University of Massachusetts Medical School eScholarship@UMMS

GSBS Dissertations and Theses Graduate School of Biomedical Sciences

2004-04-29

Pathways Linking Deregulated Proliferation to Apoptosis: a Dissertation

Harry A. Rogoff University of Massachusetts Medical School

Let us know how access to this document benefits ou.y Follow this and additional works at: https://escholarship.umassmed.edu/gsbs_diss

Part of the Amino Acids, Peptides, and Proteins Commons, Cells Commons, Eye Diseases Commons, and the Neoplasms Commons

Repository Citation Rogoff HA. (2004). Pathways Linking Deregulated Proliferation to Apoptosis: a Dissertation. GSBS Dissertations and Theses. https://doi.org/10.13028/18tf-2t22. Retrieved from https://escholarship.umassmed.edu/gsbs_diss/63

This material is brought to you by eScholarship@UMMS. It has been accepted for inclusion in GSBS Dissertations and Theses by an authorized administrator of eScholarship@UMMS. For more information, please contact [email protected]. A Dissertation Presented

Harr Andre Rogoff

Submitted to the Faculty of the

University of Massachusetts Graduate School of Biomedical Sciences, Worcester

In partial fulfillment of the requirements for the degree of

DOCTOR OF PHILOSOPHY

April 29, 2004

Program in Immunology and Virology COPYRIGHT NOTICE

Pars of this dissertation have appeared in the following publications:

Rogoff, H.A., Pickering, M. , Debatis, M. , Jones, S. , and T.F. Kowalik (2002). E2F1

Induces Phosphorylation of p53 that is Coincident with p53 Accumulation and Apoptosis.

Mol. Cell. BioI. 22: 5308-5318.

Rogoff, H.A. , Pickering, M. , Frame, F. , Debatis, M. , Sanchez, Y. , Jones , S. , and

F. Kowalik (2004). Apoptosis Associated with Deregulated E2F Activity is Dependent on E2F1 and AtmlbsllChk. Mol. Cel. BioI. 24: 2968-2977. Michelle Kelliher, Ph.D., Chair of Committee

Michael Brodsky, Ph.D., Member of Committee

Maria Zapp, Ph.D., Member of Committee

Stephen Doxsey, Ph.D., Member of Committee ACKNOWLEDGMENTS

I would like to thank my mentor Tim Kowalik for his guidence, support, and

friendship during my graduate career. Joining Tim s lab was possibly the best decision I

made during my graduate career and provided me the best possible place to grow as a

scientist. I would also like to thank the members of my committee, Michelle Kelliher

Stephen Doxsey, Maria Zapp, Michael Brodsky, and Karl Munger for their thoughtful

advice on my research projects. I am also grateful to the members of the Kowalik lab for

ther contributions. Fiona Frame has been a scientific colleague and, most of all, a good

friend. Fiona contributed Chapter V Figure 1B and Chapter V Figure 8. Michelle Debatis

contributed Chapter III Figure 5. Mary Pickering contributed Chapter III Figure 4 and

Chapter III Figure 12A. I would also like to thank Brad Stadler and former lab member

Jon Castilo for their helpful scientific advice, for making the Kowalik lab an enjoyable place to work, and for their friendships outside of science.

I would like to thank my parents, Martin and Linda Rogoff, for their support

during my years in graduate school. I would also like to thank the four legged members

of the Rogoff household; Andy, Bada, Marley, Teddy Bear, and Chloe for bringing love

and relaxation to my homelife. Most of all, thank you to my wife Tina, who has provided daily love and encouragment throughout my graduate career. ABSTRACT

Proper regulation of cellular proliferation is critical for normal development and

cancer prevention. Most, if not all, cancers contain mutations in the Rb/E2F pathway,

which controls cellular proliferation. Inactivation of the retinoblastoma protein (Rb) can

occur through Rb loss, mutation, or inactivation by cellular or viral oncoproteins leading

to unrestrained proliferation. This occurs primarily by de-repression and activation of the

E2F transcription factors, which promote the transition of cells from the G to S phase of

the cell cycle. In order to protect against loss of growth control, the p53 tumor suppressor

is able to induce programmed cell death, or apoptosis, in response to loss of proper Rb cell cycle regulation.

E2F! serves as the primary link between the Rb growth control pathway and the p53 apoptosis pathway. While the pathway(s) linking E2F! to p53 activation and apoptosis are unclear, it has been proposed that E2F! activates p53-dependent apoptosis

RF by transactivation ofpI leading to inhibition of Mdm2-promoted degradation ofp53.

We tested this hypothesis RF , and found that pI is not required for E2F!-induced

apoptosis. Instead , we find that expression of E2F I leads to covalent modifications of p53 that correlate with p53 activation and are required for apoptosis.

The observation that E2F! induces covalent modification ofp53 is consistent with the p53 modifications observed following DNA damage. We therefore hypothesized that

E2F! may be activating components of the DNA damage response to activate p53 and kill cells. Consistent with the DNA damage response, we find that E2FI-induced apoptosis is compromised in cells from patients with the related disorders ataxia

telangiectasia and Nijmegen breakage syndrome, lacking functional Atm and Nbsl gene

products, respectively. E2FI-induced apoptosis and p53 modification also requires the

human checkpoint kinase Chk2, another component of the DNA damage response. We

find that the commitment step in E2F I- induced apoptosis is the induction of Chk2.

Having found that E2FI requires DNA damage kinases to induce apoptosis, we

next examined events upstream of kinase activation. To this end, we observe relocalization of the DNA damage repair MRN complex (composed of Mrel I , NbsI , and

Rad50) to nuclear foci specifically following expression of E2F!. Expression of E2FI also induces relocalization of the DNA damage recognition proteins yH2AX and 53BPI

to nuclear foci, consistent with the location of these complexes observed following DNA

double strand breaks. As a consequence of activating some or all of these DNA damage

signaling proteins, expression of E2F I blocks cell cycle progression in diploid human

fibroblasts. The observed block in cell cycle progression is found to be , in part, due to activation of a p2I-dependent cell cycle checkpoint.

The E7 protein from the oncogenic human papilomavirus (HPV) is able to bind to and inactivate members of the Rb family. HPV infects quiescent, non-cycling cells that lack expression of DNA replication machinery that is essential for replication of the viral genome. By expression of the E7 protein, HPV is able to bypass normal Rb-mediated growth control and induce quiescent cells to enter S phase where the host cell DNA replication enzymes are present for viral replication. We find that expression of E7 can also result in apoptosis that is dependent specifically on E2F!. Additionally, E7-induced V11

apoptosis, like E2FI-induced apoptosis, requires Atm, NbsI , and Chk2. Expression of

, like that of E2FI , induces E2Fl-dependent covalent modification of p53 that correlates with apoptosis induction.

These findings demonstrate that deregulation of the Rb/E2F growth control pathway leads to activation of an apoptosis program with some similarity to the pathways activated by DNA damage. Our observations suggest that E2FI not only functions as sensor for deregulation of Rb, but may also play an important role in regulating cellular growth control in response to other oncogenic stimuli......

V11

T ABLE OF CONTENTS

COPYRIGHT NOTICE......

ACKNOWLEDGEMENTS......

ABSTRACT...... ,...... v

TABLE OF CONTENTS...... viii

LIST OF FIGURES...... ix

CHAPTER I. Introduction...... I

CHAPTER II. Materials and Methods......

CHAPTER III. E2FI induces phosphorylation of p53 that is coincident with

p53 accumulation and apoptosis...... 44

CHAPTER IV. Apoptosis associated with deregulated E2F activity is

dependent on E2F I and Atmlbs I /Chk ...... 71

CHAPTER V. E2F I induces Mre II foci formation and blocks cell cycle

pro gression in human fi bro blasts...... II 0

CHAPTER VI. Discussion ...... 138

CHAPTER VII. Appendix A...... 156

CHAPTER VIII. References...... 161 ......

LIST OF FIGURES

Figure 1. 1. Cyclin/Cdk regulation of the cell cycle ...... 5

Figure 1.2. Rb/E2F regulation of the cell cycle...... 7

Figure 1.3. Schematic representation of the E2F family...... 10

Figure 1.4. Models of Rb/E2F regulation ofE2F regulated promoters ...... 15

Figure 1.5 Model of p5 3 activation following cellular stress...... 26

Figure 1.6. Model of DNA double strand break signaling ...... 31 AR protein accumulation...... Figure 3. 1. E2FI induces p I9

Figure 3.2. E2FI induces apoptosis in the absence of pI 9ARF AR ...... Figure 3.3. E2FI induces p53-dependent apoptosis in the absence of pI 9

Figure 3.4. E2FI reqires pI9ARF to induce p53 accumulation...... AR and Mdm2 to increase p53 protein levels...... 53 Figure 3.5. E2FI requires pI9

Figure 3.6. Expression ofE2FI or E2F2 results in an increase in p53 protein

levels and levels ofthe phospho-serine 18 form of p53 in

wildtype MEFs......

Figure 3.7. E2FI specifically induces accumulation ofp53 and the phospho- AR ...... 57 serine 18 form ofp53 in cells lacking pI9

Figure 3.8. E2F2 does not induce apoptosis in wildtype MEFs ...... AR protein accumulation...... 60 Figure 3.9. E2F I and E2F2 induce p I9 AR to induce p53 protein and alter levels of the Figure 3. 10. E2F2 requires pI9

phospho-serine 15 form of p53 ...... "'''''' ......

Figure 3. 11. E2FI induces p53 protein and levels of the phospho-serine 15

and phospho-serine 20 forms of p5 3 in cells lacking p I9AR ...... 63 Figure 3.12. p53 phosphorylation correlates with E2FI- induced apoptosis ......

Figure 3. 13. Caffeine inhibits E2FI-induced apoptosis ...... 66 Figure 3.14. Model ofE2FI-induced apoptosis and p53 accumulation...... 69

Figure 4. 1. E2F I-induced apoptosis is reduced in human fibroblasts lacking

functional Atm or Nbsl ...... 74

Figure 4.2. E2FI-induced apoptosis is reduced in mouse embryo fibroblasts

lacking atm......

Figure 4.3. E2FI and E2F2 require Atm and Nbsl to efficiently induce

phosphorylation of p53...... 76

Figure 4.4. AdE2FI and AdE2F2 are expressed to similar levels in normal

, and NBS fibroblasts...... """'" ""'"'' ...... 78 Figure 4.5. Increased doses of AdE2F2 does not lead to apoptosis...... 79 Figure 4.6. Increased doses of AdE2F2 leads to increased levels of phospho-

serine 15 p53 but not phospho-serine 20 p53

Figure 4.7. Dominant negative Chk2, but not dominant negative ChkI reduces

E2F I-induced apoptosis ...... 82

Figure 4. 8. siRNA targeting ofChk inhibits E2FI-induced apoptosis ...... 83

Figure 4. 9. DN-Chk, but not DN-ChkI , inhibits phosphorylation ofp53 at

serine 20...... 85 Figure 4. 10. siRNA targeting ofChk inhibits E2FI-induced phosphorylation

of p53 at serine 20......

