and consistent with published time course

experiments (Woisetschlaeger et al., 1989; Woisetschlaeger et al., 1990). (B) The ratio of

Wp to Cp mRNA expression levels from (A) is plotted versus division and through LCL

outgrowth. (C) Protein expression of EBNA-LP, EBNA2, EBNA3A, and EBNA3C are

shown from sorted infected PDs and a polyclonal LCL from the same donor. (D)

51

Proteins detect by Western blotting from three independent normal donors similar to

those in panel (C) were quantified. The average ratio of total EBNA-LP protein (i.e., all

isoforms) relative to total EBNA3A or EBNA3C +/- SEM is plotted versus PD through

LCL. (E) Average CD23 surface expression as mean fluorescence intensity (MFI) is

plotted versus PD +/- SEM for two donors. (F, left) The expression level of c-Myc mRNA

is plotted versus sorted PD. (F, right) The activity of the c-Myc target gene expression

signature (Bild et al., 2006) is plotted from the average expression of targets in

microarray samples from four independent donors of resting B cells (B), early

proliferating B cells (Prolif), and monoclonal LCLs. Error bars represent SEM.

To more rigorously assess this, we analyzed EBNA2 targets including

CD23 (Wang et al., 1991) and c-Myc (Kaiser et al., 1999). In both cases, these EBNA2

targets were highly induced in early cell divisions and then attenuated through LCL

outgrowth, still remaining significantly higher than resting B cell levels (Fig. 11E-F). The

consequences of the transient increase in c-Myc mRNA was manifested in an increase in

the c-Myc target gene expression signature (Bild et al., 2006) during early proliferating

cells that was attenuated in LCLs, though still greater than resting B cell levels (Fig. 11F).

Given the importance in titrating this potentially genotoxic oncoprotein and the known

role of ATM in suppressing c-Myc oncogenesis (Hong et al., 2006; Murphy et al., 2008),

these findings strongly support a model of acute oncogenic stress early after EBV

infection that is modulated through the well described Wp to Cp switch enabling

modest EBNA2 activity critical for indefinite EBV-infected cell outgrowth.

2.1.3.9. EBNA3C is required to attenuate the EBV-induced DNA damage response

While the induction of the DNA damage response after EBV infection requires

latent gene expression and proliferation, a definitive role for viral latent genes in

52

attenuating this response was not demonstrated. In order to determine which latent

genes are critical for DDR attenuation during late divisions after infection we chose to

interrogate the EBNA3 proteins, EBNA3A and EBNA3C, as they are known to modulate

EBNA2 activity. Infection of primary B cells with EBV B95-8, EBNA3A knockout (KO),

or EBNA3C KO virus (Anderton et al., 2008) supported early B cell proliferation (Fig.

12A-C).

Figure 12: Expression of viral latency genes during early cell divisions through

LCL outgrowth.

The expression of (A) EBNA-3A, EBNA-3B, and EBNA-3C, (B) EBNA-1, (C)

LMP1, and (D) LMP2A was detected by quantitative real-time PCR. EBV-infected PBMC

were sorted 5 and 7 days post infection by CD19 and CFSE for population doublings and

also a polyclonal LCL was derived from the same infected donor (day 35 post infection).

53

RNA was extracted and SYBR green-based qRT-PCR was performed using primers

spanning exons as previously described (EBNA1 Y3/U/K, LMP1 exon 2/3, and LMP2A

exon 1/2 from (Bell et al., 2006) and EBNA-3A (exon1/2), EBNA-3B (exon 1/2), and

EBNA-3C (exon 1/2) from (Sengupta et al., 2006).Data from one normal donor are shown

in the graphs which are representative of three independent infections. The relative

mRNA abundance is plotted normalized to an HPRT mRNA control (primers from

(Ranuncolo et al., 2007)). We note that for each mRNA, an increase is observed from

PD5-6 through LCL outgrowth, which is likely due to an increase in genome copy

number.

However, upon sorting these early proliferating cells we observed that EBNA3C

KO virus infected cells displayed increased activation of the DNA damage response,

while EBNA3A KO infected cells were similar to WT B95-8 infection in DDR activation

(Fig. 13A-B). Indeed, greater than 80% of EBNA3C KO-infected cells were -H2AX

positive relative to ~50% of WT or EBNA3A KO-infected cells (Fig. 13C).

54

Figure 13: EBNA3C attenuates the EBV-induced DNA damage response.

(A) Representative IF images are shown of -H2AX staining (red) from WT,

EBNA3A KO ( 3A), and EBNA3C KO ( 3C) infected and sorted PD1-4 B cells. DAPI

DNA stained (blue) and DAPI/ -H2AX merged images are also shown. (B)

Representative IF images are shown of 53BP1 staining (red) from WT, 3A, and 3C

infected and sorted PD1-4 B cells. (C) Quantification of IF data from (A) is plotted as

percentage -H2AX positive cells. Average values are plotted for infected cells,

uninfected B cells, and 5 Gy -irradiated B cells. (D) Model for EBV-induced DDR/hyper

proliferative period and attenuation during LCL outgrowth. Early in infection EBNA2

and EBNA-LP associate with cellular transcription factors (TF) to potently up-regulate

expression of growth control genes and B cell activation markers, including c-Myc and

CD23, activating the host DNA damage response (left). Later in infection, the activity of

the EBNA3 proteins, in particular EBNA3C, down-regulate EBNA2 function, as LMP1

and LMP2 are up-regulated and may cooperate in the constitutive, but attenuated

expression of host growth control genes and enhanced cell survival (right).

55

Similarly, EBNA3C KO-infected cells accumulated 53BP1 DDR foci to a greater

extent than WT or EBNA3A KO-infected cells (p<0.001, 3C KO v. WT; p>0.1, 3A KO v.

WT). Thus, while B cells infected with either EBNA3A KO or EBNA3C KO virus were

crippled for long-term outgrowth (Fig. 14B-C), these experiments define a critical role

for EBNA3C in attenuating the host DNA damage response to EBV infection early after

infection. These data strongly support our model of a latent gene expression triggered

hyper-proliferation induced DDR, followed by proper expression of the EBNA3

proteins, in particular EBNA-3C, in order to attenuate a potentially genotoxic and

growth suppressive signaling pathway (Fig. 13D).

56

Figure 14: Characterization of EBNA-3A KO and EBNA-3C KO viruses.

(A) The protein expression of EBNA-3A, -3C, -2, -LP and LMP-1 was analyzed 10

days after B cell infection with EBV by western blotting. EBV-negative BJAB and EBV

positive EF-3D LCL were used as controls. (B) (Top) Dot plot analysis of EBV-driven B

cell proliferation is depicted using the CD19 B-cell marker (y-axis) and CellTrace Violet

dye (x-axis). Both EBNA-3A and 3C KO viruses drive B cell proliferation at 7 days post

infection. (Bottom) Similar dot plots of CD19/Violet indicate that EBNA-3A and EBNA-

3C KO viruses are impaired in driving B cell proliferation through 14 days after infection

compare to WT virus. (C) Quantification of proliferating CD19+ B cells, infected with

WT, EBNA-3A KO and EBNA-3C KO EBV at day 7 and day 14 post infection. Average

for two normal donors is shown +/- SEM.

57

2.1.4. Discussion

It has long been recognized that Epstein-Barr virus transformation efficiency is

on the order of 1-10% of infected primary human B cells (Henderson et al., 1977; Sugden

and Mark, 1977). However, little is known about the molecular mechanism responsible

for this low efficiency. We hypothesize that a robust innate tumor suppressor response

is activated by latent viral oncoproteins and blocks outgrowth of the majority of infected

cells. Recent evidence suggests that activated oncogene expression is sufficient to trigger

a growth-suppressive DNA damage responsive signaling pathway (Halazonetis et al.,

2008) and other oncogenic viruses have been shown to induce the DDR through

multiple means (reviewed in Chapter 1.4 and (Nikitin and Luftig, 2012). Therefore, in

this study we asked whether EBV was capable of inducing a DNA damage response in

primary B cells and, importantly, whether this response resulted in the low

transformation efficiency. We observed that as EBV-infected cells initiated proliferation,

a transient DNA damage response (DDR) was activated as evidenced by

phosphorylation of ATM Ser1981, H2AX Ser139 ( -H2AX), Chk2 Thr68, and

accumulation of 53BP1 in nuclear foci. Modulation of this signaling pathway by

chemical antagonism of ATM and its downstream target Chk2 markedly increased EBV-

mediated B cell polyclonal expansion and transformation efficiency thereby

demonstrating that the DDR contributes to an EBV-induced innate tumor suppressor

58

pathway. This is the first molecular pathway to be identified that restricts EBV

transformation.

2.2. Mitogen-Induced B Cell Proliferation Activates Chk2-dependent G1/S Cell Cycle Arrest

2.2.1. Contributions

M.L. and P.N. designed experiments and P.N. performed them. C.Y. made an

initial observation of elevated marks of the DDR in CpG-induced B cells. P.N. wrote the

chapter.

2.2.2. Introduction

In the first part of Results, I reported the fast division of Epstein-Barr virus-

infected human B cells, termed hyperproliferation, activating ATM and its downstream

kinase Chk2-dependent signaling. However, whether non-viral activators of B cell

hyper-proliferation activate the DDR remains unknown. Further, the downstream

effectors that mediated the DDR signaling in B lymphocytes are yet to be investigated.

Therefore, in the second part of Results I will discuss the role of the DDR in regulating

mitogen-induced B cell proliferation and the mechanism of Chk2-dependent

suppression of proliferation.

B cell mitogens. Two established ways to drive the proliferation of primary

human B cells in vitro include stimulation of the TLR9 receptor or activation with CD40

ligand coupled with cytokines (Hawkins et al., 2007a; Rousset et al., 1991; Tangye et al.,

2003). The human B cell TLR9 receptor may be potently and selectively activated with a

59

single-stranded oligonucleotide enriched for GC content that mimics the bacterial DNA

(termed “CpG”) (Cunningham-Rundles et al., 2006; Hartmann and Krieg, 2000). CpG

induces T cell-independent activation and accelerates proliferation of B cells that

recognize common pathogen patterns, such as bacterial DNA (Cunningham-Rundles et

al., 2006; Hartmann and Krieg, 2000) the (Fagarasan and Honjo, 2000)to . CpG DNA

binding activates the TLR9 dimer, which results in the recruitment of MyD88 (Hacker et

al., 2000; Latz et al., 2007). MyD88 interacts with IRAK4 and TRAF6 to induce mitogen-

activated protein kinase (MAPK) p38 and NF B transcription factors p50/p65, activator

protein (AP)-1, and activating transcription factor (ATF)-2 (Hartmann and Krieg, 2000;

Peng, 2005; Yi et al., 1998). In addition, MyD88 recruitment initiates a Pyk2-Src-Syk-

STAT3 downstream cascade that is required for cellular proliferation in a DOCK8-

dependent manner (Jabara et al., 2012). TLR9 stimulation of B cells in vitro potently

initiates proliferation of human and murine B cells. The kinetics of CpG-induced

proliferation of B cells has beenbeen well-studied by Phillip Hodgkin’s group (Hawkins

et al., 2007a; Turner et al., 2008). Intriguingly, computational analysis predicted two

distinct factors to control B-cell proliferation in early and later division, with the latter

being programmed cell death while the former remains unknown (Markham et al.,

2010).

60

Signaling through CD40 coupled with IL4 mimics the Th2-cell survival signal

and depends on TRAF6 (Lomaga et al., 1999). Notably, αCD40+IL4-induced

proliferation does not depend on DOCK8 (Jabara et al., 2012).

2.2.3. Results

2.2.3.1. Mitogen stimulation of primary B cells results in a period of robust

proliferation

We first determined the proliferative kinetics of human CD19+ B cells from three

normal donors in response to diverse stimuli. B cells within the context of PBMCs were

induced with two known human B-cell mitogens: the TLR9 ligand CpG (ODN 2006)

(Hartmann and Krieg, 2000) and the G28-5 agonistic anti-CD40 antibody combined with

recombinant human IL-4 (Clark and Ledbetter, 1986; Rousset et al., 1991; Tangye et al.,

2002). As a control, we infected PBMC with the B -cell- targeting virus EBV. Using the

fluorescent proliferation-tracking dye CellTrace Violet, we measured the proliferation

profile over time by FACS and calculated mean division number (MDNs) for each time

point (Fig. 15A-C) as described previously (Hawkins et al., 2007a; Nikitin et al., 2010).

Figure 15: Multiple B cell mitogens induce hyper-proliferation.

61

Kinetics of B cell proliferation in response to Epstein-Barr virus infection (A), constant

stimulation with TLR9 ligand CpG (2.5µg/mL) (B), and anti-CD40 antibody G28-5

(1µg/mL) coupled with interleukin-4 (20 pg/mL)treatment (C). Mean division number is

calculated as in Fig. 5 for PBMC from 3-6 normal human donors.

In EBV-infected cells we observed a bi-phasic proliferation of EBV-infected

PBMC, including a faster hyper-proliferative period followed by a slower stage of

proliferation (Nikitin et al., 2010) (Fig.15A). In CpG and αCD40/IL4-induced cells we

observed a similar initial period of robust proliferation rate as cells began to divide,

although the time to first division differed among these mitogens (Fig. 15B,C). Notably,

αCD40/IL4 stimulation induced a slower proliferation rate compared to EBV and CpG

(Fig. 15C).

2.2.3.2. Mitogen stimulation activates the ATM signaling pathway in hyper-

proliferating cells

Aberrant oncogene and growth factor induced cellular proliferation has been

shown to activate DNA replicative stress and a subsequent growth-suppressive DNA

damage responsive signaling pathway (Bartkova et al., 2006; Di Micco et al., 2006;

Halazonetis et al., 2008). We previously revealed activated hallmarks of the DDR,

including formation of γH2AX foci and ATM-downstream signaling in EBV-driven

proliferation (Fig. 4). Moreover, the DDR activation did not depend on viral DNA

replication, but rather correlated with the speed of cellular proliferation and was present

in early proliferating cells (Nikitin et al., 2010). In order to separate mitogen-induced

cycling (“proliferating”) and quiescent (“non-proliferating”) cells, we employed FACS

62

sorting. We observed elevated levels of γH2AX in proliferating B cells in all three

conditions (Fig. 16A-B).

Figure 16: B cell mitogens induce the ATM signaling pathways in hyper-

proliferating cells.

(A) Immunofluorescent microscopy of sorted early proliferating B cells. Non Prolif, non-

proliferating sorted B cells; Prolif, proliferating sorted B cells. DAPI, nuclei stain; H2AX,

phosphorylated histone H2AX; B cells, untreated primary B cells; IR, γ-irradiation. (B)

Number of cells with γH2AX intensity >5X over background plotted as a ratio from total

from three normal donors. Erorr bars are SEM. (C) Western blot analysis of Chk2 and its

ATM-phosphorylation site at Thr68. Protein lysates normalized to a total protein

concentration. P, proliferating; NP, non-proliferating cells; Cntr, untreated cells of

Burkitt’s lymphoma cell line 2 (BL2); IR, 5Gy irradiated BL2. Shown blot is a

representative analysis from three normal donors.

Consistent with previous work, ATM-dependent phosphorylation of Chk2 at

threonine-68 increased, suggesting the role of ATM/Chk2 kinases in the mediation of the

63

signal (Fig. 16C). Finally, we observed higher phosphorylation of pChk2-Thr68 in sorted

proliferating EBV-infected or CpG-induced cells, compared to αCD40/IL4-driven

proliferating cells.

2.2.3.3. EBV and mitogen-induced B-cell proliferation is suppressed by Chk2

Previously reported work has identified a role for ATM and its downstream effector

Chk2 in suppressing lymphomagenesis (Hirao et al., 2002; Maclean et al., 2008;

McPherson et al., 2004; Nikitin et al., 2010; Tort et al., 2002). Given our previous findings

indicating that EBV-induced B -cell proliferation was suppressed by Chk2 (Fig. 17A,

left), we assessed the role of Chk2 in suppressing mitogen-stimulated B-cell growth.

Consistently, treatment with a potent and specific small molecule inhibitor of Chk2

(Arienti et al., 2005) increased the number of dividing human B lymphocytes stimulated

with either CpG or CD40/IL4 (Fig. 17).

64

Figure 17: Chk2 inhibition increase proliferation of human B cells.

(A) FACS profile of EBV infected or mitogen-induced CD19+ cells at day 6 (EBV, CpG)

or day 7 (αCD40/IL4) post infection/stimulation in the presence of DMSO (red) or 5 M

Chk2i (blue). Gated CD19+ proliferating cells are shown in inset. (B) Relative number of

proliferating cells in the first three divisions. Cells from A. that divided up to 3 times

were gated and counted over internal beads control and normalized to DMSO-treated

sample. Experiment was repeated on PBMC from 3-6 normal donors. Error bars are

SEM.

Under all stimulating conditions, we did not observe a change in the time to first

division; rather inhibition of Chk2 increased the number of cells entering subsequent

divisions.

65

2.2.3.4. B-cell mitogens induce caspase 3/7-dependent apoptosis independent of Chk2

Chk2 downstream signaling induces apoptosis and cell cycle arrest (Sato et al.,

2010). Therefore, we assayed the role of Chk2 kinase in both of these processes during B-

cell mitogen driven hyper-proliferation. Both CpG and αCD40/IL4 stimulation of B cells

induced apoptosis, revealed by increased signal from a fluorescently labeled caspase 3/7

substrate DEVD-FAM (Fig. 18A, B) and by Western analysis of cleaved PARP and

caspase 3 (Fig. 18C). However, inhibition of Chk2 did not prevent the activated

apoptosis in proliferating cells upon CpG and αCD40/IL4 stimulations (compare Fig.18A

top to bottom panels). Surprisingly, we did not observe apoptotic cells in EBV-infected

early proliferating B cells (Fig.18. A-C). Unexpectedly, a modest induction of apoptosis

was observed in EBV-infected non-proliferating cells (Violethi) but not in proliferating

cells (Violetlo).

66

Figure 18: B-cell mitogens, but not EBV induces caspases 3/7-dependent apoptosis.

(A) Activated apoptosis in proliferating B cells treated with DMSO (top panels) or Chk2i

(bottom panels) and measured with DEVD-FAM, a fluorescently labeled soluble caspase

3/7 inhibitor. FACS profiles of CD19+ single cells. (B) Percent of apoptotic cells in non-

proliferating (NP) or proliferating (Prol) CD19+ cells treated with DMSO (blue) or Chk2i

(red). Activation of caspases was compared to LCL and BL2 cells treated with 10µM

Nutlin-3 or 15µM camptothecin. (C) Cleavage of caspase targets (PARP and self-

processing of caspase-3) revealed by western blot analysis of sorted proliferating (Prol)

or non-proliferating (NP) EBV-infected or mitogen-stimulated B cells. Casp3, cleaved

form of Caspase 3.

Therefore, Chk2 inhibition does not relieve activation of caspases 3/7 in early

proliferating B cells.

2.2.3.6. Mitogen-induced hyper-proliferation of human B cells induced a Chk2-

dependent G1/S cell cycle arrest

As discussed in the introduction, Chk2 phosphorylation stabilizes p53, thus

promoting G1/S phase arrest, and activates CDC25s to induce an intra-S phase and

G2/M checkpoints (Falck et al., 2001; Hirao et al., 2000; Matsuoka et al., 1998). We have

67

assayed the cell cycle profile of early proliferating EBV-infected or mitogen-stimulated

cells following a 2 hours pulse of the thymidine analog BrdU. We revealed that Chk2-

inhibition increased the number of cells entering S-phase in CpG-induced or EBV-

infected proliferating cells. Further, αCD40/IL4-stimulation led to a high number of cells

entering the first S-phase that was mildly increased upon Chk2i treatment (Fig. 19A,

left). Finally, in all three stimuli, we did not observe a significant accumulation of cells in

G2/M stage or a release of BrdU-negative cells (intra-S phase checkpoint) upon

inhibition of Chk2. Strikingly, EBV-transformed lymphoblastoid cell lines with

attenuated expression of proliferation-driven genes (Nikitin et al., 2010) were not

responsive to inhibition of Chk2 and had a normal cell cycle profile. Therefore, we

conclude that Chk2 induced a G1/S checkpoint in human B-cells.

68

Figure 19: Mitogen stimulation or EBV infection of human B cells activates

Chk2-dependent cell cycle arrest.

(A) Cell cycle profile of EBV-infected or mitogen-induced proliferating B cells treated

with DMSO (red) or Chk2i (blue) measured by FACS. Cells were pulsed with 25µM

BrdU for 2 hours and subsequently stained with anti-BrdU antibody and propidium

iodide (PI). Cells in the first division were gated as for previous figure. G1, S, G2-M,

stages of the cell cycle. (B) Number of cells (from A) in S-phase normalized to DMSO-

treated samples from three normal donors. Error bars are SEM. (C) p21 mRNA level in

sorted EBV-infected early proliferating cells treated with DMSO (red) or Chk2i (blue),

EBV-immortalized LCL (black) or LCL induced with 10uM Nutlin-3 (stripe pattern) and

normalized to an SETDB1. Shown value is an average for two normal donors. Error bars

are SEM.

69

2.2.3.7. Activated Chk2 induces expression of the CDK inhibitor p21

As we have observed a Chk2-dependent G1/S phase arrest in EBV-infected or

mitogen-induced normal human B-cells, we next assayed the expression of p53-

dependent effector p21 likely to play a role in limiting the proliferation of human B cells.

We assayed p21 mRNA levels in early proliferating cells treated with DMSO or Chk2i by

qPCR. We found that p21 expression was decreased in Chk2i treated EBV-infected early

proliferating cells by 2-5 folds in two donors (Fig. 19C) to the level of LCL. As a positive

control we used previously reported Nutlin-3 treatment of LCL, that upregulates the p21

level (Forte and Luftig, 2009).

2.2.4. Discussion

We identified a Chk2-dependent G1/S phase cell cycle checkpoint as a limit to

mitogen-induced proliferation of human B-cells. The activated checkpoint was

uncoupled from apoptosis in mitogen-induced or EBV-infected B-cells. Notably, the

pharmacological inhibition of Chk2 increased the number of dividing cells, but did not

change the time to first division nor did it affect the rate of cellular division. Instead,

Chk2 inhibition of EBV-infected early proliferating cells resulted in decreased expression

of G1/S checkpoint effector p21.

EBV infection activates the DDR-dependent G1/S cell cycle arrest and

immortalizes B cells with inactivated p16. Consistent with previously reported findings

(Nikitin et al., 2010), we observed DDR–dependent growth suppression of EBV-driven

70

proliferation of B cells. Surprisingly, we did not detect apoptosis in dividing cells; rather

we observed the DDR-dependent expression of p21 and induction of G1/S phase cell

cycle arrest. In addition, we detected the activation of the senescence-associated beta-

galactosidase in arrested, but not in proliferating cells (data not shown).Notably, we did

not detect an active apoptosis in infected cells (Fig. 18). Expression of viral BCL2

homologs likely prevents activation of apoptosis in EBV-infected cells ((Altmann and

Hammerschmidt, 2005) and Micah Luftig and Jay Tourigny, unpublished).

Further, the DDR-dependent G1/S arrest was readily detected in early

proliferating B cells but not in indefinitely transformed LCL, despite the latter retained

thea low level of γH2AX positivity (Nikitin et al., 2010). Recent reports have begun to

address how the initial DNA damage results in the formation of persistent

heterochromatin foci with active DDR signaling (Di Micco et al., 2011; Rodier et al.,

2011). Specifically, (Rodier et al., 2011) identified DNA-SCARS as foci with persistent

ATM-downstream signaling in dividing cells with inactivated p16. EBNA-3C attenuates

the DDR-mediated growth arrest (Nikitin et al., 2010) and induces epigenetic

inactivation of p16 in transformed LCL (Maruo et al., 2011; Skalska et al., 2010).

Therefore, we hypothesize that EBNA2-dependent transcription of host growth

promoting genes, such as c-Myc, initiates the oncogene-induced replicative stress and

activation of ATM/Chk2-dependent G1/S growth arrest of the majority of infected

proliferating cells. Further, EBNA2 driven EBNA3C attenuates the expression of

71

oncogenes and inactivates p16 allowing subsequent cellular proliferation despite the

presence of active ATM signaling.

DNA damage response in mitogen-induced B cells. Chk2-dependent G1/S cell

cycle arrest was detected in the first three cell divisions of CpG-induced or EBV-infected

cells; however αCD40/IL4-induced cells demonstrated the most apparent response to

Chk2 inhibition during the first division only. Considering the observed decline in the

proliferation speed of αCD40/IL4-induced cells (Fig. 15 and BrdU profiles not shown),

we conclude that Chk2 activates G1/S in hyper-proliferating cells in the first division

upon stimulation with this mitogen. Notably, we previously reported similar transient

activation of Chk2 in EBV-infected hyper-proliferating cells that decreased with the slow

proliferation rate (Nikitin et al., 2010).

TLR9 stimulation of B cells activates two independent suppressive

mechanisms. We have demonstrated a bi-phasic profile of human B cell proliferation in

response to TLR9 stimulation with CpG in vitro. Particularly, during the first three

divisions proliferating B cells were free of apoptotic markers and displayed Chk2-

dependent and reversible G1/S cell cycle arrest. However, divisions four and higher

displayed apoptotic markers, including activated Caspases 3 and 7 and cleaved PARP.

Consistent with our findings, CpG-induced dividing murine BCL2 transgenic B cells

remained alive while arrested in higher divisions (Hawkins et al., 2007b). Therefore,

conclude that the two factors regulating TLR9-induced proliferation of human B-cells in

72

vitro (Markham et al., 2010) are, first, a DDR-dependent G1/S cell cycle arrest and,

second, a DDR-independent proliferation associated apoptosis.

Implications in vivo. TLR9 ligand stimulation, but not αCD40/IL4 treatment,

requires DOCK8 (Jabara et al., 2012), a guanine nucleotide exchange factor that activates

Cdc42 and promotes cellular proliferation (Cote and Vuori, 2002; Harada et al., 2012). In

a recently proposed model Goodnow et al. suggest that DOCK8 upregulates MDM2

through the activation of Akt and therefore silence p53 activity in the germinal center B

cells (reviewed in (Goodnow et al., 2010)). However, DDR signaling activates ATM and

Chk2 which in turn stabilize p53 by direct phosphorylations on Ser 15 and 20 (Hirao et

al., 2000). Strikingly, the ATR/Chk1 shoulder of the DDR is potently down-regulated,

while the expression of pro-survival BCL6 is upregulated in germinal center B cells

(Ranuncolo et al., 2007). Therefore, degraded p53 and down-regulated ATR likely

protect hyper-proliferating B-cells from cell cycle arrest in germinal center, while G1/S

checkpoint may restrict an extra-follicular proliferation of B cells.

Overall, we have identified a Chk2-dependent growth arrest upon EBV-driven or

mitogen-induced proliferation of B-cells in the absence of T cells. Chk2 kinase inhibition

resulted in increased DNA synthesis, but did not affect the induction of apoptosis. We

therefore conclude that Chk2-dependent DDR may limit extrafollicular proliferation of

human B cells.

73

2.3. Enhanced Method of Epstein-Barr Virus Mediated Transformation of B Cells Used for Generation of Human Monoclonal Antibodies

2.3.1. Contributions

The following section of Results has been presented at CFAR retreat by P.N. and

included in patent application (Kwan-Ki Hwang, 2009). Micah Luftig, Barton Haynes

initiated and designed the project. P.N., M.L. and Kwan-Ki Hwang designed the

experimental approach, and P.N. and K.H. performed experiments.

2.3.2. Introduction

Broadly neutralizing monoclonal antibody (BnAB) therapy is a novel promising

method to control chronic infection and to prevent de novo infection with highly

pathogenic agents. The vastly growing pool of isolated human monoclonal antibodies

includes BnAbs to anthrax (Smith et al., 2009), chikungunya virus (Warter et al., 2011),

cytomegalovirus (Macagno et al., 2010), dengue virus, HIV (reviewed in (Haynes et al.,

2012)), influenza virus (Corti et al., 2010; Krause et al., 2012; Yu et al., 2008) (including

“avian” strain H5N1 (Hu et al., 2012; Simmons et al., 2007)), respiratory syncytial virus

(Collarini et al., 2009), rotavirus (Di Niro et al., 2010; Tian et al., 2008), SARS (Traggiai et

al., 2004) and tetanus (Poulsen et al., 2011) (summarized in (Wilson and Andrews,

2012)).