Figure 4.11. Expression ofE2FI , not E2F2, leads to increased Chk2 protein

levels in normal, AT, and NBS cells...... 87

Figure 4. 12. Expression of E2F Ie13 fails to induce apoptosis, increase Chk

protein levels or induce phosphorylation of p53 ...... """'"'''''' 88 Figure 4. 13. Expression ofE2FI or E2F21eads to an increase in the phospho-

serine 1981 form of Atm while leaving total Atm protein levels

unchanged...... 90

Figure 4. 14. E2FI , leads to an increase in , not E2F2 Chk2 mRNA levels but

not Atm mRNA levels...... 91

Figure 4. 15. E2FI and E2F2 cooperate with Chk in apoptosis induction...... 93

Figure 4. 16. Chk2 cooperates with E2F2 to induce phosphorylation of p53 at

serine 20...... 94

Figure 4. 17. HPV- I6 E7 induces apoptosis in human fibroblasts ...... 96

Figure 4. 18. HPV- I6 E7-induced apoptosis is reduced in cells lacking

functional Atm or Nbsl ......

Figure 4. 19. siRNA targeting ofE2FI or Chk inhibits E7-induced apoptosis ...... 98 Figure 4.20. siRNA knockdown of E2F2 and E2F3 ...... 100

Figure 4.21. HPV- I6 E7 requires E2FI but not E2F2 or E2F3 to induce

apoptosis ...... 101 X11

Figure 4.22. HPV- I6 E7 expression results in p53 accumulation and

modification, similar to those observed following expression of

E2F I ...... 102

Figure 4.23. HPV- I6 E7 expression results in an increase in Chk2 protein

levels and levels of the phospho-serine 1981 form of Atm

while leaving total Atm protein levels unchanged...... 104

Figure 4.24. HPV- I6 E7 requires E2FI and Chk to increase the levels of the

phospho-serine 20 form ofp53 ...... 105

Figure 4.25. HPV- I6 E7 requires E2FI to induce Chk2...... 106

Figure 4.26. Model ofp53 activation following deregulation ofE2FI ...... 108

Figure 5. 1. E2F I specifically induces Mre II foci formation...... 113

Figure 5.2. Expression ofE2FI , but not E2F3a, results in an increase in the

phospho-serine 20 form ofp53 ...... 114

Figure 5.3. E2FI , but not E2F3a, induces apoptosis in human fibroblasts ...... 115

Figure 5.4. E2FI fails to induce MreII foci in NBS cells ...... 116

Figure 5. 5. E2F I induces Mre II foci in AT cells...... 118

Figure 5.6. E2FI induces relocalization of DNA damage response proteins...... 119

Figure 5.7. Expression ofE2FI induces MreII foci that do not correlate with

BrdU incorporation...... 120

Figure 5. 8. E2FIl-283 induces MreII foci formation similar to full length E2FI ...... 121

Figure 5.9. E2FIe13 induces MreII foci formation similar to E2FI ...... 122

Figure 5. 10. E2FI , but not E2F3a, blocks cell cycle progression...... 124 ......

X11

Figure 5. 11. Expression of E2F I and E2F3a results in increased levels of the

phospho-serine 1981 form of Atm 125

Figure 5. 12. Roles for Nbsl and 53BPI in blocking cell cycle progression...... 127

Figure 5. 13. Expression of E2F I leads to accumulation ofp2I protein ...... 128

Figure 5. 14. Role for p2I in blocking cell cycle progression...... 129

Figure 5. 15. Role for p53 in blocking cell cycle progression and p2I

accumulation...... 130

Figure 5. 16. Reduced levels of 53BPI ofp2Ileads to enhanced E2FI-induced

apoptosis ...... 133

Figure 5. 17. Model of E2F 1- induced apoptosis and growth arrest...... 13 5

Figure 6. 1. Model ofE2F signaling network...... 149

Figure AI. E2F I induces apoptosis...... 157

Figure A2. siChk2 inhibits E2F I-induced apoptosis ...... 158

Figure A3. DNChkI and DNChk do not effect E2FI expression...... 159

Figure A4. Specific requirement for E2FI in HPV- I6 E7 induced apoptosis...... 160 CHAPTER I

INTRODUCTION A. Cell Cycle

The cell cycle is a tightly controlled sequence of events in which genetic material

is precisely copied so that each daughter cell will receive an identical copy of the genetic

material following cell division. The eukaryotic cell cycle is divided into mitosis (M

phase), the phase in which chromosomes are segregated and cell division occurs, and an interphase period that occurs between subsequent mitosis. Interphase consists of a gap

phase that occurs following cell division and prior to DNA replication known as the G

phase. DNA is then replicated during S phase, and is followed by another gap phase, G

prior to cells re-entering mitosis. Many cells in the body reach a state of terminal differentiation where they will exit from the cell cycle following mitosis and enter a

resting, or G , state (Figure 1. 1). Malfunctions during the cell cycle, including errors in

DNA replication, spindle formation, cytokinesis, or DNA damage can result in activation

of a cell cycle checkpont to allow for repair to take place so that mutations are not passed to daughter cells (365).

B. Cell eycle regulation

1. Cyetins and eyclin dependent kinases

Precise control of the cell cycle is necessary for cells to grow and divide at the proper time and to ensure passage of properly replicated DNA to daughter cells. Multiple levels of regulation are present to ensure that the cell cycle will not advance to the next phase if the cellular or environmental conditions are not conducive for cell cycle progression. Progression from one phase of the cell cycle to the next is achieved mainly by the action of cellular cyclins and cyclin-dependent kinases (Cdk) (Figure 1.2). Cdk are a family of serine-threonine protein kinases that control progression of the cell cycle by phosphorylating target proteins at specific times (272, 318). Cdk protein levels remain constant during the cell cycle, so their activity is regulated by expression of the cyclin family of proteins (281 , 318, 319). Timed expression and rapid ubiquitin mediated proteolysis limits cyclin expression to specific intervals of the cell cycle, limiting the activity of the Cdk to certain phases of the cell cycle (102, 126 , 281 , 282, 330).

Beginning in G , the cyclin D family (consisting of cyclin DI , D2, and D3) forms a complex with Cdk4 or Cdk6. The cyclin D-Cdk4/6 complex begins the cells entry into S phase primarily by phosphorylation of the retinoblastoma (Rb) family of proteins (364).

This is followed by cyclin E-Cdk2 activity and additional Rb phosphorylation pushing the cell from G into S phase (298). The complex of cyclin A and Cdk2 is required for cells to progress through S phase (121 , 422). In the G and M phases of the cell cycle cyclin A forms a complex with CdkI , and this complex is required for transition from to M (446). Mitosis is then further regulated by activity of the cyclin B-Cdki complex

(11 , 200) (Figure 1. 1).

2. Cdk inhibitors

Additional layers of Cdk regulation take the form of cyclin dependent kinase inhibitors and protein localization. Cyclin dependent kinase inhibitors play important roles in coordinating proliferation during normal development and differentiation, as well as during cellular stress (63, 223). Cyclin dependent kinase inhibitors can be divided into INK4a the INK4 family and the Cip/Kip family (367). The INK4 family, including pI6 target Cdk4 and Cdk6 and prevent their association with cyclin D (45). Without cyclin D association, Cdk4/6 remain inactive and the cell cycle is unable to progress through G

(325, 360). The Cip/Kip family of cyclin dependent kinase inhibitors, including p2I and p27, are able to inactivate multiple cyclin-Cdk complexes and are able to block cell cycle progression in G and, to a lesser extent, Sand M phases of the cell cycle (139, 150 221

320). Unlike pI6, p2I and p27 promote a stable association between cyclin D and their

Cdk partners (215). Following cellular growth signals, increased levels of cyclin D-Cdk4 complexes will titrate p2I and p27 from cyclin E-Cdk2 complexes allowing for progression through G I (32). The localization of both Cdk activators and inhibitors also contributes to proper cell cycle progression. For example, to prevent inappropriate mitosis the cyclin B-Cdki inhibitor Wee I is kept in the nucleus, while the cyclin B-Cdki activator Cdc25 is sequestered in the cytoplasm prior to M phase (144, 237, 312, 451).

3. The retinoblastoma protein family

In the late G phase of the cell cycle, cells must pass through the G restriction point in order to enter into S phase and begin DNA replication (307). Passing the G restriction point means that the cell can continue through the remainder of the cell cycle

and back into G without additional growth factor requirements (307, 363). The G restriction point is controlled by the retinoblastoma (Rb) family of proteins. The Rb

family consists ofthree related genes;pRb (known as Rb), pI07, andp130 (387). Rb was

originally identified due to homozygous loss of the Rb allele that predisposes children to Figure 1.1. Cyetin/Cdk regulation of the eell eycle. Eukaryotic cells replicate DNA during S phase and divide during mitosis (M phase).

Separating the S and M phases of the cell cycle are two gap phases, G and G2. Activity of cellular cyclins and cyclin dependent kinases (Cdk) regulate progression from one phase of the cell cycle to the next. Sites of activity of Cyclin-Cdk complexes during the cell cycle are depicted. development of retinoblastoma, a malignant tumor of the retina (116 , 118 , 203 , 224). pI07 and p130 have been identified more recently, and have structural and functional similarities to Rb (59, 105, 120, 135 355 414 456 461). Together, Rb, pI07, and p130 are known as the pocket proteins due to a large, conserved protein-protein interaction domain (58, 220). Studies of Rb family knockout mouse models and cells from those mice reveal some functional overlap between family members in regards to development

(62, 222) and cell cycle regulation (67, 345). Rb family members also play important roles in terminal differentiation (67), however, the role of individual Rb family members in differentiation is unclear and may depend on the experimental system analyzed (44

293 334 352 453).

The Rb family fuction mainly as negative regulators of cell cycle control. During

, the Rb family are in an active, unphosphorylated state (429). In this state, the pocket proteins are able to interact with transcription factors, such as the E2F family, that control

the entrance of the cell into S phase (4, 429). While bound to Rb, the E2F family are

unable to transactivate the expression of genes required for S phase (4, 429). A major

function of the Rb family of proteins is to repress transcription from E2F regulated

promoters (5 , 89 , 109 , 114). At the restriction point, the Rb family becomes

sequentially phosphorylated by the cellular cyclin-Cdk complexes (225). D type cyclins

complexed to either Cdk4 or Cdk6 phosphorylate Rb during Gl (104, 193). This is

followed by cyclin E-Cdk2 phosphorylation leaving Rb in a hyperphosphorylated state

(154, 206, 230). In this state, the Rb family is unable to bind to and inhibit activity of the

S phase promoting E2F family members (55) (Figure 1.2). ..

Eukaryotic G Cell Cycle R 1

Figure 1.2. Rb/E2F regulation of the cell cycle.

Rb/E2F complexes regulate entry of cells from the G to S phase of the cell cycle. Prior

to the restriction point (R), Rb/E2F complexes repress E2F regulated promoters

preventing entry into S phase. Phosphorylation of Rb at the restriction point by

cyclin/Cdk complexes promotes dissociation of Rb and E2F, allowing expression of E2F regulated genes. 4. The E2F family c-- The Rb family controls cell cycle progression mainly through their ability to

interact with the E2F family of transcription factors. E2F was first identified as a cellular

transcription factor capable of binding to two consensus E2F "TTTCGCGC" sites within

the adenovirus E2 promoter (208, 209, 286). Human E2FI was later cloned by screening

cDNA libraries for proteins that interact with the pocket domain of Rb (148, 186, 362).

Many cellular genes regulated by the E2F family have been identified including genes

involved in controlling cell cycle regulation, DNA synthesis, and DNA replication. These

genes include cyclin A, cyclin E, cyclin DI , dihydrofolate reductase (DHFR), thmidine

kinase (TK), DNA polymerase a, and ORCI (382). E2F homo logs have been identified

in mouse (232), Drosophila melanogaster (297), yeast (249, 418), Xenopus laevis (315

391), and plants (326, 359).

To date, seven members of the human E2F family have been described (69, 81

404) (Figure 1.3). The E2F family can be subdivided into categories based on their ability

to interact with the Rb family and on their transcriptional activity. E2FI , E2F2, and

E2F3a are potent activators of transcription and interact mainly with Rb (3 , 148 , 149

174, 183 , 226, 227, 324). E2F4, and E2F5 are transcriptional repressors, and interact

primarily with the Rb family members pI07 and p130 (26, 153, 171 270 347 414). The

functions of the remaining members of the E2F family remain somewhat unclear. E2F3b

encoded by an alternate transcript from the E2F3 locus, interacts with Rb and may

function as a transcriptional repressor in quiescent cells (227). E2F6 shares little

homology with the other E2F family members outside of the DNA binding domain, does not interact with any of the Rb family members, lacks a transactivation domain, and is reported to repress transcription by interaction with polycomb proteins (275 , 403).

Likewise, E2F7 is unable to interact with Rb proteins, lacks a transactivation domain, and appears to act as a transcriptional repressor (69 81).

In cells , E2F proteins act in conjunction with their heterodimeric binding partners the DP family of proteins. The DP family consists of DPI and DP2. DPI was first identified as a ubiquitously expressed binding partner for E2FI that enhanced the transactivation ability of E2FI (18 , 125 , 149, 213). DP2 shares high homology to DPI and is also acts as an E2F binding partner that can enhance the transactivation ability of

E2F family members (299, 435 , 458). Unlike DPI , DP2 is expressed as tissue specific splice variants (72, 299, 339 458). In vitro, all combinations ofE2F-DP heterodimers can form and recognize similar E2F consensus binding sites (38, 226, 458). The large number of E2F-DP combinations possible in cells suggests that different heterodimers may differentially regulate genes. While some evidence of this exists (398), the role of the different E2F-DP heterodimers in regulating gene expression remains unclear.

5. Importanee of the E2F family in growth control

The important and unique roles for the E2F proteins have become partially realized through genetic studies in mice. Cells from mice lacking E2F I display limited cell cycle defects except for a prolonged Go phase, suggesting that E2FI may have a role in timing S phase entry from quiescence (113 , 425 , 449), while cells lacking E2F2 show little defect in proliferation (228). Cells from mice lacking E2F3 exhibit a delayed cell j .""""" '"...': , ...,... ' ,. ~~~- .'''' ' _.. .. "

Cyclin A Binding Dimerization Pocket Protein Binding

DNA Binding Marked Box Transactivation

''''''M'' .W"W E2F1 I"yJM"%'mm

E2F2 :''r- ?:''r r*-*-" M"' E2F3a F1l7k;;;IV..I'

"c""F'?- E2F3b r"' O=I I..

E2F4 r"7;57 -0=

f1r' E2F5 ISHZ

E2F6 I'YmHA, ?!,,"'D

E2F7

Figure 1.3. Sehematie representation of the E2F family.

The E2F family consists of seven members, distinguised by their transactivational ability and preference for association with Rb family members. E2FI , E2F2, and E2F3a are potent activators of transcription, and interact mainly with Rb. E2F4 and E2F5 are transcriptional repressors, and interact preferentially with pI 07 and p 130. E2F6 and E2F7 lack Rb family binding and transactivation function, and may function as transcriptional repressors. Shaded regions represent conserved domains. cycle , failure to upregulate many E2F responSIve genes II response to mitogen

stimulation, centriole duplication, and premature centriole separation (167 , 343).

However, cells from mice lacking all three activator E2Fs show severe proliferative

defects and fail to induce E2F target genes that are critical for the transition from G to S

(438). Cells lacking E2F4 or E2F5, or both, do not display the cell cycle defects or loss of

gene expression associated with loss ofE2F3 (166 331). However, loss ofE2F4 or E2F5

does result in a failure to properly exit from the cell cycle in response to growth

INK4a (37 inhibitory signals such as expression ofpI6 , 166 331).

E2F family members also playa role in the regulation of differentiation, or exit

from the cell cycle. Repression of E2F 1 regulated promoters is essential to promote and

maintain exit from the cell cycle in various cell types (82, 424). Mutation of Rb in some

cell types can lead to re-entry into the cell cycle and uncontrolled proliferation and an

increase in apoptosis (60 , 179 , 219). Repression of E2FI in some cell types is accomplished by the activity of the C/EBPa transcription factor (322). Active repression of E2F regulated genes by a complex composed of p130 and E2F4 or E2F5 is also essential for maintaining the differentiated state of some cell types (28, 274, 322). Taken together, E2F family members can have different roles depending on the cells state of differentiation.

6. Rb/E2F regulation of cell cycle progression

Cellular growth control by the retinoblastoma protein family occurs primarily through Rb-mediated repression of E2F regulated promoters and active transcriptional . .

repression by Rb/E2F complexes. While it is unclear which Rb/E2F function is most

important in vivo both mechanisms have been shown to occur in vitro. Rb inhibition of

E2F-mediated transactivation occurs by virtue of Rb binding to the E2F transactivation

domain and inhibiting transcriptional activation of E2F responsive promoters (Figure 1.4

A) (114, 137, 147 341). The importance ofE2F repression by Rb is demonstrated by the

observations that ectopic expression of E2FI , E2F2, or E2F3 is sufficient to drive

quiescent cells into S phase (76, 183 211).

Rb can also actively repress E2F-mediated transcriptional activation by

recruitment of chromatin remodeling enzymes, including HDACs, SWI/SNF complexes

and methyl transferases (136, 137 290 417 443). HDACs, or histone deacetylases, are a

family of enzymes involved with the removal of acetyl groups from histone tails that

promotes chromatin condensation (201 207) and have been shown to be associated with

transcriptional repressor complexes (8, 130, 140, 141 , 145). HDACI and HDAC2 contain

LxCxE motifs, allowing them to bind to Rb at the LxCxE binding site, while HDAC3

lacks the LxCxE motif and binds to Rb at another location (34, 243 , 247). E2F family

members bind to Rb at a site distinct from the LxCxE motif and HDAC3 binding site

allowing complexes containing HDACI , HDAC2, or HDAC3 , Rb, and E2F to form.

Interaction between Rb and HDACs is at least partially responsible for Rb-mediated

repression of E2F responsive promoters (34, 216 , 243 , 247 , 457). Additionally,

recruitment of HDACs to Rb/E2F repressor complexes serves to inhibit the function of

the histone acetyl transferases (HAT) p300/CBP that is recruited to promoters by

interaction with E2FI (405). HATs function by adding acetyl groups to histone tails relaxing chromatin structure, and allowing transcriptional machinery access to promoters

(201). By recruitment ofHDACs to corepressor complexes, Rb is able to actively repress

E2F -mediated transcription by altering chromatin structure (Figure 1.4 B). Repressor complexes containing pI 07 and p 13 0 have not been characterized as thoroughly as Rb repressor complexes, but studies have shown association of pI 07 and p 130 with HDAC proteins (112, 169, 194 386).

Active repression by Rb also occurs by recruitment of A TP-dependent SWI/SNF complexes. Rb recruits SWI/SNF complexes by interaction with the ATPase components of the complexes, Brgi and Brm (87, 375, 389). Similar to HDAC complexes, repressor complexes containing Brm, Rb, and E2F can form at promoters containing E2F binding sites (406). Dominant-negative ATPase mutant Brgi or Brm can block Rb-mediated growth suppression (87, 388) and expression of Brgi in cells lacking BrgI and Brm results in a growth arrest that is dependent on interaction with Rb (87). Additionally, expression of p I61NK4a induces a Rb-dependent growth arrest that requires functional

Brgi (434 457).

The formation of multiple Rb/E2F repressor complexes during the G phase of the cell cycle prevents expression of S phase promoting genes and thus prevents entry into S phase. However, different Rb/E2F complexes also form at promoters during the different phases of the cell cycle. Most cell cycle regulated promoters bind both E2FI 3/Rb complexes and E2F4 5/pI07 or p130 complexes. The use of chromatin immunoprecipitation (ChIP) allows analysis of promoter occupancy during distinct phases of the cell cycle and has provided a picture of the dynamic regulation by Rb/E2F complexes (329, 395 , 431). Regulation ofthe b-myb gene is an example of expression by

derepression. The b-myb promoter is occupied by an E2F4/pI07 repression complex during Gl. During S phase, the E2F4/pI07 complex dissociates and E2F4 no longer binds

, 431). to the b-myb promoter allowing expression of b-myb during S phase (395

Regulation of the dihydrofolate reductase (DHFR) promoter is an example of

transcriptional activation. The DHFR promoter is occupied in Go and by an

E2F4/pI07 repression complex. As cells progress from G into S phase, the complex is replaced by an E2FI/Rb complex, and following hyperphosphorylation ofRb, free E2FI

allowing expression of DHFR in early S phase (Figure 1.4 C). This complex is quickly replaced in late S phase with an E2F4/pI07 repression complex (431). Consistent with their proposed roles as activator E2Fs, E2FI , 2, and 3 occupy many E2F responsive promoters during S phase, while E2F 4 is the predominant E2F occupying these same promoters during Go and G . During S phase, some of the promoters tested also showed preference to binding by E2FI , E2F2, or E2F3 , such as DHFR , TK (thymidine kinase),

and Cdc6 while other promoters such aspI07 and Cdc2 showed no preference for E2F binding (329, 395 431).