In the case of infectious agents with low immunogenicity of conserved epitopes,

such as HIV, influenza and HCV, inefficient vaccination fails to provide broad and long-

74

lasting protection (Haynes et al., 2012). For instance, the first limited neutralizing Ab

response to HIV gp120 appears by 16 weeks after transmission (Richman et al., 2003;

Wei et al., 2003). Moreover, only ~5% of chronically HIV-infected patients develop

broadly neutralizing antibodies (BnAB) through a presumably atypical maturation

process that results in BnAB with high affinity to host self-antigens and extensive

somatic hyper-mutation (Haynes et al., 2005; Haynes et al., 2012). Therefore, isolation

and investigation of patient-derived BnAbs not only has the potential to generate

therapeutics, but also to provide information for vaccine design.

Currently, the four most common methods to isolate human nAB, include: (1)

phage display, (2) in vitro proliferation and immortalization of memory B cells, (3)

single B cell expression cloning with antigen baiting and (4) plasmablasts expression

cloning after the exposure to an antigen. However, each method suits different needs

and has limitations. Particularly, selection through a phage display leads to bacterial

processing of resulted heavy or light chain; single B cell expression cloning requires a

known and immunogenic baiting antigen; while a single-cell expression cloning of

plasmablasts transiently and briefly detected after the antigen exposure (Wilson and

Andrews, 2012). Finally, the immortalization of memory B cells, usually with EBV, has

long been used as an option to immortalize antibody-secreting cells (Kozbor and Roder,

1981; Steinitz et al., 1977). However, as discussed in previous Chapters, the efficiency of

viral transformation remains below 1-10%. Introduction of B-cell mitogens, such as the

75

TLR9 ligand CpG, increased viral transformation (Iskra et al., 2010; Traggiai et al., 2004).

Particularly, this approach was used to isolate BnAB against SARS coronavirus, H5N1

“avian” influenza virus, and dengue virus from convalescent patients as well as recently

various BnAB to HIV from chronic HIV-infected individuals (Friberg et al., 2012; Hicar

et al., 2010; Hu et al., 2012; Simmons et al., 2007; Traggiai et al., 2004).

In the two previous chapters we identified the DDR as one of the major growth-

suppressive mechanisms to control EBV-driven or CpG-induced proliferation of human

B cells. Therefore, we reasoned that the inhibition of key kinases ATM and Chk2 would

facilitate the isolation of patient-derived human monoclonal antibodies following EBV

infection of memory B cells.

2.3.3. Results

2.3.3.1. Pharmacological inhibition of ATM or Chk2 increases EBV-mediated

transformation of low numbers of B cells

We first identified the minimal number of total primary B cells sufficient to

achieve long-term outgrowth in a single well of a 96-well plate following EBV infection.

To provide supportive growth signals we incubated EBV-infected B cells with irradiated

feeder cells, including autologous PBMC from the same donor or mouse macrophages

J774.A1. We plated EBV-infected total B cells at 1-1000 cells per well density. Five weeks

post infection we detected EBV-transformed clones in 10% of wells plated with 30

cells/well and in all wells plated with 1000 cells/well (Fig. 20, black circles).

76

Figure 20: Pharmacological inhibition of DDR kinases increases EBV

transformation of low numbers of total B cells derived from a normal donor.

Total B cells were infected with EBV and plated at low density in 96-well plate

with iiradiated autologous PBMC. Five weeks post infection wells with growing LCL

were counted as positive. Black dots, DMSO-treated; Blue square, Chk2i-treated; Red

triangle-ATMi treated cells.

We next assayed whether EBV-mediated transformation of a low number of B

cells changed upon small molecule antagonism of ATM or Chk2. As expected, inhibition

of the DDR kinases ATM or Chk2 (DDRi) increased the efficiency of EBV-mediated

transformation (Figure 20, red triangles and blue squares). Notably, DDRi decreased the

minimal cell number required for EBV transformation to ~5 cells / well (six fold) and the

minimal cell number to achieve the 100% transformation to ~100 cells / well (tenfold).

77

2.3.3.2. Inhibition of Chk2 coupled with TLR9 stimulation additively enhances EBV-

mediated transformation of memory B cells from normal donors

We aimed to improve the EBV-mediated immortalization of IgG-producing cells.

We next focused on memory B cells derived from a normal donor. Previously, TLR9

stimulation with CpG DNA was shown to significantly improve EBV-transformation

efficiency (Traggiai et al., 2004). Therefore, we assayed EBV-transformation efficiency in

the presence of CpG and Chk2 inhibitor. We sorted CD19hi CD27hi IgDlo memory B cells

from a normal donor (Fig. 21, left) and infected them with EBV.

Figure 21: Chk2 inhibitor and TLR ligand CpG additively increase EBV-

mediated transformation of human memory B cells.

Left. CD19 B cells were sorted from a marked gate by FACS. Right. Sorted

CD19+ CD27+IgD- memory B cells were EBV infected and treated with DMSO (black),

CpG (blue), Chk2i (brown) or CpG combined with Chk2 (red) and plated at 300

cells/well density in 96-well plates over irradiate mouse macrophages. Five weeks later

the percent of positive wells was determined as in previous figure.

78

Both CpG and Chk2i treatment increased the EBV transformation of memory B

cells plated at 300 cells per well. EBV-infection or TLR9 stimulation of B cells activates

cell cycle arrest (Chapter. 2.2); however, whether the combined stimulation may

overcome cell cycle arrest remained unknown. We found that the combined treatment

additively elevated EBV transformation efficiency from 70% (EBV+CpG) to 90%

(EBV+CpG+Chk2i) (Fig. 21, right panel). We thus referred to CpG and Chk2i dual

treatment as an enhanced EBV transformation method.

2.3.3.3. Combined treatment with Chk2i and TLR9 ligand increases EBV-mediated

transformation of memory B cells from a chronic HIV-infected patient

As mentioned above, the efficient selection and immortalization of BnAB-

producing memory B cells will facilitate the production of Ig-based therapeutics and

inform vaccine design. In order to identify such BnAB it is important to identify patients

that have recovered or chronically infected with a pathogen. Therefore, we tested our

method of enhanced EBV transformation on B cells derived from a HIV chronically -

infected patient in collaboration with Bart Haynes group at the Duke Human Vaccine

Institute

Thawed PBMC from the chronic HIV patient were FACS sorted as shown above

and infected with EBV in the presence of TLR9 ligand CpG. Five weeks after EBV

infection we observed transformation in ~20% of wells plated with 3000 PBMC and in

~25% of wells plated with 100 memory B cells (Fig. 22, blue bars).

79

Figure 22: Enhanced EBV transformation of memory B cells derived from a

chronic HIV patient.

Frozen PBMC sample from CHAVI’s chronic HIV patient was thawed, FACS

sorted as for previous figure and EBV infected. Unsorted PBMC and sorted memory B

cells were treated with CpG (blue bars) or CpG combined with Chk2i (red bars) and

plated at 3000 cells per well and 100 cells per well density over irradiated macrophages

in 96-well plate. Transformation was determined as in previous figures. c/w, cells per

well.

Strikingly, upon inhibition of Chk2 EBV transformation efficiency increased to

~60% for PBMC (three fold over CpG alone) and to ~55% (two fold over CpG alone) for

Memory B cells.

80

2.3.4. Discussion

We have identified the DNA damage response as a growth suppressive

mechanism to restrict EBV-mediated transformation of low numbers of total B and

memory B cells. Further, we found that EBV transformation during pharmacological

inhibition of the DDR can be performed with as low as 3 B cells per well. Finally, in a

proof-of-concept experiment we demonstrated that TLR9 stimulation coupled with EBV

infection and inhibition of Chk2 kinase increased transformation of memory B cells

derived from a chronic HIV patient.

The results of our work were included in a patent application filed in 2009

(Kwan-Ki Hwang, 2009). Since that time, the inhibition of the DDR kinase Chk2 (Nikitin

et al., 2010), along with TLR9 stimulation (Traggiai et al., 2004), became a common way

to increase EBV mediated transformation of patient-derived memory B cells.

Particularly, this combined treatment was used to isolate HIV-1 envelope-specific BnAB

and study the V2/V3 conformational epitope presentation mechanism (Bonsignori et al.,

2011), to select BnAB specific to a conserved epitope on influenza H1N1 hemagglutinin

(Krause et al., 2011), to isolate weak nAB and study the antibody-mediated enhancement

of secondary dengue virus infection (Smith et al., 2012) and identify early BnAB to HIV-

1 that induce viral immune escape (Bar et al., 2012).

81

3. Conclusions and future directions

We identified the first tumor suppressive pathway to limit EBV immortalization

of primary human B cells. Particularly, we revealed that a transient, viral-driven

elevated expression of growth control genes induces the cellular hyper-proliferation and

activates the growth suppressive ATM/Chk2-dependent DDR resulting in G1/S cell cycle

arrest in most of early proliferating B cells. However, viral protein EBNA3C down-

regulates the host DDR and promotes immortalization of infected cells into

lymphoblasts.

Our work also provides a new paradigm for EBV infection of primary B cells de

novo, elucidating the intermediate period between resting and transformed cells

happened early in infection. Early proliferating cells display an atypically high level of

EBNA-LP expression, robust proliferation rate and activated hallmarks of the DDR

resulting in p21-mediated G1/S cell cycle arrest.

However, there are several questions remained. We found the activated state of

DDR signaling pathway, but we did not detect DNA damage in sorted proliferating cells

by comet assay (not shown). Therefore, whether initial hyper-proliferation leads to a

replication stress and double-stranded breaks, exposed telomeres or induces a formation

of heterochromatin foci remains unknown. Next question is whether transformed

lymphoblastic cells still contain such heterochromatin foci or any traces of DNA

damage. The rationale for this question is the presence of low, but detectable γH2AX

82

signal in LCL. Further, what is the significance of early proliferating phase in vivo?

Replicative stress and high Myc level may facilitate the mutagenesis and selection for

clones with constantly high Myc expression. This process coupled with negative

selection for EBNA3s due to a high immunogenicity would favor the Myc translocation

under Ig promoter and drives early proliferating cells towards latency I Burkitt’s

lymphoma. Finally, we demonstrated an asynchronous proliferation of infected cells.

Does it reflect heterogeneity of latency proteins expression in different cells? If so, which

cells have higher chance to be transformed into latency III in vitro, and which cells

would give raise to latency I lymphomas in vivo?

3.1. The source of DNA damage

The first question is what activates the DNA damage response. Towards

characterizing the EBV-induced DNA damage signal, we reasoned that either viral or

cellular DNA was important for ATM activation. In addition to oncogenic stress,

replication intermediates of DNA viruses and retroviruses contain double-stranded

DNA ends that activate ATM (Lilley et al., 2007). In fact, EBV lytic replication induces a

DDR which is suppressed by inhibition of downstream transcriptional activation of p53

(Kudoh et al., 2005; Mauser et al., 2002). In our studies of primary B cell infection,

however, we found no evidence of viral lytic DNA replication or viral DNA associated

with DDR activation. First, we did not observe DDR activation within the first 3 days

after EBV infection nor did we observe activation using UV-inactivated virus or the non-

83

transforming EBV variant P3HR1 suggesting that the incoming linear DNA genome and

tegument proteins within the virion were not responsible for this signal. Second, lytic

viral DNA was not responsible for DDR activation as less than 1% of infected cells were

undergoing lytic DNA replication when greater than 50% of infected cells were -H2AX

positive. Third, we asked whether the DNA damage signal was derived from viral

episomes since DNA repair factors are recruited to the episome to ensure proper

resolution of Holliday junctions following episome replication (Deng et al., 2002;

Dheekollu et al., 2007). We observed little increase in episome number per cell and

found that viral episomes and -H2AX did not co-localize during the period of DDR

activation. These data collectively demonstrate that viral DNA is not the source of DNA

damage. Our experiments cannot rule out the possibility, however, that viral lytic gene

expression downstream of BZLF1 in the absence of lytic DNA replication (Kalla et al.,

2010) plays a role in the transient DDR early after infection. Despite this possibility, we

inferred from our data that viral latent gene expression causes an oncogenic stress

response leading to cellular DNA damage.

The initiation of cell proliferation defines the period after EBV infection when

ATM and Chk2 were active in suppressing transformation. Rigorous analysis of infected

cell division rates uncovered a period of hyper-proliferation where early population

doublings (PDs) were every 8-12h leading to DDR activation, while later divisions

displayed an attenuated rate of ~24-30h per division similar to LCLs and had little

84

evidence of DDR activation. Microarray analysis of gene expression during the

transition from resting B cell to early EBV-induced hyper-proliferation and through LCL

outgrowth strongly supported our cell-based observations. Specifically, genes involved

in proliferation and the DDR, including ATM/p53-dependent targets (Elkon et al., 2005),

were highly induced early after infection and then attenuated during the transition to

LCL. We propose that aberrant induction of cellular DNA replication early after EBV

infection activates a DNA damage response that is dependent on EBNA2 and EBNA-LP

mediated up-regulation of S phase promoting oncoproteins including c-Myc, cyclin D2,

and E2F1 ((Kaiser et al., 1999; Sinclair et al., 1994) and Fig. 11). Indeed, we observed

increased expression of c-Myc and its gene activation signature in hyper-proliferating

cells relative to LCLs. Furthermore, EBNA-LP protein levels and Wp-derived transcripts

were heightened during this early period relative to EBNA3 proteins and Cp transcripts

consistent with previous analysis of the initial cascade of viral latent gene expression at

different days post infection (Schlager et al., 1996; Woisetschlaeger et al., 1989;

Woisetschlaeger et al., 1990). Finally, EBNA3C, but not EBNA3A, deleted virus-infected

cells displayed a significantly stronger DDR during early proliferation. Thus, while both

EBNA3A and EBNA3C likely mitigate growth arrest in LCLs through p16 suppression

(Hertle et al., 2009; Skalska et al., 2010), during early outgrowth EBNA3C is also

required to modulate the DNA damage response. Collectively, our data support a model

that initial EBV-driven hyper-proliferation leads to an oncogenic stress that is ultimately

85

attenuated as EBNA3 proteins moderate EBNA2 driven c-Myc and its genotoxic and

growth suppressive consequences. This ultimate balance in viral and host gene

expression enables constitutive S phase induction without driving selection of cells with

genomic instability.