7. Rb family independent regulation of E2F

In addition to regulation by the Rb family of proteins, the E2F family is also subject to Rb independent regulation. The majority of information regarding E2F regulation has come from studies of the E2FI family member. The association of E2FI with Rb during Gl serves to protect E2FI from degradation. When Rb becomes Transcriptional Repression

Active Transcriptional Repression

Transcriptional Activation

Figure 1.4. Models of Rb/E2F regulation of E2F regulated promoters.

(A) Binding of Rb/E2F complexes to E2F regulated promoters represses E2Fs transactivation function. (B) By recruiting chromatin remodeling enzymes such as

HDACs or SWI/SNF complexes to promoter regions, Rb is able to actively repress transcription. (C) Dissociation of Rb from E2F allows expression from E2F regulated promoters. phosphorylated as cells enter into S phase, E2FI becomes acetylated by p300/CBP enhancing E2FI stability and DNA binding activity (253). As cells proceed into mid S phase, the cyclin A/Cdk2 complex physically associates with E2FI at a conserved N- terminal domain (212 310 444). Subsequent phosphorylation of both E2FI and the DP heterodimeric binding partner by the cyclin A/Cdk2 complex at the S to G phase transition reduces E2FI DNA binding ability and promotes E2FI degradation by the proteasome (131 , 252).

Because of the ability of E2FI to drive cells into the cell cycle, E2FI protein levels must be very tightly controlled. When Rb dissociates with E2F I at the G to S transition, E2FI protein begins to be degraded by the ubiquitin-proteasome pathway (42). AR (mouse pI9 The degradation process is initiated by the human pI4 ) protein that

SKP2 binds to the C-terminus of E2FI and recruits the p45 ubiquitin protein ligase (25

251). This leads to poly-ubiqutination of E2FI and degradation by the proteasome.

Additionally, in response to some stimuli, E2FI can be phosphorylated by the ataxia telangiectasia mutated (Atm) kinase and the human checkpoint kinase, Chk2 (235 , 383).

These phosphorylation events stabilize E2FI protein, possibly by interfering with p45 SKP2 binding, and can also alter E2FI DNA binding specificity (383).

8. E2F regulated genes

The role for E2F in regulating genes involved in the G to S phase transition, S phase progression, and DNA replication has been confirmed by numerous micro array studies. Genes induced by E2F expression include genes involved in S phase promotion such as cyclin E genes required for the synthesis of DNA such as ribonucleotide reductase, thymidine kinase and DNA polymerase , and genes involved in assembly of pre-recognition complexes at origins of replication including those encoding Orc proteins, Cdc6, and Mcm proteins (173 187 244 279 332 380). Microarray analysis has also identified additional classes of genes that are regulated by E2F proteins. These include genes involved in DNA damage recognition, DNA repair, and apoptosis (279

332). The findings that E2F proteins regulate genes encoding DNA damage recognition and repair enzymes is not surprising given that DNA replication is an error prone process.

Additionally, in response to DNA damaging agents, E2FI is upregulated, providing a mechanism for increased expression of DNA damage recognition and repair enzymes

(279). E2FI can also downregulate expression of genes encoding anti-apoptotic factors

(314), and upregulate expression or pro-apoptotic factors (279). The ability of E2F proteins to regulate such a diverse set of genes suggests that E2F function is crucial to many biological pathways, although it remains unclear as to how E2F regulation may

segregate by biological pathway.

C. Deregulation of the cell cycle

1. Deregulation of the eell cycle by small DNA tumor viruses

Much of what is known regarding cell cycle regulation originally came about

through studies of the small DNA tumor viruses. Small DNA tumor viruses, including

adenoviruses, human papillomaviruses (HPV), and polyomaviruses such as SV 40 are

dependent on host cell DNA replication machinery for replication of viral genomes. However, the small DNA tumor viruses replicate in terminally differentiated, non- dividing cells that lack the required DNA replication machinery. In order to replicate, a virus must push cells into S phase where host cell DNA replication machinery is available for replication of viral genomes (39 , 78).

The induction of S phase by the small DNA tumor viruses is accomplished by virally encoded proteins that inactivate members of the Rb family of proteins. Each of the small DNA tumor viruses accomplishes this through a conserved LxCxE motif that binds and inactivates Rb family members. The adenovirus EIA protein contains an LxCxE motif and binds with high affnity to all three members of the Rb family of proteins (95

13 8 , 231 , 260). By binding to Rb family members, E I A disrupts the association of

Rb/E2F complexes allowing expression of E2F responsive promoters, resulting in induction of S phase (12, 16, 19, 95 , 138 , 163 , 231 , 260, 328, 455). HPV encodes the

LxCxE motif containing E7 protein that also binds to and inactivates Rb family members resulting in increased E2F activity and deregulation of the cell cycle (54, 80, 302, 436).

Expression of E7 also results in degradation of Rb family members by the proteasome providing another mechanism to bypass cellular growth control (27 , 33 , 128). The polyomavirus SV 40 encodes the large T antigen that is also capable of binding to Rb family members and blocking Rb-mediated G growth arest (74, 90, 103 432, 454).

2. Cell eycle deregulation and caneer

Long before the mechanisms of cell cycle deregulation by the small DNA tumor viruses were known, these viruses were found to be associated with tumor formation. Adenovirus was the first human virus identified to induce tumors in an animal model

(402). HPV is capable of inducing tumors in humans, and may be the causative agents in most cervical cancers (31 , 88, 464, 465). SV 40 was first isolated from monkey kidney cells (392) and is capable of transforming some rodent cell types (92, 122- 124, 399 400).

There is also some evidence that SV 40 may be associated with some human cancers

(202). In all cases, the small DNA tumor viruses require the function of the oncogenes

EIA, E7 or large T for efficient transformation (79, 93 , 94, 180, 313 , 344).

In addition to virus-associated transformation, cancer often results from cellular mutations that result in loss of normal growth control and lead to unrestrained proliferation. The high frequency of cell cycle regulators mutated in human cancers demonstrates the importance of proper cellular growth control for the prevention of cancer (363). The cell cycle mutations most often observed in human cancers include overexpression of cellular cyclins, loss of cyclin dependent kinase inhibitors, and loss of

Rb either by mutation, deletion, or inactivation (262).

Loss or mutation of cellular Cdks although rare, has been associated with some

tumors. Cdk4 amplification and Cdk4 and Cdk6 mutations have been identified in

melanomas, sarcomas, and gliomas (91 , 433). CdkI and Cdk2 overexpression has also been reported in a subset of colon cancers (198 , 447). Occuring more frequently in human tumors is the overexpression or deregulation of cellular cyclins. Overexpression

of cyclin DI often occurs as a result of a gene translocation that puts the cyclin DI gene under the control of the gene encoding the immunoglobulin heavy chain (133, 278, 430).

Cyclin D2, cyclin D3, cyclin A and cyclin E have all been reported to be amplified or overexpressed in some tumors , and elevated levels of cyclin E expression may correlate with shorter survival (85 170 197 218 357).

In addition to alterations in expression of Cdks and cyclins, mutations in both Cdk activators and inhibitors occur in human cancer. Deregulation or overexpression of the

Cdk activating enzme Cdc25 results in deregulated activation of cyclin-Cdk complexes entry into S phase, and is associated with tumor formation. Cdc25A and Cdc25B are activated by c-myc, an oncogene that is frequently mutated in tumors, and are often overexpressed in breast cancer (119, 291). Loss or mutation of the cyclin dependent

kinase inhibitorspI6 p27 andp2Ileads to unscheduled inactivation ofRb. Mutations in cyclin dependent kinase inhibitors often correlates with tumor aggressiveness and serves as a marker for poor patient prognosis (100, 188, 369).

The Rb family of tumor suppressors is one of the most frequently studied cell cycle regulators due to its involvement in viral and mutation-induced tumors. As discussed earlier, the Rb protein is the target of many viral oncoproteins that deregulate the cell cycle in order to provide a conducive environment for viral replication. In human tumors, the Rb protein is mutated in retinoblastoma, lung cancer, and acute lymphoblastic leukemia (162, 410). Moreover, it is estimated that greater than 90% of human tumors contain mutations in the Rb growth control pathway (133). While mutations in the Rb growth control pathway are common in human tumors, mutations in E2F family members

are rarely found in human tumors. E2F expression has been reported to be either reduced or amplified in different tumor types (99, 106 , 110 , 157, 177). The lack of direct E2F family mutations in human tumors suggests that cells must maintain a certain level of r::, E2F proteins in order to properly grow and avoid apoptosis (discussed below).

D. Apoptosis

1. Overview of apoptosis

Cell death has long been observed as a function of normal development (248).

The observation that cells appeared to be destined to die at specific times during

development was proposed in 1965 by Lockshin and Wiliams and termed programed cell

death (239). The term apoptosis, Greek for falling of leaves, was coined in 1972 to

describe the morphological changes associated with programmed cell death (195 , 196).

Apoptosis is characterized morphologically by cellular shrinking, condensation of the

chromatin, nucleosomal DNA fragmentation, convolution of the plasma membrane

known as blebbing, and formation of the dying cells into apoptotic bodies that are

surounded by an intact plasma membrane (415 , 442). Since the initial characterization of

the phenomenon of apoptosis in the I970s, there has been extensive work done to

determine its biological importance and underlying molecular mechanisms. The apoptosis

program has been found to be vital for normal development, for maintaining tissue

homeostasis, and for proper function of the immune system. Deregulation of the

apoptosis program can lead to disastrous consequences such as neurodegeneration

autoimmunity, and in its absence, cancer.

2. The p53 tumor suppressor The p53 protein plays an essential role in mediating both cellular growth arrest

and apoptosis in response to diverse environmental stimuli. The importance of p53 in

maintaining genomic stability is suggested by findings that greater than 50% of human

tumors contain mutations in the p53 gene (159). Activation of p53 was first observed in

cells exposed to ultraviolet radiation (250). It was then demonstrated that p53 was

essential for mediating a cellular growth arrest in response to irradiation or DNA-

damaging agents (192). The role of p53 in apoptosis induction was discovered by the

introduction of p53 into a p53 deficient cell line (452). The ability of p53 to induce apoptosis was later shown to correlate with the ability of p53 to act as a tumor suppressor

(393). Depending on the cellular context or duration or extent of DNA damage, p53 can

induce a cellular growth arrest, primarily through transactivation of the p2I promoter, to

allow for repair of damaged DNA or induce apoptosis (168 , 192, 229). Consequently, p53 has become known as a sensor of genomic stability due to its ability to respond to

damaged DNA, hypoxia, nucleotide depletion, viral oncogene expression, and other

genotoxic stresses (323).

3. Apoptotic targets of p53

The apoptosis program has been divided into two major pathways, the intrinsic pathway and the extrinsic pathway (2). Much of the data involving p53-mediated apoptosis have linked p53 to activation of the intrinsic, or cellular stress pathway. In the apoptosis pathway, the p53 protein functions as a sequence specific transcription factor

(204). Activation of the apoptosis program by p53 has been linked to its transactivation function (15 , 57 , 181 , 349). Perhaps the most important proapoptotic transcriptional targets of p53 in the intrinsic pathway are members of the Bcl-2 family. Transactivation by p53 of the Bc1- 2 family members Bax, Bid, Puma and Noxa ultimately leads to permeablization of the outer mitochondrial membrane, release of proapoptotic factors from the damaged mitochondria, activation of cysteine proteases known as caspases, and apoptosis (2, 214, 269, 284, 295, 350, 368). p53 also transactivates components of the

apoptotic machinery, including ApafI and the effector caspase, caspase-6. Apafl helps to initiate the caspase cascade by activating caspase-9 (190, 276, 336), while an increase in caspase-6 protein levels may sensitize cells to release of cytochrome-c from mitochondria

(245).

The extrinsic apoptosis pathway involves activation of cell surface death receptors by their appropriate ligands, resulting in caspase activation and apoptosis (14). The contribution of p53 to apoptosis resulting from activation of the extrinsic pathway remains unclear. p53 may elevate sensitivity to death receptor signaling by transactivation of genes encoding cell surface receptors or ligands. Direct transcriptional

targets of p53 include Fas and Fas ligand and studies using mutant p53 constructs have proposed a role for p53 in upregulation of these receptors as a means of promoting apoptosis in tumor cells (129, 280 301).

In addition to the two major apoptotic pathways, p53 is able to induce many apoptotic genes that are not directly associated with these pathways. These genes include p53AipI which is able to disrupt mitochondrial function and induce apoptosis in some tumor cell lines (257 , 296). PTEN is another transcriptional target of p53 that plays an /-

important role in p53-mediated apoptosis (378). PTEN does not act as a direct inducer of

apoptosis, but instead acts to inhibit the antiapoptotic protein Akt. Akt is a kinase that

inhibits cell death by phosphorylating numerous targets. PTEN functions as

phosphatase, preventing activation of Akt and the subsequent cellular survival signals

(420).

4. p53 aetivation

In response to cellular stressors , p53 is covalently modified, including

phosphorylation at numerous N- and C- terminal serine residues and acetylation on C-

terminal lysine residues (10). While it remains unclear as to which posttranslational

modifications are required for p53 activation under specific cellular stressors, these

modifications play an important role in mediating p53 function. p53 modifications have

been shown to increase transactivation function, increase protein stability, alter protein-

protein interactions, and even change promoter specificity (10) (Figure 1.5).

Several cellular kinases have been identified that play critical roles in the

activation of p53 following DNA damage, the best studied of the stress responses that

activates p53. These kinases include Atm, the kinase mutated in ataxia telangiectasia

(AT), Atr, the Atm and Rad3 related kinase, and their downstream kinase substrates

checkpoint kinase I (ChkI) and checkpoint kinase 2 (Chk) (1, 372). The role of Atr in regulating the p53 response to DNA damage is not well understood due in part to the

early embryonic lethality of atr mouse embryos (36, 71). However, it has been proposed that Atr is activated in response to certain types of DNA damage and can phosphorylate p53 on serine 15 (43 134 199 217). Atr is also able to phosphorylate and activate ChkI (132, 146 289), which can subsequently phosphorylate the N-terminus of p53 (370).

The role of Atm in activating p53 following DNA damage is better understood. In response to y-irradiation or genotoxic drugs that induce DNA double strand breaks, Atm is activated and can directly phosphorylate p53 at serine 15 (191). In cells from AT patients, there is a delay in activation of p53 following y-irradiation (374). In addition to directly phosphorylating p53 , Atm can phosphorylate and activate the human checkpoint kinase Chk2 (35 , 52 , 258, 259, 264). Chk2 is able to further phosphorylate p53 at additional N-terminal serine residues including serine 15 and serine 20, causing increased p53 stability and transcriptional activation (53 , 155 370). The importance ofChk in this pathway is demonstrated in dominant negative Chk2 expressing cells (53) and Chk2 deficient mice, which exhibit a defect in apoptosis and a decrease in p53 stabilization in response to y-irradiation (396).

5. Deregulated protiferation and p53

Inactivation of Rb by cellular or viral oncoproteins or loss of Rb wil stimulate cells to bypass GO/Gl and enter S phase (96 , 101 , 273 , 286). Ectopic expression of viral oncogenes, such as the high risk HPV - 16 E7 protein, can also lead to the induction of apoptosis (164, 304). The ability of E7 to induce apoptosis is dependent on its ability to interact with Rb (304). E7-induced apoptosis can be inhibited by expression of the viral 48-

Figure 1.5. Model of p53 activation following cellular stress.

The Atm and Atr kinases are activated by certain forms of DNA damage or replicative

stress. Atm and Atr can directly phosphorylate p53 at serine 15 as well as phosphorylate

and activate ChkI and Chk. ChkI and Chk can subsequently phosphorylate serine 20.

These modifications may stabilize p53 by promoting interaction with p300/CBP and inhibiting interaction with Mdm2. Additional kinases and acetyl transferases (shown in green) are activated in response to certain types of stress and may be important increasing p53 stabilzation and transactivation fuction. protein E6 (304). The HPV E6 protein is able to bind to and promote the degradation of

p53 (351). The studies with HPV and other small DNA tumor viruses suggest a link

between Rb/E2F cell cycle control and p53-mediated apoptosis. There is evidence that

ectopic expression of various E2F family members can induce both S phase progression

and apoptosis depending on the levels of expressed cDNA or cell type context (56 , 152

211 324 419 440 462). However, in fibroblasts, exogenous expression ofE2FI , but not

E2F2 or E2F3 , causes apoptosis (76, 210).

The link between E2FI and p53 dependent apoptosis has been demonstrated by genetic studies in mice. Apoptosis observed in transgenic mouse models expressing a

fragment of the SV -40 large T antigen and in the central nervous system of Rb- mouse

embryos is p53 dependent and associated with E2FI (246, 305, 408). Expression of an

E2FI transgene in the skin of K5E2FI transgenic mice results in increased proliferation

and p53 dependent apoptosis (316). Additionally, mice deficient for E2F I have an excess of matue T cells due to a defect in thymocyte apoptosis (113).

Expression of E2FI leads to an accumulation of p53 protein (152, 211). A potential mechanism for the stabilization of p53 observed following expression of E2F is activation of the pI9AR/Mdm2 pathway. Expression of E2FI has been shown to

transactivate the expression of the mouse ARF p I ~RF and human p I4 promoters (25 , 172

335). pI~RF encodes a protein that modulates the activity of Mdm2 (321 , 459). Mdm2 is an E3-1ike ubiquitin ligase that regulates the stability of p53 by promoting its degradation by the proteasome (115 , 117, 142, 160). By inhibiting Mdm2 activity, p19ARF allows for stabilization, activation, and accumulation of p53 protein (321 , 459). Thus, it has been ARF hypothesized that pI9 is a key protein linking the Rb/E2F and p53 pathways (25

189). One hypothesis for how aberrations in the Rb/E2F pathway are recognized by p53

is that de-regulated E2F expression activates p53 by inducing pI ~RF

D. DNA damage and E2F

1. DNA double strand break reeognition

Activation of p53 often occurs as a result of damaged DNA. The p53 activation

pathways begin when specialized proteins recognize a DNA break. The DNA damage

sensors are sensitive enough to recognize a single double strand break, an important task

for maintaining genomic integrity and preventing inactivation of critical genes or

chromosomal abnormalities (333 , 416). The double strand break repair pathway is

divided into two major pathways, non-homologous end joining (NHEJ) and homologous

recombination (HR). The NHEJ pathway is the predominant form of repair activated by

double strand breaks.

Within thirty minutes of the induction of DNA double strand breaks caused by ionizing radiation or genotoxic drugs, a complex containing MreII , NbsI , and Rad50

(MRN complex) relocalizes to sites of the DNA breaks (285). The Mre II component contains both exonuclease and endonuclease activities, and is thought to process broken

DNA ends prior to repair (308, 407). Nbsl is required for proper localization of the complex to DNA breaks following ionizing radiation (47). The role ofRad50 in the MRN complex is to hold the broken DNA ends in close proximity allowing for MreII process the broken DNA ends, and repair by other DNA repair factors (70, 161). Three distinct MRN complexes have been observed in cells. Type I foci are thought be involved

in telomere length regulation and can be observed in untreated cells (241 , 437). Type II and Type III foci form early and late, respectively, following exposure of cells to ionizing radiation, and have been termed ionizing radiation induced foci (IRIF) (255). Type IV

MRN foci are associated with normal cellular DNA replication, and may form in order to repair DNA breaks that arise during S phase (64). Type IV foci are thought to relocalize to sites near origins of DNA replication by virtue of an interaction between E2F I and the

Nbsl component of the complex (254).

2. Checkpoint signating

Following relocalization of the MRN complex to sites of DNA double strand breaks, a cell cycle checkpoint is activated resulting in a pause in cell cycle progression or apoptosis. Cells from patients with the rare human disorders Nijniegen breakage syndrome (NBS) and ataxia-telangiectasia-like disease (A TLD), lacking functional Nbsl and Mre II , respectively, fail to undergo growth arrest in response to ionizing radiation

(371 , 384, 394). A functional MRN complex is required for activation of Atm (412) that leads to activation of Chk2 and subsequent phosphorylation and activation of p53 as discussed.