Furthermore, in our system, we did not observe increased transformation in the

presence of anti-oxidants including N-acetyl cysteine and citric acid (P.N. and M.L. data

not shown) suggesting that ROS was not responsible for the EBV-induced DDR. We also

did not observe changes in BubR1 or ATM expression through LCL outgrowth (data not

shown). However, we anticipate that genomic instability may ensue in the setting of

aberrant latent oncoprotein expression that may exist in BL and other EBV-associated

tumors. Consistent with this notion and our findings, a recent report suggests that while

LCLs maintain a stable karyotype, early DNA damaging events may lead to non-clonal

chromosomal aberrations including telomere fusions (Lacoste et al., 2009). This report

supports our findings of an early hyper-proliferation associated oncogenic stress that

may induce such structures leading to ATM activation (Karlseder et al., 1999) and

suppression of long-term outgrowth. Thus, only cells with the ability to maintain a

stable karyotype emerge as LCLs.

3.2. DDR signaling in transformed cells

Second question is why EBV-transformed LCL display a residual level of γH2AX

positivity. EBV infection of immortalized lymphoma cells or modulation of expression

86

of viral latent genes in LCL revealed different aspects of viral interaction with the DDR

(summarized in Chapter 1.4). However, we did not observe overt genomic aberrations in

LCL that constitutively express nine viral latent proteins at the physiological level.

Furthermore, while LCL displayed low levels of DDR hallmarks such as γH2AX, we did

not detect the downstream consequences of the DDR activation, such as cell cycle arrest

or apoptosis (Fig. 18 and 19). In contrast, Burkitt’s lymphoma-derived cell line BL2 that

has a wild type p53 readily displayed a Chk2-dependent G1/S cell cycle arrest (Fig. 19).

Observed residual level of γH2AX activation in LCL may reflect the persistent

chromatin changes formed upon DNA damage (Di Micco et al., 2011; Rodier et al., 2011).

Further, DDR-dependent heterochromatin foci display ATM-downstream signaling and

growth arrest in p16-intact cells, but continue to proliferate in the absence of p16 (Rodier

et al., 2011). Intriguingly, in EBV-transformed LCL viral protein EBNA3C epigenetically

represses p16 (Maruo et al., 2011; Skalska et al., 2010). Therefore, we suggest that EBV-

driven cellular hyper-proliferation causes DNA damage and induces formation of

persistent heterochromatin foci with localized ATM signaling. However, EBNA3C

affects the cause of DDR and mediates cell cycle checkpoint through attenuation of the

cellular proliferation and repression of p16.

3.3. Implication in vivo

The third question is how early proliferating cells progress in vivo? Our findings

of an intermediate state early in infection has implications to the germinal center model

87

for EBV infection (Roughan and Thorley-Lawson, 2009) in the context of B cell

lymphomagenesis. In particular, our observed hyper-proliferative phase early after

infection in vitro may be similarly induced by EBV in vivo and is reminiscent of B cell

proliferation rates in the germinal center (MacLennan, 1994). However, there are

important differences between B cell proliferation in germinal center and due to EBV

infection. First, Bcl-6 down-regulation of the DDR mitigates the consequences of

centroblast hyper-proliferation in the germinal center (Ranuncolo et al., 2007), while

EBV potently suppresses Bcl-6 early after infection leaving DDR checkpoints intact

(Siemer et al., 2008). Second, high MDM2 expression in GC cells silences p53 (Goodnow

et al., 2010), while viral protein EBNA3C attenuates DDR later in infection that prevents

p53 activation (Nikitin et al., 2010). Therefore, EBV-infected B cells may not only migrate

into GCs as suggested previously (Thorley-Lawson and Allday, 2008), but may

additionally promote extra-follicular B cell maturation by-passing the germinal center

(Heath et al., 2012). However, a critical balance must be struck between the aberrant

latent oncoprotein-driven proliferation early after infection and the stable proliferative

signals found in LCLs to maintain an activated, immortalized state. Perturbations in this

balance in vivo may select for mutations driving lymphomagenesis. For example, the

IgH/c-myc translocation common in Burkitt’s lymphoma (BL) may be the consequence of

high EBNA-LP expression in the absence of EBNA3C. Given our findings, it is plausible

that imbalances in EBV latent gene expression may provide a milieu of cells with an

88

increased potential for genomic instability. Recent work in BL cell lines suggests that this

is likely the case.

3.4. Expression of viral latency proteins in early proliferating B cell

Finally, we investigated a bulk population of proliferating cells, whether arrested

or continued to proliferate. However, whether EBV latency proteins are expressed at

different levels in different cells is poorly understood. While we demonstrated dynamic

changes in EBNA-LP to EBNA3s expression ratio, we have not investigated the

abundance of these viral proteins in cells that either entered a cell cycle and arrested or

continued to proliferate. Therefore, the key question to be addressed is whether EBV

infection generates distinct subsets of cells with different viral gene expression and thus, potency

to induce oncogenesis.

3.4.1. How is EBNA3C activity regulated in infected cells?

We and others demonstrated the role of EBNA3C in deactivating host tumor

suppression. Particularly, EBNA3C downregulates the DDR (Nikitin et al., 2010) and

epigenetically inactivates p16 (Maruo et al., 2011; Skalska et al., 2010). However, the

expression pattern of EBNA3C per single cell remains unknown. Western blot analysis

detected two isoforms of EBNA3C in early proliferating cells, but not in LCL (Nikitin et

al., 2010). Further, our group revealed host genome-wide alterations in alternative exon

isoform usage in infected cells ((Robinson et al., 2012) and Nicholas Homa and M.L. in

preparation). The genome-wide alternations in initiation and splicing found in EBV

89

transformed cells suggest fundamental changes in mRNA processing machinery

affecting both host and viral gene expression early and late in B cell infection. Therefore,

the functional activity of EBNA3C is likely attributed to a particular isoform expressed

in a given cell, which controls the survival of this cell.

3.4.2. What is the role of LMP1 in restricting the DDR and why is its expression is delayed?

LMP1 expression is delayed in infected B cells in vitro (Price et al., 2012). Notably,

LMP1 expression during latency III is controlled by its own promoter and depends on

EBNA2 and EBNA3C activity (discussed in introduction). Therefore, expression of

LMP1 likely depends on progression through cell divisions and occurs in cells that have

already escaped the DDR-imposed growth arrest, i.e. in cells expressing an active

EBNA3C with attenuated c-Myc level and inactivated p16. LMP1 exclusive expression in

cells with active EBNA3C may particularly explain the stable karyotype in LCL, as only

cells experienced low oncogenic stress progress far enough to receive LMP1-mediated

survival stimuli. Finally, such division-linked regulation may prevent the competition

between DDR-induced p53 and LMP1-driven NF-kB downstream signaling (reviewed

by (Perkins, 2012)) and provide the required survival stimulus to cycling cells.

3.4.3. How EBNA-LP length and expression control EBV-driven oncogenesis?

EBNA-LP co-operates in transcriptional activation of EBNA2 target genes

(Rickinson and Kieff, 2006). We detected a transient up-regulation of EBNA-LP

90

expression, EBNA2 activity and c-Myc mRNA levels in early proliferating infected cells.

Furthermore, and consistent with previous work, we detected up to nine EBNA-LP

isoforms due to the initiation from or splicing through a variable number of W repeats

used early in infection, followed by selection of 1-2 isoforms in transformed cells.

Intriguingly, the number of W repeats found in vivo determines EBV transformation

efficiency (Tierney et al., 2011). Therefore, it is highly possible that newly infected B cells

in vivo express EBNA-LP that contain different combinations of repeats, and therefore

display variable levels of EBNA2 activity. Further, the heterogeneity EBNA-LP/EBNA2

activity results in subsets of cells expressing different levels of growth control genes,

such as c-Myc, and therefore experiencing different extend of oncogenic stress. Finally,

subsets of cells with different levels of expressed oncogenes are likely a basis for

mutagenesis, such as c-Myc translocation under Ig promoter, found in Burkitt’s

lymphomas.

3.5. A proposed model of EBV infection of primary B cells

In summary, I propose that EBV infection results in subsets of B cells expressing

atypical combinations of latent proteins and their isoforms. Therefore, instead of the

linear sequence of events upon infection, schematically written as:

EBNA-LPEBNA2 activityEBNA3CLMP1 transformation

viral infection results in early proliferating cells expressing combinations of proteins :

(EBNA-LPW1-9 and EBNA2 and/or EBNA3Con/off)LMP1early/late

91

Further, considering the tumor suppressive role of EBNA3B (White et al., 2012), the

subset of early proliferating EBV infected cells is likely described by the following

schematic:

[(EBNA-LPW1-9 and EBNA2 and/or (EBNA3Con/off + EBNA3Bon/off)]LMP1early/late.

In addition, each infected cell contains EBNA1 and may contain EBNA3A and LMP2s.

Finally, EBV infected early proliferated cells may express different subsets of viral non-

coding RNAs (not discussed in this dissertation, but emerge as important regulators of

viral latency (Skalsky et al., 2012)).

Hence, viral infection results in subsets of cells with various levels of EBNA-LP

that determines the oncogenic potential of this cell, EBNA3C that promotes

transformation but induces the strong cytotoxic immune response and LMP-1 that likely

is expressed later in infection and provides the required survival stimuli. Each latency

variant expressed in early proliferating B cells may be beneficial in different conditions,

such as immunosuppression, environmental stress or additional pathogens infection.

Overall, EBV-infected early proliferating B cells likely consist of multiple

populations expressing different levels of latency components and provide a broad

range of variants for in vivo selection by the host immune response and cell-intrinsic

growth suppression.

92

References

Alfieri, C., Birkenbach, M., and Kieff, E. (1991). Early events in Epstein-Barr virus

infection of human B lymphocytes. Virology 181, 595-608.

Altmann, M., and Hammerschmidt, W. (2005). Epstein-Barr virus provides a new

paradigm: a requirement for the immediate inhibition of apoptosis. PLoS biology 3,

e404.

Anderton, E., Yee, J., Smith, P., Crook, T., White, R.E., and Allday, M.J. (2008). Two

Epstein-Barr virus (EBV) oncoproteins cooperate to repress expression of the

proapoptotic tumour-suppressor Bim: clues to the pathogenesis of Burkitt's lymphoma.

Oncogene 27, 421-433.

Arienti, K.L., Brunmark, A., Axe, F.U., McClure, K., Lee, A., Blevitt, J., Neff, D.K.,

Huang, L., Crawford, S., Pandit, C.R., et al. (2005). Checkpoint kinase inhibitors: SAR

and radioprotective properties of a series of 2-arylbenzimidazoles. J Med Chem 48, 1873-

1885.

Ariumi, Y., Kaida, A., Lin, J.Y., Hirota, M., Masui, O., Yamaoka, S., Taya, Y., and

Shimotohno, K. (2000). HTLV-1 tax oncoprotein represses the p53-mediated trans-

activation function through coactivator CBP sequestration. Oncogene 19, 1491-1499.

Arrand, J.R., and Rymo, L. (1982). Characterization of the major Epstein-Barr virus-

specific RNA in Burkitt lymphoma-derived cells. Journal of virology 41, 376-389.

Babcock, G.J., Decker, L.L., Volk, M., and Thorley-Lawson, D.A. (1998). EBV persistence

in memory B cells in vivo. Immunity 9, 395-404.

Bar, K.J., Tsao, C.Y., Iyer, S.S., Decker, J.M., Yang, Y., Bonsignori, M., Chen, X., Hwang,

K.K., Montefiori, D.C., Liao, H.X., et al. (2012). Early low-titer neutralizing antibodies

impede HIV-1 replication and select for virus escape. PLoS Pathog 8, e1002721.

93

Bartkova, J., Horejsi, Z., Koed, K., Kramer, A., Tort, F., Zieger, K., Guldberg, P., Sehested,

M., Nesland, J.M., Lukas, C., et al. (2005). DNA damage response as a candidate anti-

cancer barrier in early human tumorigenesis. Nature 434, 864-870.

Bartkova, J., Rezaei, N., Liontos, M., Karakaidos, P., Kletsas, D., Issaeva, N., Vassiliou,

L.V., Kolettas, E., Niforou, K., Zoumpourlis, V.C., et al. (2006). Oncogene-induced

senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints.

Nature 444, 633-637.

Belgnaoui, S.M., Fryrear, K.A., Nyalwidhe, J.O., Guo, X., and Semmes, O.J. (2010). The

viral oncoprotein tax sequesters DNA damage response factors by tethering MDC1 to

chromatin. The Journal of biological chemistry 285, 32897-32905.

Bell, A.I., Groves, K., Kelly, G.L., Croom-Carter, D., Hui, E., Chan, A.T., and Rickinson,

A.B. (2006). Analysis of Epstein-Barr virus latent gene expression in endemic Burkitt's

lymphoma and nasopharyngeal carcinoma tumour cells by using quantitative real-time

PCR assays. The Journal of general virology 87, 2885-2890.

Bester, A.C., Roniger, M., Oren, Y.S., Im, M.M., Sarni, D., Chaoat, M., Bensimon, A.,

Zamir, G., Shewach, D.S., and Kerem, B. (2011). Nucleotide deficiency promotes

genomic instability in early stages of cancer development. Cell 145, 435-446.

Bild, A.H., Yao, G., Chang, J.T., Wang, Q., Potti, A., Chasse, D., Joshi, M.B., Harpole, D.,

Lancaster, J.M., Berchuck, A., et al. (2006). Oncogenic pathway signatures in human

cancers as a guide to targeted therapies. Nature 439, 353-357.

Boichuk, S., Hu, L., Hein, J., and Gjoerup, O.V. (2010). Multiple DNA damage signaling

and repair pathways deregulated by simian virus 40 large T antigen. Journal of virology

84, 8007-8020.

Bonsignori, M., Hwang, K.K., Chen, X., Tsao, C.Y., Morris, L., Gray, E., Marshall, D.J.,

Crump, J.A., Kapiga, S.H., Sam, N.E., et al. (2011). Analysis of a clonal lineage of HIV-1

envelope V2/V3 conformational epitope-specific broadly neutralizing antibodies and

their inferred unmutated common ancestors. Journal of virology 85, 9998-10009.