In addition to MRN relocalization, other DNA damage signaling proteins localize to sites of double strand breaks, including yH2AX and 53BP1. Following DNA damage the histone protein H2AX becomes phosphorylated by Atm, and yH2AX rapidly relocalizes to sites of DNA breaks at or near the formation of MRN foci (205 , 338). yH2AX relocalization is though to be important for the recruitment of DNA repair factors

to sites of DNA breaks (309). 53BPI is a DNA damage response protein originally

identified due to its binding to p53 and enhancement ofp53 transactivation function (175

176). 53BPI is recruited to nuclear foci following DNA damage by binding to yH2AX

(426). The importance of 53BPI in cell cycle checkpoint activation has been

demonstrated by mouse models and siRNA gene knockdown experiments in human cells.

Loss of 53BPI results in a defect in Sand G /M checkpoint activation, hypersensitivity to

ionizing radiation, and the development of thymic lymphomas (84, III , 423 , 427).

53BPI , like H2AX, is phosphorylated by Atm (327) and is required for the Atm-

dependent activation of Chk (427) (Figure 1.6).

3. E2F! and DNA damage signating

Following treatment of cells with DNA damaging agents, E2FI protein

accumulates (30, 158, 165, 235, 266, 294), and is phosphorylated at an N-terminal Atm recognition sequence that is unique to E2FI among the E2F family members (235). This phosphorylation of E2FI is largely dependent on Atm and is required for efficient E2FI

stabilization following DNA damage (235). Chk has also been shown to phosphorylate

and stabilize E2FI following DNA damage, and this modification has been shown to be required for E2FI dependent apoptosis following DNA damage by altering its promoter

specifcity (383). Additionally, DNA damage induced apoptosis is compromised in thymocytes from E2Fr mice (235), suggesting that E2FI has multiple roles in DNA damage signaling. 53BP1

Growth Apoptosis Arrest

Figure 1.6. Model of DNA double strand break signating.

DNA double strand breaks are initially detected by the MRN complex, composed of

MreII , NbsI , and RadSO. Relocalization of 53BPI and yH2AX to sites at or near DNA double strand breaks and phosphorylation of these components by Atm acts to transduce through Chk2, the detection of a DNA break. Phosphorylation and activation of pS3 by

Atm, Chk, and possibly other kinses, can lead to activation of a cell cycle checkpoint resulting in growth arrest or apoptosis. E. Thesis aims

The objective of this dissertation was to determine the pathway(s) by which deregulation of the Rb growth control pathway results in p53-dependent apoptosis. Given AR can modulate p53 stability through inhibition of Mdm2 activity, it had been that pI9 proposed that this was the pathway by which E2FI induces apoptosis. We tested this

p I ~RF is not required hypothesis , and the data presented in Chapter III demonstrate that for E2FI-induced apoptosis. Instead, apoptosis correlated with the ability of E2FI to induce covalent modification of p53. The observation that E2FI could induce p53 modifications similar to those observed following DNA damage, prompted us to examine the role of DNA damage kinases in mediating signaling to p53 and apoptosis. The experiments in Chapter IV demonstrate a role for the DNA damage kinases Atm and induced Chk2, as well as the DNA damage recognition and repair protein Nbsl in E2FI-

apoptosis. Comparing the ability of E2F family members E2FI and E2F2, we found that correlated with p53 the induction of Chk2 expression was specific for E2FI and

activation and apoptosis.

To determine if ectopic E2F expression properly modeled Rb family inactivation

we confrmed our results obtained by E2F overexpression by expression of HPV - 16 E7.

In this biologically relevant context, E7 specifically required E2FI and activation of Atm

Nbs I , and Chk2 to induce apoptosis. The experiments in Chapter V address the role of

Nbs I in this apoptosis pathway. Expression of E2F specifically induced the

relocalization of the Nbs I containing MRN complex to nuclear foci that correlated with

the formation of 53BPI and yH2AX containing foci. Due to the role of the MRN complex in cell cycle checkpoint activation following DNA damage, we hypothesized that a similar cell cycle checkpoint may be activated following expression of E2FI. We found that expression of E2FI in normal human fibroblasts blocked cell cycle progression that is mediated, in part, by activating a p2I-dependent checkpoint. These findings provide further insight into the pathways linking deregulated proliferation control to p53 and apoptosis. CHAPTER II

MA TERIALS AND METHODS A. Cell culture

1- Early passage wildtype and genetically matched p53- and MdmT /p5T MEFs were isolated from mouse embryos as described (184). MEFs derived from pI~RF- mouse embryos were a generous gift from Charles Sherr (St. Jude Children s Research

I- Hospital, Memphis , TN). MEFs derived from pI~RF- /p5T mouse embryos were a

generous gift from Gerard Zambetti (University of Tennessee, Memphis , TN). Ink4a

MEFs were kindly provided by Ronald DePinho (Harvard Medical School, Boston, MA).

atm I- and genetically matched wildtype mice were purchased from The Jackson

Laboratory, Bar Harbor, ME. 293 cells were generously provided by Joseph Nevins

(Duke University Medical Center, Durham, NC). MEFs and 293s were cultured in

Dulbecco s modified Eagle medium (DMEM) supplemented with 10% fetal bovine

serum and I % penicilin-streptomycin. Primary human dermal fibroblasts GM003I6B

and GM02270A (Normal), primary human embryonic lung fibroblasts GMOI604 (HEL

fibroblasts), GM03395C and GM05823C (AT), and GM07I66A (NBS) were obtained

from Coriell Cell Repositories, Camden, NJ. Human cells were cultured as recommended

by Coriell.

B. Adenoviral veetors

Recombinant adenoviruses encoding p53, E2FI , E2F2, and E2F3a have been

described previously (76, 211 , 354). An adenovirus encoding an E2FI DNA binding

mutant, E2FI 132 (183), was created by homologous recombination in 293 cells (211

288). An adenovirus encoding the E2FIl-283 mutant was a generous gift from W. Douglas Cress (H. Lee Moffitt Comprehensive Cancer Center, Tampa, FL). The p53N/C virus was

generated from the pCB6+p53N/C construct generously provided by Karen Vousden

(National Cancer Institute, Frederick, MD). The ChkI , DN-ChkI , Chk2, DN-Chk2, and

HPV - 16 E7 recombinant adenoviruses were created by homologous recombination in

Escherichia coli (143). DN-ChkI contains an aspartic acid to alanine substitution at

position 330. Plasmids encoding Chk2 and DN-Chk2 constructs were generously

provided by David Johnson (M.D. Anderson Cancer Center, Smithvile, Texas). DN-

Chk2 contains a serine to alanine substitution at position 347. A plasmid encoding HPV-

16 E7 was generously provided by Karl Munger (Harvard Medical School, Boston

Massachusetts). Control viruses encode either an empty expression cassette or 13-

galactosidase (f3-gal). Infection with control virus had no effect on parameters tested

relative to mock infection (data not shown). Viruses were propagated in 293 cells and

purified by centrifugation through cesium-chloride gradients. Virus titers were

determined by immunohistochemical staining for the adenovirus hexon protein with an

anti-adenovirus antibody (Biodesign International) and visualized using a 3

diaminobenzidine (DAB) substrate kit from Vector Laboratories (50).

C. Virus infection

Cells were washed once with PBS and serum free DMEM containing virus was added to the cells. Infections were carried out at 37 C in 5% carbon dioxide (C0 ) for I hour (211). The viral innoculum was then removed and replaced with DMEM containing the appropriate seru concentrations and cultured under the conditions described above. D. Western blot analysis

Whole cell extracts were harvested from recombinant adenovirus infected cells at

24 hours post infection unless otherwise noted. Cells were washed twice with cold PBS

and lysed in whole-cell extract buffer (SO mM HEPES, 2 mM magnesium chloride 2S0 mM sodium chloride , 0. 1 mM EDTA, I mM EGTA , 0. 1% Nonidet P- , I dithiothreitol, Ix mammalian protease inhibitor cocktail (Sigma), Ix phosphatase inhibitor cocktails I and II (Sigma)) by incubation for 30 min on ice followed by sonication. Soluble proteins were separated by centrifugation at 13 000 x g in a micro centrifuge and supernatants stored at - C. Proteins were separated by SDS-P AGE analysis and transferred to PVDF membrane (Perkin Elmer) by electro blotting. pI9

ARF was detected using anti-pI9 polyclonal antisera at I :SOO (Novus), E2FI was detected using monoclonal antibody KH-95 at I :500 (Santa Cruz Biotechnology), E2F2 was detected using polyclonal antisera C- 20 at I :SOO (Santa Cruz Biotechnology). p53 protein was detected using monoclonal antibody DO- I at I :500 or polyclonal antisera Ab-7 at

I :2000 (Oncogene Research Products), phospho-serine 15 and -serine 20 forms of p53 the phospho-threonine 68 form of Chk, and the phospho-serine 345 form of ChkI were detected using polyclonal antisera specific for each modification at I: I 000 (Cell

Signaling Technologies). ChkI was detected using polyclonal antisera FL-476 at 1:500

(Santa Cruz Biotechnology). Chk was detected using poly clonal antisera H-300 at 1:500

(Santa Cruz Biotechnology) or monoclonal antibody clone #7 at I :500 (Lab Vision

Corporation). Atm was detected using polyclonal antisera Ab-3 at I: ISO (Oncogene Research Products) and the phospho-serine 1981 form of Atm was detected using polyclonal antisera at I :600 (Rockland). p2I was detected using monoclonal antibody F- at I :400 (Santa Cruz Biotechnology). Actin was detected using polyc1onal antisera I-I9 at

I: I 000 (Santa Cruz Biotechnology). Immunoreactive proteins were detected with a chemiluminescence kit (Perkin Elmer) according to the manufacturers recommendations.

Actin blots are shown as protein loading controls. Relative changes in the levels of p53 were estimated from scanned images of western blots using Multianalyst software

(BioRad).

E. Apoptosis analysis by flow eytometry

At the indicated times post infection, cells were trypsinized, combined with any floating cells, pelleted, washed with PBS, repelleted, and resuspended in 400 l of PBS.

All centrifugations were at SOO x g for 5 min at 4 C. Subsequently, cells were fixed in cold ethanol (final concentration, 70%) and stored at 4 C. Cells were processed for propidium iodide (PI) staining as described (211). Flow cytometric analysis was performed by the University of Massachusetts Medical School Flow Cytometry Core

Facility. Cells undergoing apoptosis were defined as the population that contains less than a 2N DNA content.

F. Apoptosis analysis by ELISA

Cells were plated in 10 cm dishes at 6000 cells per cm , or 24 well plates at I x

cells per well. Virus infections were performed 24 hours after plating. At 96 hours post infection, cells were centrifuged at 500 x g for 10 min at 4 , lysed, and Cell Death

Detection ELISAplus assay performed as described by the manufacturer (Roche). The ordinate axis represents DNA fragmentation relative to control which is defined as

Error bars represent standard deviations calculated from experiments performed in triplicate.