94

Borel, J.F., Feurer, C., Magnee, C., and Stahelin, H. (1977). Effects of the new anti-

lymphocytic peptide cyclosporin A in animals. Immunology 32, 1017-1025.

Bouvard, V., Baan, R., Straif, K., Grosse, Y., Secretan, B., El Ghissassi, F., Benbrahim-

Tallaa, L., Guha, N., Freeman, C., Galichet, L., et al. (2009). A review of human

carcinogens--Part B: biological agents. Lancet Oncol 10, 321-322.

Brooks, L., Yao, Q.Y., Rickinson, A.B., and Young, L.S. (1992). Epstein-Barr virus latent

gene transcription in nasopharyngeal carcinoma cells: coexpression of EBNA1, LMP1,

and LMP2 transcripts. Journal of virology 66, 2689-2697.

Cai, X., Schafer, A., Lu, S., Bilello, J.P., Desrosiers, R.C., Edwards, R., Raab-Traub, N.,

and Cullen, B.R. (2006). Epstein-Barr virus microRNAs are evolutionarily conserved and

differentially expressed. PLoS pathogens 2, e23.

Chandhasin, C., Ducu, R.I., Berkovich, E., Kastan, M.B., and Marriott, S.J. (2008). Human

T-cell leukemia virus type 1 tax attenuates the ATM-mediated cellular DNA damage

response. Journal of virology 82, 6952-6961.

Chang, J.T., and Nevins, J.R. (2006). GATHER: a systems approach to interpreting

genomic signatures. Bioinformatics 22, 2926-2933.

Chang, M.H., Ng, C.K., Lin, Y.J., Liang, C.L., Chung, P.J., ChenMl, Tyan, Y.S., Hsu, C.Y.,

Shu, C.H., and Chang, Y.S. (1997). Identification of a promoter for the latent membrane

protein 1 gene of Epstein-Barr virus that is specifically activated in human epithelial

cells. DNA and cell biology 16, 829-837.

Chen, W., Hilton, I.B., Staudt, M.R., Burd, C.E., and Dittmer, D.P. (2010). Distinct p53,

p53:LANA, and LANA complexes in Kaposi's Sarcoma--associated Herpesvirus

Lymphomas. Journal of virology 84, 3898-3908.

Cheng, S., Schmidt-Grimminger, D.C., Murant, T., Broker, T.R., and Chow, L.T. (1995).

Differentiation-dependent up-regulation of the human papillomavirus E7 gene

reactivates cellular DNA replication in suprabasal differentiated keratinocytes. Genes

Dev 9, 2335-2349.

95

Choudhuri, T., Verma, S.C., Lan, K., Murakami, M., and Robertson, E.S. (2007). The

ATM/ATR signaling effector Chk2 is targeted by Epstein-Barr virus nuclear antigen 3C

to release the G2/M cell cycle block. Journal of virology 81, 6718-6730.

Christophorou, M.A., Ringshausen, I., Finch, A.J., Swigart, L.B., and Evan, G.I. (2006).

The pathological response to DNA damage does not contribute to p53-mediated tumour

suppression. Nature 443, 214-217.

Clark, E.A., and Ledbetter, J.A. (1986). Activation of human B cells mediated through

two distinct cell surface differentiation antigens, Bp35 and Bp50. Proceedings of the

National Academy of Sciences of the United States of America 83, 4494-4498.

Collarini, E.J., Lee, F.E., Foord, O., Park, M., Sperinde, G., Wu, H., Harriman, W.D.,

Carroll, S.F., Ellsworth, S.L., Anderson, L.J., et al. (2009). Potent high-affinity antibodies

for treatment and prophylaxis of respiratory syncytial virus derived from B cells of

infected patients. J Immunol 183, 6338-6345.

Corti, D., Suguitan, A.L., Jr., Pinna, D., Silacci, C., Fernandez-Rodriguez, B.M., Vanzetta,

F., Santos, C., Luke, C.J., Torres-Velez, F.J., Temperton, N.J., et al. (2010). Heterosubtypic

neutralizing antibodies are produced by individuals immunized with a seasonal

influenza vaccine. The Journal of clinical investigation 120, 1663-1673.

Cote, J.F., and Vuori, K. (2002). Identification of an evolutionarily conserved superfamily

of DOCK180-related proteins with guanine nucleotide exchange activity. Journal of cell

science 115, 4901-4913.

Cotsiki, M., Lock, R.L., Cheng, Y., Williams, G.L., Zhao, J., Perera, D., Freire, R.,

Entwistle, A., Golemis, E.A., Roberts, T.M., et al. (2004). Simian virus 40 large T antigen

targets the spindle assembly checkpoint protein Bub1. Proceedings of the National

Academy of Sciences of the United States of America 101, 947-952.

Cunningham-Rundles, C., Radigan, L., Knight, A.K., Zhang, L., Bauer, L., and

Nakazawa, A. (2006). TLR9 activation is defective in common variable immune

deficiency. J Immunol 176, 1978-1987.

96

DeCaprio, J.A., Ludlow, J.W., Figge, J., Shew, J.Y., Huang, C.M., Lee, W.H., Marsilio, E.,

Paucha, E., and Livingston, D.M. (1988). SV40 large tumor antigen forms a specific

complex with the product of the retinoblastoma susceptibility gene. Cell 54, 275-283.

Deng, Z., Lezina, L., Chen, C.J., Shtivelband, S., So, W., and Lieberman, P.M. (2002).

Telomeric proteins regulate episomal maintenance of Epstein-Barr virus origin of

plasmid replication. Molecular cell 9, 493-503.

Derheimer, F.A., and Kastan, M.B. (2010). Multiple roles of ATM in monitoring and

maintaining DNA integrity. FEBS letters 584, 3675-3681.

Dheekollu, J., Deng, Z., Wiedmer, A., Weitzman, M.D., and Lieberman, P.M. (2007). A

role for MRE11, NBS1, and recombination junctions in replication and stable

maintenance of EBV episomes. PLoS One 2, e1257.

Di Micco, R., Fumagalli, M., Cicalese, A., Piccinin, S., Gasparini, P., Luise, C., Schurra, C.,

Garre, M., Nuciforo, P.G., Bensimon, A., et al. (2006). Oncogene-induced senescence is a

DNA damage response triggered by DNA hyper-replication. Nature 444, 638-642.

Di Micco, R., Sulli, G., Dobreva, M., Liontos, M., Botrugno, O.A., Gargiulo, G., dal Zuffo,

R., Matti, V., d'Ario, G., Montani, E., et al. (2011). Interplay between oncogene-induced

DNA damage response and heterochromatin in senescence and cancer. Nature cell

biology 13, 292-302.

Di Niro, R., Mesin, L., Raki, M., Zheng, N.Y., Lund-Johansen, F., Lundin, K.E.,

Charpilienne, A., Poncet, D., Wilson, P.C., and Sollid, L.M. (2010). Rapid generation of

rotavirus-specific human monoclonal antibodies from small-intestinal mucosa. J

Immunol 185, 5377-5383.

DiTullio, R.A., Jr., Mochan, T.A., Venere, M., Bartkova, J., Sehested, M., Bartek, J., and

Halazonetis, T.D. (2002). 53BP1 functions in an ATM-dependent checkpoint pathway

that is constitutively activated in human cancer. Nature cell biology 4, 998-1002.

Duensing, S., Lee, L.Y., Duensing, A., Basile, J., Piboonniyom, S., Gonzalez, S., Crum,

C.P., and Munger, K. (2000). The human papillomavirus type 16 E6 and E7 oncoproteins

97

cooperate to induce mitotic defects and genomic instability by uncoupling centrosome

duplication from the cell division cycle. Proceedings of the National Academy of

Sciences of the United States of America 97, 10002-10007.

Durkin, S.S., Guo, X., Fryrear, K.A., Mihaylova, V.T., Gupta, S.K., Belgnaoui, S.M.,

Haoudi, A., Kupfer, G.M., and Semmes, O.J. (2008). HTLV-1 Tax oncoprotein subverts

the cellular DNA damage response via binding to DNA-dependent protein kinase. The

Journal of biological chemistry 283, 36311-36320.

Dyson, N., Howley, P.M., Munger, K., and Harlow, E. (1989). The human papilloma

virus-16 E7 oncoprotein is able to bind to the retinoblastoma gene product. Science (New

York, NY 243, 934-937.

Efeyan, A., Garcia-Cao, I., Herranz, D., Velasco-Miguel, S., and Serrano, M. (2006).

Tumour biology: Policing of oncogene activity by p53. Nature 443, 159.

Elkon, R., Rashi-Elkeles, S., Lerenthal, Y., Linhart, C., Tenne, T., Amariglio, N., Rechavi,

G., Shamir, R., and Shiloh, Y. (2005). Dissection of a DNA-damage-induced

transcriptional network using a combination of microarrays, RNA interference and

computational promoter analysis. Genome Biol 6, R43.

Elmore, L.W., Hancock, A.R., Chang, S.F., Wang, X.W., Chang, S., Callahan, C.P., Geller,

D.A., Will, H., and Harris, C.C. (1997). Hepatitis B virus X protein and p53 tumor

suppressor interactions in the modulation of apoptosis. Proc Natl Acad Sci U S A 94,

14707-14712.

Fagarasan, S., and Honjo, T. (2000). T-Independent immune response: new aspects of B

cell biology. Science (New York, NY 290, 89-92.

Falck, J., Mailand, N., Syljuasen, R.G., Bartek, J., and Lukas, J. (2001). The ATM-Chk2-

Cdc25A checkpoint pathway guards against radioresistant DNA synthesis. Nature 410,

842-847.

98

Fennewald, S., van Santen, V., and Kieff, E. (1984). Nucleotide sequence of an mRNA

transcribed in latent growth-transforming virus infection indicates that it may encode a

membrane protein. Journal of virology 51, 411-419.

Forte, E., and Luftig, M.A. (2009). MDM2-dependent inhibition of p53 is required for

Epstein-Barr virus B-cell growth transformation and infected-cell survival. Journal of

virology 83, 2491-2499.

Forte, E., and Luftig, M.A. (2011). The role of microRNAs in Epstein-Barr virus latency

and lytic reactivation. Microbes and infection / Institut Pasteur 13, 1156-1167.

Friberg, H., Jaiswal, S., West, K., O'Ketch, M., Rothman, A.L., and Mathew, A. (2012).

Analysis of human monoclonal antibodies generated by dengue virus-specific memory

B cells. Viral immunology 25, 348-359.

Friborg, J., Jr., Kong, W., Hottiger, M.O., and Nabel, G.J. (1999). p53 inhibition by the

LANA protein of KSHV protects against cell death. Nature 402, 889-894.

Goodnow, C.C., Vinuesa, C.G., Randall, K.L., Mackay, F., and Brink, R. (2010). Control

systems and decision making for antibody production. Nature immunology 11, 681-688.

Gorgoulis, V.G., Vassiliou, L.V., Karakaidos, P., Zacharatos, P., Kotsinas, A., Liloglou, T.,

Venere, M., Ditullio, R.A., Jr., Kastrinakis, N.G., Levy, B., et al. (2005). Activation of the

DNA damage checkpoint and genomic instability in human precancerous lesions.

Nature 434, 907-913.

Greenwell, P.W., Kronmal, S.L., Porter, S.E., Gassenhuber, J., Obermaier, B., and Petes,

T.D. (1995). TEL1, a gene involved in controlling telomere length in S. cerevisiae, is

homologous to the human ataxia telangiectasia gene. Cell 82, 823-829.

Gruhne, B., Sompallae, R., Marescotti, D., Kamranvar, S.A., Gastaldello, S., and Masucci,

M.G. (2009a). The Epstein-Barr virus nuclear antigen-1 promotes genomic instability via

induction of reactive oxygen species. Proceedings of the National Academy of Sciences

of the United States of America 106, 2313-2318.

99

Gruhne, B., Sompallae, R., and Masucci, M.G. (2009b). Three Epstein-Barr virus latency

proteins independently promote genomic instability by inducing DNA damage,

inhibiting DNA repair and inactivating cell cycle checkpoints. Oncogene 28, 3997-4008.

Grundhoff, A., Sullivan, C.S., and Ganem, D. (2006). A combined computational and

microarray-based approach identifies novel microRNAs encoded by human gamma-

herpesviruses. RNA 12, 733-750.

Gupta, S.K., Guo, X., Durkin, S.S., Fryrear, K.F., Ward, M.D., and Semmes, O.J. (2007).

Human T-cell leukemia virus type 1 Tax oncoprotein prevents DNA damage-induced

chromatin egress of hyperphosphorylated Chk2. The Journal of biological chemistry 282,

29431-29440.

Hacker, H., Vabulas, R.M., Takeuchi, O., Hoshino, K., Akira, S., and Wagner, H. (2000).

Immune cell activation by bacterial CpG-DNA through myeloid differentiation marker

88 and tumor necrosis factor receptor-associated factor (TRAF)6. The Journal of

experimental medicine 192, 595-600.

Halazonetis, T.D., Gorgoulis, V.G., and Bartek, J. (2008). An oncogene-induced DNA

damage model for cancer development. Science (New York, NY 319, 1352-1355.

Harada, Y., Tanaka, Y., Terasawa, M., Pieczyk, M., Habiro, K., Katakai, T., Hanawa-

Suetsugu, K., Kukimoto-Niino, M., Nishizaki, T., Shirouzu, M., et al. (2012). DOCK8 is a

Cdc42 activator critical for interstitial dendritic cell migration during immune responses.

Blood 119, 4451-4461.

Harper, J.W., and Elledge, S.J. (2007). The DNA damage response: ten years after.

Molecular cell 28, 739-745.

Hartmann, G., and Krieg, A.M. (2000). Mechanism and function of a newly identified

CpG DNA motif in human primary B cells. J Immunol 164, 944-953.

Hawkins, E.D., Hommel, M., Turner, M.L., Battye, F.L., Markham, J.F., and Hodgkin,

P.D. (2007a). Measuring lymphocyte proliferation, survival and differentiation using

CFSE time-series data. Nature protocols 2, 2057-2067.

100

Hawkins, E.D., Turner, M.L., Dowling, M.R., van Gend, C., and Hodgkin, P.D. (2007b).

A model of immune regulation as a consequence of randomized lymphocyte division

and death times. Proceedings of the National Academy of Sciences of the United States

of America 104, 5032-5037.

Haynes, B.F., Fleming, J., St Clair, E.W., Katinger, H., Stiegler, G., Kunert, R., Robinson,

J., Scearce, R.M., Plonk, K., Staats, H.F., et al. (2005). Cardiolipin polyspecific

autoreactivity in two broadly neutralizing HIV-1 antibodies. Science (New York, NY

308, 1906-1908.

Haynes, B.F., Kelsoe, G., Harrison, S.C., and Kepler, T.B. (2012). B-cell-lineage

immunogen design in vaccine development with HIV-1 as a case study. Nature

biotechnology 30, 423-433.

Heath, E., Begue-Pastor, N., Chaganti, S., Croom-Carter, D., Shannon-Lowe, C., Kube,

D., Feederle, R., Delecluse, H.J., Rickinson, A.B., and Bell, A.I. (2012). Epstein-Barr virus

infection of naive B cells in vitro frequently selects clones with mutated immunoglobulin

genotypes: implications for virus biology. PLoS Pathog 8, e1002697.

Hein, J., Boichuk, S., Wu, J., Cheng, Y., Freire, R., Jat, P.S., Roberts, T.M., and Gjoerup,

O.V. (2009). Simian virus 40 large T antigen disrupts genome integrity and activates a

DNA damage response via Bub1 binding. Journal of virology 83, 117-127.

Henderson, E., Miller, G., Robinson, J., and Heston, L. (1977). Efficiency of

transformation of lymphocytes by Epstein-Barr virus. Virology 76, 152-163.

Henkel, T., Ling, P.D., Hayward, S.D., and Peterson, M.G. (1994). Mediation of Epstein-

Barr virus EBNA2 transactivation by recombination signal-binding protein J kappa.

Science (New York, NY 265, 92-95.

Hertle, M.L., Popp, C., Petermann, S., Maier, S., Kremmer, E., Lang, R., Mages, J., and

Kempkes, B. (2009). Differential gene expression patterns of EBV infected EBNA-3A

positive and negative human B lymphocytes. PLoS pathogens 5, e1000506.

101

Hicar, M.D., Kalams, S.A., Spearman, P.W., and Crowe, J.E., Jr. (2010). Emerging studies

of human HIV-specific antibody repertoires. Vaccine 28 Suppl 2, B18-23.

Hickson, I., Zhao, Y., Richardson, C.J., Green, S.J., Martin, N.M., Orr, A.I., Reaper, P.M.,

Jackson, S.P., Curtin, N.J., and Smith, G.C. (2004). Identification and characterization of a

novel and specific inhibitor of the ataxia-telangiectasia mutated kinase ATM. Cancer Res

64, 9152-9159.

Hirao, A., Cheung, A., Duncan, G., Girard, P.M., Elia, A.J., Wakeham, A., Okada, H.,

Sarkissian, T., Wong, J.A., Sakai, T., et al. (2002). Chk2 is a tumor suppressor that

regulates apoptosis in both an ataxia telangiectasia mutated (ATM)-dependent and an

ATM-independent manner. Molecular and cellular biology 22, 6521-6532.

Hirao, A., Kong, Y.Y., Matsuoka, S., Wakeham, A., Ruland, J., Yoshida, H., Liu, D.,

Elledge, S.J., and Mak, T.W. (2000). DNA damage-induced activation of p53 by the

checkpoint kinase Chk2. Science (New York, NY 287, 1824-1827.

Hong, S., Pusapati, R.V., Powers, J.T., and Johnson, D.G. (2006). Oncogenes and the

DNA Damage Response: Myc and E2F1 Engage the ATM Signaling Pathway to Activate

p53 and Induce Apoptosis. Cell Cycle 5.

Hu, H., Voss, J., Zhang, G., Buchy, P., Zuo, T., Wang, L., Wang, F., Zhou, F., Wang, G.,

Tsai, C., et al. (2012). A human antibody recognizing a conserved epitope of H5

hemagglutinin broadly neutralizes highly pathogenic avian influenza H5N1 viruses.

Journal of virology 86, 2978-2989.

Hurley, E.A., and Thorley-Lawson, D.A. (1988). B cell activation and the establishment

of Epstein-Barr virus latency. The Journal of experimental medicine 168, 2059-2075.

Iskra, S., Kalla, M., Delecluse, H.J., Hammerschmidt, W., and Moosmann, A. (2010). Toll-

like receptor agonists synergistically increase proliferation and activation of B cells by

epstein-barr virus. Journal of virology 84, 3612-3623.

Jabara, H.H., McDonald, D.R., Janssen, E., Massaad, M.J., Ramesh, N., Borzutzky, A.,

Rauter, I., Benson, H., Schneider, L., Baxi, S., et al. (2012). DOCK8 functions as an

102

adaptor that links TLR-MyD88 signaling to B cell activation. Nature immunology 13,

612-620.

Jin, D.Y., Spencer, F., and Jeang, K.T. (1998). Human T cell leukemia virus type 1

oncoprotein Tax targets the human mitotic checkpoint protein MAD1. Cell 93, 81-91.

Johannsen, E., Luftig, M., Chase, M.R., Weicksel, S., Cahir-McFarland, E., Illanes, D.,

Sarracino, D., and Kieff, E. (2004). Proteins of purified Epstein-Barr virus. Proc Natl

Acad Sci U S A 101, 16286-16291.

Johannsen, E., Miller, C.L., Grossman, S.R., and Kieff, E. (1996). EBNA-2 and EBNA-3C

extensively and mutually exclusively associate with RBPJkappa in Epstein-Barr virus-

transformed B lymphocytes. Journal of virology 70, 4179-4183.

Kaiser, C., Laux, G., Eick, D., Jochner, N., Bornkamm, G.W., and Kempkes, B. (1999). The

proto-oncogene c-myc is a direct target gene of Epstein-Barr virus nuclear antigen 2.

Journal of virology 73, 4481-4484.

Kalla, M., Schmeinck, A., Bergbauer, M., Pich, D., and Hammerschmidt, W. (2010). AP-1

homolog BZLF1 of Epstein-Barr virus has two essential functions dependent on the

epigenetic state of the viral genome. Proc Natl Acad Sci U S A 107, 850-855.

Kamranvar, S.A., and Masucci, M.G. (2011). The Epstein-Barr virus nuclear antigen-1

promotes telomere dysfunction via induction of oxidative stress. Leukemia 25, 1017-

1025.

Karlseder, J., Broccoli, D., Dai, Y., Hardy, S., and de Lange, T. (1999). p53- and ATM-

dependent apoptosis induced by telomeres lacking TRF2. Science (New York, NY 283,

1321-1325.

Kaye, K.M., Izumi, K.M., and Kieff, E. (1993). Epstein-Barr virus latent membrane

protein 1 is essential for B-lymphocyte growth transformation. Proceedings of the

National Academy of Sciences of the United States of America 90, 9150-9154.

103

Khanna, K.K., and Jackson, S.P. (2001). DNA double-strand breaks: signaling, repair and

the cancer connection. Nature genetics 27, 247-254.

Khanna, R., Burrows, S.R., Kurilla, M.G., Jacob, C.A., Misko, I.S., Sculley, T.B., Kieff, E.,

and Moss, D.J. (1992). Localization of Epstein-Barr virus cytotoxic T cell epitopes using

recombinant vaccinia: implications for vaccine development. The Journal of

experimental medicine 176, 169-176.

Kieff, E., and Rickinson, A. (2006). Epstein-Barr virus and its replication. In Fields

Virology, D.M. Knipe, and P.M. Howley, eds. (Philadelphia: Lippincott, Williams, and

Wilkins), pp. 2603-2654.

Kim, S.T., Lim, D.S., Canman, C.E., and Kastan, M.B. (1999). Substrate specificities and

identification of putative substrates of ATM kinase family members. The Journal of

biological chemistry 274, 37538-37543.

Kinjo, T., Ham-Terhune, J., Peloponese, J.M., Jr., and Jeang, K.T. (2010). Induction of

reactive oxygen species by human T-cell leukemia virus type 1 tax correlates with DNA

damage and expression of cellular senescence marker. Journal of virology 84, 5431-5437.

Klein, N.P., and Schneider, R.J. (1997). Activation of Src family kinases by hepatitis B

virus HBx protein and coupled signaling to Ras. Molecular and cellular biology 17, 6427-

6436.

Knight, J.S., and Robertson, E.S. (2004). Epstein-Barr virus nuclear antigen 3C regulates

cyclin A/p27 complexes and enhances cyclin A-dependent kinase activity. Journal of

virology 78, 1981-1991.

Koopal, S., Furuhjelm, J.H., Jarviluoma, A., Jaamaa, S., Pyakurel, P., Pussinen, C.,

Wirzenius, M., Biberfeld, P., Alitalo, K., Laiho, M., et al. (2007). Viral oncogene-induced

DNA damage response is activated in Kaposi sarcoma tumorigenesis. PLoS Pathog 3,

1348-1360.

104

Korzeniewski, N., Treat, B., and Duensing, S. (2011). The HPV-16 E7 oncoprotein

induces centriole multiplication through deregulation of Polo-like kinase 4 expression.

Mol Cancer 10, 61.

Kozbor, D., and Roder, J.C. (1981). Requirements for the establishment of high-titered

human monoclonal antibodies against tetanus toxoid using the Epstein-Barr virus

technique. J Immunol 127, 1275-1280.

Krause, J.C., Tsibane, T., Tumpey, T.M., Huffman, C.J., Albrecht, R., Blum, D.L., Ramos,

I., Fernandez-Sesma, A., Edwards, K.M., Garcia-Sastre, A., et al. (2012). Human

monoclonal antibodies to pandemic 1957 H2N2 and pandemic 1968 H3N2 influenza

viruses. Journal of virology 86, 6334-6340.

Krause, J.C., Tsibane, T., Tumpey, T.M., Huffman, C.J., Basler, C.F., and Crowe, J.E., Jr.

(2011). A broadly neutralizing human monoclonal antibody that recognizes a conserved,

novel epitope on the globular head of the influenza H1N1 virus hemagglutinin. Journal

of virology 85, 10905-10908.

Kress, M., May, E., Cassingena, R., and May, P. (1979). Simian virus 40-transformed cells

express new species of proteins precipitable by anti-simian virus 40 tumor serum.

Journal of virology 31, 472-483.

Kudoh, A., Fujita, M., Zhang, L., Shirata, N., Daikoku, T., Sugaya, Y., Isomura, H.,

Nishiyama, Y., and Tsurumi, T. (2005). Epstein-Barr virus lytic replication elicits ATM

checkpoint signal transduction while providing an S-phase-like cellular environment.

The Journal of biological chemistry 280, 8156-8163.

Kulwichit, W., Edwards, R.H., Davenport, E.M., Baskar, J.F., Godfrey, V., and Raab-

Traub, N. (1998). Expression of the Epstein-Barr virus latent membrane protein 1

induces B cell lymphoma in transgenic mice. Proceedings of the National Academy of

Sciences of the United States of America 95, 11963-11968.

Kwan-Ki Hwang, M.L. (2009). MONOCLONAL ANTIBODY PRODUCTION IN B

CELLS AND USES THEROF, USPTO, ed. (USA: Duke University).

105

Lacoste, S., Wiechec, E., Dos Santos Silva, A.G., Guffei, A., Williams, G., Lowbeer, M.,

Benedek, K., Henriksson, M., Klein, G., and Mai, S. (2009). Chromosomal

rearrangements after ex vivo Epstein-Barr virus (EBV) infection of human B cells.

Oncogene.

Lane, D.P., and Crawford, L.V. (1979). T antigen is bound to a host protein in SV40-

transformed cells. Nature 278, 261-263.

Latz, E., Verma, A., Visintin, A., Gong, M., Sirois, C.M., Klein, D.C., Monks, B.G.,

McKnight, C.J., Lamphier, M.S., Duprex, W.P., et al. (2007). Ligand-induced

conformational changes allosterically activate Toll-like receptor 9. Nature immunology

8, 772-779.

Leao, M., Anderton, E., Wade, M., Meekings, K., and Allday, M.J. (2007). Epstein-barr

virus-induced resistance to drugs that activate the mitotic spindle assembly checkpoint

in Burkitt's lymphoma cells. Journal of virology 81, 248-260.

Leidal, A.M., Cyr, D.P., Hill, R.J., Lee, P.W., and McCormick, C. (2012). Subversion of

autophagy by Kaposi's sarcoma-associated herpesvirus impairs oncogene-induced

senescence. Cell host & microbe 11, 167-180.

Lilley, C.E., Schwartz, R.A., and Weitzman, M.D. (2007). Using or abusing: viruses and

the cellular DNA damage response. Trends Microbiol 15, 119-126.

Liu, B., Hong, S., Tang, Z., Yu, H., and Giam, C.Z. (2005). HTLV-I Tax directly binds the

Cdc20-associated anaphase-promoting complex and activates it ahead of schedule.

Proceedings of the National Academy of Sciences of the United States of America 102,

63-68.

Liu, J., Martin, H.J., Liao, G., and Hayward, S.D. (2007). The Kaposi's sarcoma-associated

herpesvirus LANA protein stabilizes and activates c-Myc. Journal of virology 81, 10451-

10459.

Lomaga, M.A., Yeh, W.C., Sarosi, I., Duncan, G.S., Furlonger, C., Ho, A., Morony, S.,

Capparelli, C., Van, G., Kaufman, S., et al. (1999). TRAF6 deficiency results in

106

osteopetrosis and defective interleukin-1, CD40, and LPS signaling. Genes Dev 13, 1015-

1024.

Macagno, A., Bernasconi, N.L., Vanzetta, F., Dander, E., Sarasini, A., Revello, M.G.,

Gerna, G., Sallusto, F., and Lanzavecchia, A. (2010). Isolation of human monoclonal

antibodies that potently neutralize human cytomegalovirus infection by targeting

different epitopes on the gH/gL/UL128-131A complex. Journal of virology 84, 1005-1013.