G. Immunohistochemieal staining for p53 protein aeeumulation

Cells were infected with the appropriate adenovirus constructs and

immunohistochemically stained for p53 protein as described (210). AdCon, AdE2FI

AdE2FI 13 were infected at an MOI of 100. At the time of harvest (48 hpi), cells were

washed three times with PBS and then fixed for 15 min each in 3.7% formaldehyde

followed by methanol. The cells were then washed in PBS- 5% Tween 20. Cells were

then incubated with anti-p53 monoclonal antibody pAb42I (Oncogene Research

Products) in the presence of 1% BSA in PBS- 5% Tween-20 for 45 min at room

temperature. The cells were washed three times with PBS- 5% Tween 20, and bound

antibody was detected using Vectastain DAB substrate kit as described by the

manufacturer.

H. p53 immunofluorescenee

Cells were infected with AdCon, AdE2FI , or AdE2F2 at an MOI of 100 and at 24

hpi the cells were fixed in 3.7% formaldehyde followed by methanol. Fixed cells were

incubated with anti-p53 monoclonal antibody pAb42I at I :200 (Oncogene Research Products) and polyclonal antisera specific for phospho-Serine 15 p53 at I :200 (Cell

Signaling Technologies). Bound antibodies were detected using fluorescein isothiocyanate (FITC) or Rhodamine Red-X conjugated secondary antibodies (Jackson

Immunoresearch Laboratories, Inc. and Southern Biotechnology Associates, Inc.

I. Cell fraetionation immunofluorescence

Glass coverslips were pre-treated with 40% hydrochloric acid for 2 min followed by a 5 min wash with 70% ethanol. Cells were plated on treated coverslips at 3000 cells per cm . Cells were infected with recombinant adenoviral vectors and at 24 hpi the cells were washed three times with PBS and incubated for 5 min in cytoskeleton buffer (10 mM PIPES pH 6. 100 mM NaCl, 300 mM sucrose, 3 mM MgCh, I mM EGTA, 0.

Triton X- IOO) and 5 min in cytoskeleton stripping buffer (10 mM Tris-Hcl pH 7.4, 10 mM NaCl, 3 mM MgCh, 1% Tween 40 , 0. 5% sodium deoxycholate). Cells were then washed 3 times with PBS and fixed in fixing buffer (200 mM sucrose and 4% paraformaldehyde in PBS, pH 7.4) for 30 min at room temperature and permeablized

(0.5% Triton X- IOO, 20 mM HEPES pH 7.4 50 mM NaCl, 3 mM MgCh, 300 mM sucrose) for 15 min at room temperature (267). Fixed cells were then blocked in 10%

FBS for I hour at room temperature. Cells were then incubated with MreII monoclonal antibody I2D7 at I: 1500 (GeneTex), y H2AX polyclonal antisera at I :600 (Upstate

Biotechnology), 53BPI polyclonal antisera at 1:400 (Novus Biologicals), or Nbsl polyclonal antisera at 1:400 (Novus Biologicals). Bound antibodies were detected using FITC or Rhodamine Red-X conjugated secondary antibodues (Jackson Immunoresearch

Laboratories, Inc. and Southern Biotechnology Associates, Inc.

J. S phase analysis

Cells were plated at 3000 cells per cm . Cells were infected with recombinant

adenoviral vectors and pulsed with 10 mM 5-Bromo- deoxyuridine (BrdU) (Roche)

immediately following infection. At 24 hpi, cells were fixed and permeablized as

described above. Following permeablization, cells were incubated in 50 mM glycine for

10 min at room temperature. DNA was denatured with 4 N HCl with 0.1 % Triton X- IOO

for 10 min at room temperature, and the samples washed extensively with PBS followed

by a wash in 50 mM glycine (254). Cells were then blocked in 10% FBS and incubated

for I hour with polyclonal antisera specific for BrdU at 1:200 (Immunologicals Direct).

Bound antibody was detected using a Rhodamine Red-X conjugated secondary antibody

(Jackson Immunoresearch Laboratories, Inc).

K. Northern blot analysis

Poly-(A) RNA was isolated using the Micro-FastTrack mRNA isolation kit from

cells as described by the manufacturer (Invitrogen). Total cellular RNA was isolated'

using Trizol as described by the manufacturer (Invitrogen). Biotinylated Chk, Atm, and

GAPDH probes were generated by PCR using primers as described by the manufacturer

(KPL). Hybridized probes were visualized with a chemiluminescence kit (KPL) as described by the manufacturer. Blots were sequentially probed and stripped. PCR primers used to make biotinylated probes:

ChkF (5' ATGTCTCGGGAGTCGGATGTTG-

ChkR (5' GCACCACTTCCAAGAGTTTTTGAC-

AtmAF (5' ACGATGCCTTACGGAAGTTGC-

AtmAR (5' GGACAGAGAAGCCAATACTGGACTG-

GAPDHF (5' CAAGGTCATCCATGACAAC-

GAPDHR (5' TGGTCGTTGAGGGCAATG-

L. RNA interferenee

siRNAs used in this study were generated by Qiagen, Valencia, CA. siRNA

oligonucleotides were transfected into cells at a concentration of 100 nM using

Lipofectamine 2000 (Invitrogen) as described by the manufacturer. Control siRNAs

(siCon) recognize either GFP or retrovirus L TR, and had no effect on parameters tested

relative to mock transfection. siRNAs used in this study:

siGFP (5' CGUAAACGGCCACAAGUUC-

siLTR (5' GAUCCAGCAUAUAAGCAGC-

siE2FIa (5' GGCCCGAUCGAUGUUUUCC-

siE2FI b (5' -CUGACCAUCAGU ACCUGGC-

siE2FIc (5' - GUCACGCUAUGAGACCUCA-

siChka (5' -CUCCAGCCAGUCCUCUCAC- Chkb (5' -GAACCUGAGGACCAAGAAC- siE2F2a (5' -GUGCAUCAGAGUGGAUGGC- siE2F2b (5' CAAGAGGCUGGCCUAUGTG- siE2F3a (5' -AGCGGUCAUCAGUACCUCU- siE2F3b (5' -CUGUUAACCGAGGAUUCAG- sip2Ia (5' AGGCCCGCUCUACAUCUUC- sip2Ib (5' CUAGGCGGUUGAAUGAGAG- siAtr (5' GACGGUGUGCUCAUGCGGC- siAtm (5' -GCGCCUGAUUCGAGAUCCU- siNbsia (5' -AUCAUGCUGUGUUAACUGC- siNbsib (5' AUGUUGAUCUGUCAGGACG- sipI4 a (5' ATCCTAAAGGCCGCGGAGT- sipI4ARFb (5' AATAACACCTTCGCTGTC- sip53 (5' GCAUGAACCGGAGGCCCAU- CHAPTER III

E2Fl INDUCES PHOSPHORYLATION OF p53 THAT IS COINCIDENT WITH p53 ACCUMULATION AND APOPTOSIS A. Introduction

During normal cell proliferation, E2F family members modulate the expression of (287). many genes involved in the transition from Gl to S phase and DNA replication E2F transcriptional activity is regulated by interaction with members of the retinoblastoma (Rb) family of proteins (137). Inactivation of Rb by cellular or viral oncoproteins or loss of Rb wil stimulate cells to bypass GO/Gl and enter S phase (96 , or E2F3 is exogenously 101 273 286). S phase induction also occurs when E2FI , E2F2 , but not expressed in quiescent cells (76 , 183 211). In fibroblasts, expression of E2FI

E2F2 or E2F3 , can result in both S phase progression and apoptosis (76, 210).

Apoptosis induction by E2FI occurs primarily by activation of the p53 apoptotic signaling pathway. The link between E2FI and p53-dependent apoptosis has been , 408 demonstrated in cell culture and by genetic studies in mice (211 , 246, 305, 324

440). It has been proposed that the E2F I transcription factor serves as a link between the

Rb/E2F proliferation pathway and the p53 apoptosis pathway by inducing the expression

of p I ~RF a protein that regulates p53 stability. We tested this hypothesis to determine

the requirement for p I ~RF in the E2F I apoptosis pathway and to better understand the

mechanism ofE2FI-induced apoptosis. /-

B. p19ARF is not required for E2Fl-mediated apoptosis

ARF HumanpI4 and mouse pI~RF promoters contain multiple E2F binding sites that are responsive to ectopic expression of E2FI (25, 172 , 335). Because pI ~RF mRNA levels increase upon E2FI expression (76), we wanted to determine whether the elevated AR protein levels. Indeed mRNA levels corresponded to an increase in p I9 , we found that infecting MEFs with an adenovirus encoding E2FI (AdE2FI) led to increased levels of AR protein (Figure 3. AR expression leads to p53 accumulation via P I9 1). Given that p I9 inactivation of Mdm2 functions and that E2FI expression leads to p53-dependent AR was required for E2FI-mediated apoptosis, we determined whether pI9 , p53- dependent apoptosis. We find that E2FI expression induced apoptosis regardless of 2N DNA content (Figure pI~RF status as measured by the percentage of cells with sub-

2). Apoptosis is observed at all multiplicities of infection (MOI) in the wildtype MEFs

ARF but only at the highest dose ofE2FI in the pI~RF- MEFs, suggesting that while pI9 is not required, it may contribute to E2FI-mediated apoptosis. MEFs infected with an adenovirus encoding an E2FI DNA binding mutant, E2FI 13, showed little or no increase in apoptosis over controls. The percentage of apoptotic cells was also measured

I- I- apoptosis in pI ~RF- and p I ~RF- /p5T MEFs to test the requirement for p53 in the

I- observed inpI~RF- MEFs. The pI~RF- /p53- MEFs were less susceptible to apoptosis

as compared to MEFs lacking only pI ~RF (Figure 3.3). In addition, expression of E2FI

l- l- also induced apoptosis in ink4a MEFs (unpublished observations). ink4a mice do not

INK4A express p I9AR because of the deletion of a shared exon with p I6 which encodes a ...... ,-, *:.

wt MEFs p19ARF-I- MEFs

p19ARF

ns- !I.. !c.nc:.

actin -

Figure 3.1. E2Fl induees pl9ARF protein aeeumulation.