Maclean, K.H., Dorsey, F.C., Cleveland, J.L., and Kastan, M.B. (2008). Targeting

lysosomal degradation induces p53-dependent cell death and prevents cancer in mouse

models of lymphomagenesis. The Journal of clinical investigation 118, 79-88.

MacLennan, I.C. (1994). Germinal centers. Annu Rev Immunol 12, 117-139.

Markham, J.F., Wellard, C.J., Hawkins, E.D., Duffy, K.R., and Hodgkin, P.D. (2010). A

minimum of two distinct heritable factors are required to explain correlation structures

in proliferating lymphocytes. Journal of the Royal Society, Interface / the Royal Society 7,

1049-1059.

Martin, H.J., Lee, J.M., Walls, D., and Hayward, S.D. (2007). Manipulation of the toll-like

receptor 7 signaling pathway by Epstein-Barr virus. Journal of virology 81, 9748-9758.

Maruo, S., Zhao, B., Johannsen, E., Kieff, E., Zou, J., and Takada, K. (2011). Epstein-Barr

virus nuclear antigens 3C and 3A maintain lymphoblastoid cell growth by repressing

p16INK4A and p14ARF expression. Proceedings of the National Academy of Sciences of

the United States of America 108, 1919-1924.

Matsuoka, S., Huang, M., and Elledge, S.J. (1998). Linkage of ATM to cell cycle

regulation by the Chk2 protein kinase. Science (New York, NY 282, 1893-1897.

Mauser, A., Saito, S., Appella, E., Anderson, C.W., Seaman, W.T., and Kenney, S. (2002).

The Epstein-Barr virus immediate-early protein BZLF1 regulates p53 function through

multiple mechanisms. Journal of virology 76, 12503-12512.

107

McPherson, J.P., Lemmers, B., Hirao, A., Hakem, A., Abraham, J., Migon, E., Matysiak-

Zablocki, E., Tamblyn, L., Sanchez-Sweatman, O., Khokha, R., et al. (2004). Collaboration

of Brca1 and Chk2 in tumorigenesis. Genes Dev 18, 1144-1153.

Miyazato, A., Sheleg, S., Iha, H., Li, Y., and Jeang, K.T. (2005). Evidence for NF-kappaB-

and CBP-independent repression of p53's transcriptional activity by human T-cell

leukemia virus type 1 Tax in mouse embryo and primary human fibroblasts. Journal of

virology 79, 9346-9350.

Moody, C.A., and Laimins, L.A. (2009). Human papillomaviruses activate the ATM

DNA damage pathway for viral genome amplification upon differentiation. PLoS

Pathog 5, e1000605.

Munger, K., Werness, B.A., Dyson, N., Phelps, W.C., Harlow, E., and Howley, P.M.

(1989). Complex formation of human papillomavirus E7 proteins with the

retinoblastoma tumor suppressor gene product. The EMBO journal 8, 4099-4105.

Murphy, D.J., Junttila, M.R., Pouyet, L., Karnezis, A., Shchors, K., Bui, D.A., Brown-

Swigart, L., Johnson, L., and Evan, G.I. (2008). Distinct thresholds govern Myc's

biological output in vivo. Cancer Cell 14, 447-457.

Murray, R.J., Kurilla, M.G., Brooks, J.M., Thomas, W.A., Rowe, M., Kieff, E., and

Rickinson, A.B. (1992). Identification of target antigens for the human cytotoxic T cell

response to Epstein-Barr virus (EBV): implications for the immune control of EBV-

positive malignancies. The Journal of experimental medicine 176, 157-168.

Nguyen, C.L., and Munger, K. (2009). Human papillomavirus E7 protein deregulates

mitosis via an association with nuclear mitotic apparatus protein 1. Journal of virology

83, 1700-1707.

Nikitin, P.A., and Luftig, M.A. (2012). The DNA damage response in viral-induced

cellular transformation. British journal of cancer 106, 429-435.

Nikitin, P.A., Yan, C.M., Forte, E., Bocedi, A., Tourigny, J.P., White, R.E., Allday, M.J.,

Patel, A., Dave, S.S., Kim, W., et al. (2010). An ATM/Chk2-mediated DNA damage-

108

responsive signaling pathway suppresses Epstein-Barr virus transformation of primary

human B cells. Cell Host Microbe 8, 510-522.

Osborn, A.J., Elledge, S.J., and Zou, L. (2002). Checking on the fork: the DNA-replication

stress-response pathway. Trends in cell biology 12, 509-516.

Park, H.U., Jeong, J.H., Chung, J.H., and Brady, J.N. (2004). Human T-cell leukemia virus

type 1 Tax interacts with Chk1 and attenuates DNA-damage induced G2 arrest

mediated by Chk1. Oncogene 23, 4966-4974.

Park, H.U., Jeong, S.J., Jeong, J.H., Chung, J.H., and Brady, J.N. (2006). Human T-cell

leukemia virus type 1 Tax attenuates gamma-irradiation-induced apoptosis through

physical interaction with Chk2. Oncogene 25, 438-447.

Parker, G.A., Touitou, R., and Allday, M.J. (2000). Epstein-Barr virus EBNA3C can

disrupt multiple cell cycle checkpoints and induce nuclear division divorced from

cytokinesis. Oncogene 19, 700-709.

Pathmanathan, R., Prasad, U., Chandrika, G., Sadler, R., Flynn, K., and Raab-Traub, N.

(1995). Undifferentiated, nonkeratinizing, and squamous cell carcinoma of the

nasopharynx. Variants of Epstein-Barr virus-infected neoplasia. The American journal of

pathology 146, 1355-1367.

Peng, S.L. (2005). Signaling in B cells via Toll-like receptors. Current opinion in

immunology 17, 230-236.

Perkins, N.D. (2012). The diverse and complex roles of NF-kappaB subunits in cancer.

Nature reviews Cancer 12, 121-132.

Pfeffer, S., Zavolan, M., Grasser, F.A., Chien, M., Russo, J.J., Ju, J., John, B., Enright, A.J.,

Marks, D., Sander, C., et al. (2004). Identification of virus-encoded microRNAs. Science

(New York, NY 304, 734-736.

109

Pise-Masison, C.A., Mahieux, R., Radonovich, M., Jiang, H., Duvall, J., Guillerm, C., and

Brady, J.N. (2000). Insights into the molecular mechanism of p53 inhibition by HTLV

type 1 Tax. AIDS Res Hum Retroviruses 16, 1669-1675.

Poulsen, T.R., Jensen, A., Haurum, J.S., and Andersen, P.S. (2011). Limits for antibody

affinity maturation and repertoire diversification in hypervaccinated humans. J

Immunol 187, 4229-4235.

Powers, J.T., Hong, S., Mayhew, C.N., Rogers, P.M., Knudsen, E.S., and Johnson, D.G.

(2004). E2F1 uses the ATM signaling pathway to induce p53 and Chk2 phosphorylation

and apoptosis. Molecular cancer research : MCR 2, 203-214.

Price, A.M., Tourigny, J.P., Forte, E., Salinas, R.E., Dave, S.S., and Luftig, M.A. (2012).

Analysis of Epstein-Barr Virus-Regulated Host Gene Expression Changes through

Primary B-Cell Outgrowth Reveals Delayed Kinetics of Latent Membrane Protein 1-

Mediated NF-kappaB Activation. Journal of virology 86, 11096-11106.

Pusapati, R.V., Rounbehler, R.J., Hong, S., Powers, J.T., Yan, M., Kiguchi, K., McArthur,

M.J., Wong, P.K., and Johnson, D.G. (2006). ATM promotes apoptosis and suppresses

tumorigenesis in response to Myc. Proceedings of the National Academy of Sciences of

the United States of America 103, 1446-1451.

Raab-Traub, N. (2002). Epstein-Barr virus in the pathogenesis of NPC. Seminars in

cancer biology 12, 431-441.

Raab-Traub, N. (2007). EBV-induced oncogenesis. In Human Herpesviruses: Biology,

Therapy, and Immunoprophylaxis, A. Arvin, G. Campadelli-Fiume, E. Mocarski, P.S.

Moore, B. Roizman, R. Whitley, and K. Yamanishi, eds. (Cambridge).

Raab-Traub, N. (2012). Novel mechanisms of EBV-induced oncogenesis. Current opinion

in virology 2, 453-458.

Ranuncolo, S.M., Polo, J.M., Dierov, J., Singer, M., Kuo, T., Greally, J., Green, R., Carroll,

M., and Melnick, A. (2007). Bcl-6 mediates the germinal center B cell phenotype and

110

lymphomagenesis through transcriptional repression of the DNA-damage sensor ATR.

Nature immunology 8, 705-714.

Richman, D.D., Wrin, T., Little, S.J., and Petropoulos, C.J. (2003). Rapid evolution of the

neutralizing antibody response to HIV type 1 infection. Proceedings of the National

Academy of Sciences of the United States of America 100, 4144-4149.

Rickinson, A., and Kieff, E. (2006). Epstein-Barr virus In Fields Virology, D.M. Knipe,

and P.M. Howley, eds. (Philadelphia: Lippincott, Williams, and Wilkins), pp. 2603-2654.

Robertson, E.S., Lin, J., and Kieff, E. (1996). The amino-terminal domains of Epstein-Barr

virus nuclear proteins 3A, 3B, and 3C interact with RBPJ(kappa). Journal of virology 70,

3068-3074.

Robinson, T.J., Forte, E., Salinas, R.E., Puri, S., Marengo, M., Garcia-Blanco, M.A., and

Luftig, M.A. (2012). SplicerEX: a tool for the automated detection and classification of

mRNA changes from conventional and splice-sensitive microarray expression data.

RNA 18, 1435-1445.

Rodier, F., Coppe, J.P., Patil, C.K., Hoeijmakers, W.A., Munoz, D.P., Raza, S.R., Freund,

A., Campeau, E., Davalos, A.R., and Campisi, J. (2009). Persistent DNA damage

signalling triggers senescence-associated inflammatory cytokine secretion. Nature cell

biology 11, 973-979.

Rodier, F., Munoz, D.P., Teachenor, R., Chu, V., Le, O., Bhaumik, D., Coppe, J.P.,

Campeau, E., Beausejour, C.M., Kim, S.H., et al. (2011). DNA-SCARS: distinct nuclear

structures that sustain damage-induced senescence growth arrest and inflammatory

cytokine secretion. Journal of cell science 124, 68-81.

Rogoff, H.A., Pickering, M.T., Frame, F.M., Debatis, M.E., Sanchez, Y., Jones, S., and

Kowalik, T.F. (2004). Apoptosis associated with deregulated E2F activity is dependent

on E2F1 and Atm/Nbs1/Chk2. Molecular and cellular biology 24, 2968-2977.

Roughan, J.E., and Thorley-Lawson, D.A. (2009). The intersection of Epstein-Barr virus

with the germinal center. Journal of virology 83, 3968-3976.

111

Rousset, F., Garcia, E., and Banchereau, J. (1991). Cytokine-induced proliferation and

immunoglobulin production of human B lymphocytes triggered through their CD40

antigen. The Journal of experimental medicine 173, 705-710.

Rowe, M., Lear, A.L., Croom-Carter, D., Davies, A.H., and Rickinson, A.B. (1992). Three

pathways of Epstein-Barr virus gene activation from EBNA1-positive latency in B

lymphocytes. Journal of virology 66, 122-131.

Rowe, M., Rowe, D.T., Gregory, C.D., Young, L.S., Farrell, P.J., Rupani, H., and

Rickinson, A.B. (1987). Differences in B cell growth phenotype reflect novel patterns of

Epstein-Barr virus latent gene expression in Burkitt's lymphoma cells. The EMBO

journal 6, 2743-2751.

Sadler, R.H., and Raab-Traub, N. (1995). The Epstein-Barr virus 3.5-kilobase latent

membrane protein 1 mRNA initiates from a TATA-Less promoter within the first

terminal repeat. Journal of virology 69, 4577-4581.

Sato, K., Ohta, T., and Venkitaraman, A.R. (2010). A mitotic role for the DNA damage-

responsive CHK2 kinase. Nature cell biology 12, 424-425.

Savitsky, K., Bar-Shira, A., Gilad, S., Rotman, G., Ziv, Y., Vanagaite, L., Tagle, D.A.,

Smith, S., Uziel, T., Sfez, S., et al. (1995). A single ataxia telangiectasia gene with a

product similar to PI-3 kinase. Science (New York, NY 268, 1749-1753.

Scheffner, M., Huibregtse, J.M., Vierstra, R.D., and Howley, P.M. (1993). The HPV-16 E6

and E6-AP complex functions as a ubiquitin-protein ligase in the ubiquitination of p53.

Cell 75, 495-505.

Scheffner, M., Werness, B.A., Huibregtse, J.M., Levine, A.J., and Howley, P.M. (1990).

The E6 oncoprotein encoded by human papillomavirus types 16 and 18 promotes the

degradation of p53. Cell 63, 1129-1136.

Schlager, S., Speck, S.H., and Woisetschlager, M. (1996). Transcription of the Epstein-

Barr virus nuclear antigen 1 (EBNA1) gene occurs before induction of the BCR2 (Cp)

112

EBNA gene promoter during the initial stages of infection in B cells. Journal of virology

70, 3561-3570.

Sengupta, S., den Boon, J.A., Chen, I.H., Newton, M.A., Dahl, D.B., Chen, M., Cheng,

Y.J., Westra, W.H., Chen, C.J., Hildesheim, A., et al. (2006). Genome-wide expression

profiling reveals EBV-associated inhibition of MHC class I expression in nasopharyngeal

carcinoma. Cancer Res 66, 7999-8006.

Shiloh, Y. (2003). ATM and related protein kinases: safeguarding genome integrity.

Nature reviews Cancer 3, 155-168.

Shuda, M., Feng, H., Kwun, H.J., Rosen, S.T., Gjoerup, O., Moore, P.S., and Chang, Y.

(2008). T antigen mutations are a human tumor-specific signature for Merkel cell

polyomavirus. Proceedings of the National Academy of Sciences of the United States of

America 105, 16272-16277.

Siemer, D., Kurth, J., Lang, S., Lehnerdt, G., Stanelle, J., and Kuppers, R. (2008). EBV

transformation overrides gene expression patterns of B cell differentiation stages. Mol

Immunol 45, 3133-3141.

Simmons, C.P., Bernasconi, N.L., Suguitan, A.L., Mills, K., Ward, J.M., Chau, N.V., Hien,

T.T., Sallusto, F., Ha do, Q., Farrar, J., et al. (2007). Prophylactic and therapeutic efficacy

of human monoclonal antibodies against H5N1 influenza. PLoS medicine 4, e178.

Sinclair, A.J., Palmero, I., Peters, G., and Farrell, P.J. (1994). EBNA-2 and EBNA-LP

cooperate to cause G0 to G1 transition during immortalization of resting human B

lymphocytes by Epstein-Barr virus. The EMBO journal 13, 3321-3328.

Skalska, L., White, R.E., Franz, M., Ruhmann, M., and Allday, M.J. (2010). Epigenetic

repression of p16(INK4A) by latent Epstein-Barr virus requires the interaction of

EBNA3A and EBNA3C with CtBP. PLoS Pathog 6, e1000951.

Skalsky, R.L., Corcoran, D.L., Gottwein, E., Frank, C.L., Kang, D., Hafner, M., Nusbaum,

J.D., Feederle, R., Delecluse, H.J., Luftig, M.A., et al. (2012). The viral and cellular

microRNA targetome in lymphoblastoid cell lines. PLoS pathogens 8, e1002484.

113

Smith, K., Garman, L., Wrammert, J., Zheng, N.Y., Capra, J.D., Ahmed, R., and Wilson,

P.C. (2009). Rapid generation of fully human monoclonal antibodies specific to a

vaccinating antigen. Nature protocols 4, 372-384.

Smith, S.A., Zhou, Y., Olivarez, N.P., Broadwater, A.H., de Silva, A.M., and Crowe, J.E.,

Jr. (2012). Persistence of circulating memory B cell clones with potential for dengue virus

disease enhancement for decades following infection. Journal of virology 86, 2665-2675.

Steinitz, M., Klein, G., Koskimies, S., and Makel, O. (1977). EB virus-induced B

lymphocyte cell lines producing specific antibody. Nature 269, 420-422.

Stiff, T., Walker, S.A., Cerosaletti, K., Goodarzi, A.A., Petermann, E., Concannon, P.,

O'Driscoll, M., and Jeggo, P.A. (2006). ATR-dependent phosphorylation and activation

of ATM in response to UV treatment or replication fork stalling. The EMBO journal 25,

5775-5782.

Stolz, A., Ertych, N., Kienitz, A., Vogel, C., Schneider, V., Fritz, B., Jacob, R., Dittmar, G.,

Weichert, W., Petersen, I., et al. (2010). The CHK2-BRCA1 tumour suppressor pathway

ensures chromosomal stability in human somatic cells. Nature cell biology 12, 492-499.

Stracker, T.H., Couto, S.S., Cordon-Cardo, C., Matos, T., and Petrini, J.H. (2008). Chk2

suppresses the oncogenic potential of DNA replication-associated DNA damage. Mol

Cell 31, 21-32.

Sugden, B., and Mark, W. (1977). Clonal transformation of adult human leukocytes by

Epstein-Barr virus. Journal of virology 23, 503-508.

Swaminathan, S., Tomkinson, B., and Kieff, E. (1991). Recombinant Epstein-Barr virus

with small RNA (EBER) genes deleted transforms lymphocytes and replicates in vitro.

Proc Natl Acad Sci U S A 88, 1546-1550.

Szekely, L., Pokrovskaja, K., Jiang, W.Q., Selivanova, G., Lowbeer, M., Ringertz, N.,

Wiman, K.G., and Klein, G. (1995). Resting B-cells, EBV-infected B-blasts and established

lymphoblastoid cell lines differ in their Rb, p53 and EBNA-5 expression patterns.

Oncogene 10, 1869-1874.

114

Takada, S., Kaneniwa, N., Tsuchida, N., and Koike, K. (1997). Cytoplasmic retention of

the p53 tumor suppressor gene product is observed in the hepatitis B virus X gene-

transfected cells. Oncogene 15, 1895-1901.

Tangye, S.G., Avery, D.T., and Hodgkin, P.D. (2003). A division-linked mechanism for

the rapid generation of Ig-secreting cells from human memory B cells. J Immunol 170,

261-269.

Tangye, S.G., Ferguson, A., Avery, D.T., Ma, C.S., and Hodgkin, P.D. (2002). Isotype

switching by human B cells is division-associated and regulated by cytokines. J

Immunol 169, 4298-4306.

Thorley-Lawson, D.A., and Allday, M.J. (2008). The curious case of the tumour virus: 50

years of Burkitt's lymphoma. Nature reviews Microbiology 6, 913-924.

Tian, C., Luskin, G.K., Dischert, K.M., Higginbotham, J.N., Shepherd, B.E., and Crowe,

J.E., Jr. (2008). Immunodominance of the VH1-46 antibody gene segment in the primary

repertoire of human rotavirus-specific B cells is reduced in the memory compartment

through somatic mutation of nondominant clones. J Immunol 180, 3279-3288.

Tierney, R.J., Kao, K.Y., Nagra, J.K., and Rickinson, A.B. (2011). Epstein-Barr virus

BamHI W repeat number limits EBNA2/EBNA-LP coexpression in newly infected B cells

and the efficiency of B-cell transformation: a rationale for the multiple W repeats in

wild-type virus strains. Journal of virology 85, 12362-12375.

Tort, F., Hernandez, S., Bea, S., Martinez, A., Esteller, M., Herman, J.G., Puig, X.,

Camacho, E., Sanchez, M., Nayach, I., et al. (2002). CHK2-decreased protein expression

and infrequent genetic alterations mainly occur in aggressive types of non-Hodgkin

lymphomas. Blood 100, 4602-4608.

Traggiai, E., Becker, S., Subbarao, K., Kolesnikova, L., Uematsu, Y., Gismondo, M.R.,

Murphy, B.R., Rappuoli, R., and Lanzavecchia, A. (2004). An efficient method to make

human monoclonal antibodies from memory B cells: potent neutralization of SARS

coronavirus. Nature medicine 10, 871-875.

115

Turner, M.L., Hawkins, E.D., and Hodgkin, P.D. (2008). Quantitative regulation of B cell

division destiny by signal strength. J Immunol 181, 374-382.

Verschuren, E.W., Jones, N., and Evan, G.I. (2004). The cell cycle and how it is steered by

Kaposi's sarcoma-associated herpesvirus cyclin. The Journal of general virology 85,

1347-1361.

Verschuren, E.W., Klefstrom, J., Evan, G.I., and Jones, N. (2002). The oncogenic potential

of Kaposi's sarcoma-associated herpesvirus cyclin is exposed by p53 loss in vitro and in

vivo. Cancer Cell 2, 229-241.

Wade, M., and Allday, M.J. (2000). Epstein-Barr virus suppresses a G(2)/M checkpoint

activated by genotoxins. Molecular and cellular biology 20, 1344-1360.

Wang, F., Kikutani, H., Tsang, S.F., Kishimoto, T., and Kieff, E. (1991). Epstein-Barr virus

nuclear protein 2 transactivates a cis-acting CD23 DNA element. Journal of virology 65,

4101-4106.

Wang, F., Tsang, S.F., Kurilla, M.G., Cohen, J.I., and Kieff, E. (1990). Epstein-Barr virus

nuclear antigen 2 transactivates latent membrane protein LMP1. Journal of virology 64,

3407-3416.

Wang, W.H., Hullinger, R.L., and Andrisani, O.M. (2008). Hepatitis B virus X protein via

the p38MAPK pathway induces E2F1 release and ATR kinase activation mediating p53

apoptosis. The Journal of biological chemistry 283, 25455-25467.

Wang, X.W., Forrester, K., Yeh, H., Feitelson, M.A., Gu, J.R., and Harris, C.C. (1994).

Hepatitis B virus X protein inhibits p53 sequence-specific DNA binding, transcriptional

activity, and association with transcription factor ERCC3. Proceedings of the National

Academy of Sciences of the United States of America 91, 2230-2234.

Wang, X.W., Gibson, M.K., Vermeulen, W., Yeh, H., Forrester, K., Sturzbecher, H.W.,

Hoeijmakers, J.H., and Harris, C.C. (1995). Abrogation of p53-induced apoptosis by the

hepatitis B virus X gene. Cancer research 55, 6012-6016.

116

Warter, L., Lee, C.Y., Thiagarajan, R., Grandadam, M., Lebecque, S., Lin, R.T., Bertin-

Maghit, S., Ng, L.F., Abastado, J.P., Despres, P., et al. (2011). Chikungunya virus

envelope-specific human monoclonal antibodies with broad neutralization potency. J

Immunol 186, 3258-3264.

Wei, X., Decker, J.M., Wang, S., Hui, H., Kappes, J.C., Wu, X., Salazar-Gonzalez, J.F.,

Salazar, M.G., Kilby, J.M., Saag, M.S., et al. (2003). Antibody neutralization and escape by

HIV-1. Nature 422, 307-312.

White, R.E., Ramer, P.C., Naresh, K.N., Meixlsperger, S., Pinaud, L., Rooney, C.,

Savoldo, B., Coutinho, R., Bodor, C., Gribben, J., et al. (2012). EBNA3B-deficient EBV

promotes B cell lymphomagenesis in humanized mice and is found in human tumors.

The Journal of clinical investigation 122, 1487-1502.

Wilson, P.C., and Andrews, S.F. (2012). Tools to therapeutically harness the human

antibody response. Nature reviews Immunology 12, 709-719.

Woisetschlaeger, M., Strominger, J.L., and Speck, S.H. (1989). Mutually exclusive use of

viral promoters in Epstein-Barr virus latently infected lymphocytes. Proceedings of the

National Academy of Sciences of the United States of America 86, 6498-6502.

Woisetschlaeger, M., Yandava, C.N., Furmanski, L.A., Strominger, J.L., and Speck, S.H.

(1990). Promoter switching in Epstein-Barr virus during the initial stages of infection of

B lymphocytes. Proc Natl Acad Sci U S A 87, 1725-1729.

Wu, X., Avni, D., Chiba, T., Yan, F., Zhao, Q., Lin, Y., Heng, H., and Livingston, D.

(2004). SV40 T antigen interacts with Nbs1 to disrupt DNA replication control. Genes

Dev 18, 1305-1316.

Yi, A.K., Chang, M., Peckham, D.W., Krieg, A.M., and Ashman, R.F. (1998). CpG

oligodeoxyribonucleotides rescue mature spleen B cells from spontaneous apoptosis and

promote cell cycle entry. J Immunol 160, 5898-5906.

117

Yi, F., Saha, A., Murakami, M., Kumar, P., Knight, J.S., Cai, Q., Choudhuri, T., and

Robertson, E.S. (2009). Epstein-Barr virus nuclear antigen 3C targets p53 and modulates

its transcriptional and apoptotic activities. Virology 388, 236-247.

Yu, X., Tsibane, T., McGraw, P.A., House, F.S., Keefer, C.J., Hicar, M.D., Tumpey, T.M.,

Pappas, C., Perrone, L.A., Martinez, O., et al. (2008). Neutralizing antibodies derived

from the B cells of 1918 influenza pandemic survivors. Nature 455, 532-536.

Zalvide, J., and DeCaprio, J.A. (1995). Role of pRb-related proteins in simian virus 40

large-T-antigen-mediated transformation. Molecular and cellular biology 15, 5800-5810.

Zhao, F., Hou, N.B., Yang, X.L., He, X., Liu, Y., Zhang, Y.H., Wei, C.W., Song, T., Li, L.,

Ma, Q.J., et al. (2008a). Ataxia telangiectasia-mutated-Rad3-related DNA damage

checkpoint signaling pathway triggered by hepatitis B virus infection. World J

Gastroenterol 14, 6163-6170.

Zhao, X., Madden-Fuentes, R.J., Lou, B.X., Pipas, J.M., Gerhardt, J., Rigell, C.J., and

Fanning, E. (2008b). Ataxia telangiectasia-mutated damage-signaling kinase- and

proteasome-dependent destruction of Mre11-Rad50-Nbs1 subunits in Simian virus 40-

infected primate cells. Journal of virology 82, 5316-5328.

Zheng, Z., Li, J., Sun, J., Song, T., Wei, C., Zhang, Y., Rao, G., Chen, G., Li, D., Yang, G., et

al. (2011). Inhibition of HBV replication by theophylline. Antiviral Res 89, 149-155.

Zimber-Strobl, U., Kremmer, E., Grasser, F., Marschall, G., Laux, G., and Bornkamm,

G.W. (1993). The Epstein-Barr virus nuclear antigen 2 interacts with an EBNA2

responsive cis-element of the terminal protein 1 gene promoter. The EMBO journal 12,

167-175.

Zindy, F., Williams, R.T., Baudino, T.A., Rehg, J.E., Skapek, S.X., Cleveland, J.L., Roussel,

M.F., and Sherr, C.J. (2003). Arf tumor suppressor promoter monitors latent oncogenic

signals in vivo. Proceedings of the National Academy of Sciences of the United States of

America 100, 15930-15935.

118

Biography

Pavel Nikitin was born in 1983 in Pavlodar, a city in the USSR that is now in

Kazakhstan. In 1999 Pavel was invited to attend a physics and math magnet high school

at Novosibirsk State University, Russia. In 2000 he went to Novosibirsk State University,

where he studied molecular biology and defended a “specialist” Diploma with honors in

2005. During his undergraduate school Pavel received a Potanin federal scholarship and

was awarded with the U.S. Civilian Research & Development Foundation grant to study

methods of inactivation of influenza virus.

In 2006 Pavel moved to the United States for an internship at Reproductive

Genetics Institute, a medical company in Chicago which specialized in pre-implantation

genetic diagnosis. Further, pursuing his interest in virology and genetics, Pavel went to

Duke’s Department of Molecular Genetics and Microbiology (MGM) for graduate

school. At Duke Pavel joined the recently formed laboratory of Dr. Micah Luftig to study

the host growth suppressive mechanisms that restrict Epstein-Barr virus-mediated

tumorigenesis. In Dr. Luftig’s group Pavel published one paper, two reviews and

contributed to one patent. During his time at Duke Pavel received a best oral

presentation award at the EBV Society meeting in Birmingham, UK in 2010 and was

awarded with several travel grants from the MGM Department and Duke’s Graduate

School.