Wildtype andpIgARF- MEFs were infected wth AdCon, AdE2FI , or AdE2FI 13 at MOIs of25 (E2Fbow) or 100 (E2Fhi). Cells were harvested and lysates generated at 24 hours AR protein levels. post infection (hpi). Western blot analysis was performed to detect p I9

Actin blot is shown as protein loading control. /- , --.--. . . .. - .".------

WT MEFs :: l Con

t4 t4

E2F1 e13 B::% ::J bcJ

E2F1 tGJ br.: J k :' p19ARF-I- MEFs

con :ll ID 11J.J E2Fl e132 Ewll

E2F1 :QT ll :

() DNA content

Figure 3.2. E2Fl induces apoptosis in the absence of pl9ARF

Wildtype and pI ~RF- MEFs were infected with AdCon, AdE2FI , or AdE2FI 132 at

MOIs of 100 300 , or 500. Cells were harvested for PI staining and flow cytometry at 72 hpi. Percentages represent cells with sub-2N DNA content. /- /- ,, ...... "'.... ""

19ARF-I- MEFs

Con :

E2Fl e13z

E2Fl fJ%

p19ARF /p53 1- MEFs

Con l:UJ t;t_

E2Fl e13z

() DNA content

ARF Figure 3.3. E2Fl induees p53-dependent apoptosis in the absenee of pl9

pI ~RF- and pI ~RF- /p53- MEFs were infected with AdCon, AdE2FI , or AdE2FI 13 at

MOIs of 100, 300 , or 500. Cells were harvested for PI staining and flow cytometry at 72 hpi. Percentages represent cells with sub-2N DNA content. /- /-

cyclin dependent kinase inhibitor (361). Thus, E2FI can induce apoptosis in pI~RF

deficient MEFs that are generated from independent genetic lesions in mice.

C. p19ARF and Mdm2 eontribute to p53 aecumulation AR is necessary for Given that apoptosis does not require pI~RF we asked ifpI9

E2FI-mediated changes in pS3 protein levels. To address this question we expressed

E2FI in pI~RF- MEFs and assessed pS3 protein levels by immunohistochemistry. As

shown in Figure 3.4, E2FI expression resulted in increased levels of endogenous p53

protein in wildtype MEFs. When E2FI was expressed in pI ~RF- MEFs, fewer cells

stained positive for pS3 protein and more modest changes in p53 protein levels were

observed. The changes in endogenous pS3 protein levels observed by

immunohistochemical staining following E2F I expression correlate with the changes

observed by immunoblotting for endogenous p53 (Figure 3. 10). Likewise , Nip et.a!.

observed an increase in the half-life of pS3 following E2FI expression in 32D.3 cells, a AR expression (292). In both MEF genotypes tumor cell line that lacks pI4 , expressing

the E2FIe13 DNA binding mutant had little or no effect on pS3 protein levels. These

results are consistent with a model where p I9AR can contribute to E2F I-mediated

changes in pS3 protein levels.

pI9ARF affects pS3 protein levels by binding to Mdm2 and preventing it from promoting the degradation of pS3 (321 , 459). Therefore, we determined if Mdm2 is necessary for E2F I-mediated accumulation of p53. It is not possible to duplicate the experiments described above for Mdm2 because there are no cell lines or MEFs available /-

9ARF- Con Con

E2Fl

Figure 3.4. E2Fl reqires pl9ARF to induce p53 accumulation.

Wildtype andpI~RF- MEFs were infected with AdCon, AdE2FI , or AdE2FI e13 at an MOI of 100. Cells were fixed and immunohistochemically stained for p53 protein levels at 48 hpi. that lack Mdm2 expression but retain endogenous, wildtype p53. However, MEFs can be

1- on p53 derived from Mdm2- /p53- embryos (184, 263). To analyze the effects ofE2FI protein levels in these cells , we introduced p53 in trans by infecting cells with an Adp53 recombinant adenovirus, essentially creating a "pseudo MdmX MEF. To control for the

I- addition of exogenous p53 transcript, wildtype pI ~RF- and p5 3- MEFs were also infected with Adp53 at the same low MOL As observed in the immunohistochemical and immunoblot analysis for endogenous p53 , E2FI expression resulted in a substantial (?6 fold) increase in exogenous p53 levels in wildtype MEFs in comparison to control infected cells when measured by immunoblot analysis (Figure 3. 5 and Figure 3.10).

I- Others have made similar observations in pI ~RF- MEFs following retroviral gene transfer of E2FI (463). p53 also accumulated in response to E2FI in p5T MEFs that ectopically expressed p53 (Figure 3.5). Therefore, exogenously expressed p53 protein is affected by E2FI in a manner similar to the endogenous protein. Infection with AdE2FI resulted in a modest ( 2.4 fold) increase in total p53 protein levels in Adp53-infected pI~RF-I- MEFs as compared to AdCon infection of the same cell type (Figure 3.5). The

I- basal levels of ectopically expressed p53 were higher in p I ~RF- MEFs relative to

wildtype cells. This observation is consistent with the data obtained by immunostaining

and immunoblotting for endogenous p53 inpI~RF- MEFs (Figure 3.4 and Figure 3.10),

although the difference is exaggerated when cells are infected with Adp53 (Figure 3.5).

I- Others have also reported higher basal levels of p53 in pI~RF- MEFs (73). E2FI

expression minimally affected p53 protein levels in Adp53- infected MdmX /p53- MEFs.

We suspect that the Mdm2 deficiency elevates baseline levels ofp53 in these cells due to ..-,. , ..,, /-.-

!: LL LL (+ p53 J Uo WN NW NW p53

llIlt(l - actin WT MEFs p53

iiJI: .ITI -actin p19ARF - p53 -actin p53 -

,,17 p53

TTr.'\ '-' -actin Mdm2 - /p53 -

Figure 3.5 E2Fl requires pl9ARF and Mdm2 to inerease p53 protein levels.

Wildtype p53- pI~RF- or MdmT /p5;- MEFs were infected with AdCon, AdE2FI or AdE2FI 13 at MOIs of25 (E2Fbow) or 100 (E2Fhi). Cells were harvested and lysates generated at 24 hpi. Western blot analysis was performed to determine levels of p53 protein. Actin blots are shown as protein loading controls. /-

an absence or greatly reduced level of p53 ubiquitin ligase activity. E2F Ie13 had little or no effect on p53 protein levels in each cell type , consistent with a transcriptional mechanism being at least parly responsible for E2FI-mediated accumulation ofp53. The

reduced ability of E2F I to alter p53 levels above controls in p I ~RF- MEFs, or in MEFs lacking Mdm2 but expressing p53, is consistent with a model where E2FI modulates p53 protein levels, in large part, through the pI9 /Mdm2/p53 pathway.

D. E2F expression leads to the phosphorylation of p53

In response to DNA damage, p53 is activated by covalent modifications

including the phosphorylation at certain serines and acetylation of lysine residues. Of

these modifications, phosphorylation of serine 15 on human p53, or serine 18 on mouse

p53 , is commonly observed in response to DNA damage (20, 43). Given that both DNA AR to activate p53 damage (189) and E2FI do not necessarily require pI9 , we determined

whether E2FI-mediated activation of p53 function is coincident with p53

phosphorylation. To detect phospho-serine 18 , we used an antibody specific for this

modification. We find that E2F I , and to a lesser extent E2F2, expression in wildtype

MEFs results in an increase in endogenous p53 protein levels and the phospho-serine 18

form ofp53 as detected by immunoflourescence (Figure 3.6). When merged it is apparent

that only a subset of the cells with an increase in total p53 has increased levels of the

phospho-serine 18 form ofp53. Total p53 protein and the phospho-serine 18 form ofp53

were present at low levels in cells infected with control virus (Figure 3.6). This data suggest that E2FI may signal p53 accumulation and p53 phosphorylation by separate pathways.

E. Differential abilties of E2Fl and E2F2 to phosphorylate p53 and induee

apoptosis

To discriminate between E2FI-specific induction of p53 and increased E2F

activity in general, we measured the levels of p53 in extracts of cells infected with an

adenovirus encoding E2F2 in comparison to E2F I. E2F2 is fuctionally similar to E2F I;

E2F2 is a potent inducer of S phase when expressed in quiescent cells, is normally

induced at the G to S transition, specifically interacts with Rb, and induces a similar

aray of genes when overexpressed (76 , 174 , 226, 279, 358). However, like other E2F

family members, E2F2 is unable to induce apoptosis in REF52 cells (76, 210). We find

that E2F2 expression does not lead to apoptosis in MEFs, even though it is capable of

inducing the expression of pI9AR protein (Figure 3. 8 and Figure 3.9). Ectopic E2F2

expression did result in increased levels of endogenous p53 protein in wildtype MEFs

although not as effectively as E2FI (Figure 3.6 and Figure 3.10). However, expression of

I- E2F I , but not E2F2 , alters p53 protein levels in p I ~RF- MEFs (Figure 3. 7 and Figure

10).

We next compared the ability ofE2FI and E2F2 expression to induce a change in

p53 phosphorylation. We find that ectopic expression of either E2FI or E2F2 leads to an

increase in the levels of both endogenous p53 and the phospho-serine 18 form of p53 in

wildtype MEFs, although the levels of phospho-serine 18 p53 is less in E2F2 expressing DAPI total p53

Con

E2Fl

E2F2

Figure 3.6. Expression of E2Fl or E2F2 results in an increase in p53 protein levels

wild type MEFs. and levels of the phospho-serine 18 form of p53 in

Wildtype MEFs were infected with AdCon, AdE2FI , or AdE2F2 at an MOI of 100. Cells

were fixed and stained for total p53 (rhodamine Red-X) and the phospho-serine 18 form

ofp53 (FITC) at 24 hpi. DAPI staining is shown in blue. Merged images are also shown. /-

DAPI

Con

E2Fl

E2F2

Figure 3.7. E2Fl specifcally induces accumulation ofp53 and the phospho-serine form of p53 in cells lacking pI9ARF pI~RF- MEFs were infected with AdCon, AdE2FI , or AdE2F2 at an MOI of 100. Cells were fixed and stained for total p53 (rhodamine Red-X) and the phospho-serine 18 form ofp53 (FITC) at 24 hpi. DAPI staining is shown in blue. Merged images are also shown. sample (Figure 3. 10). Similar results were observed by immunoflourescence staining for

I- p53 and phospho-serine 18 p53 inpI~RF- MEFs (Figure 3.7). Expression ofE2FI , but not E2F2, leads to increased levels of the endogenous phospho-serine 18 form of p53 in pI~RF-I- MEFs (Figure 3.7 and Figure 3. 10). In addition to phosphorylation at serine 15

(the human equivalent to serine 18 in mouse p53), phosphorylation of serine 20 (serine

23 in mouse) on human p53 is commonly observed following DNA damage (53 , 155

370). Since we have had difficulties detecting mouse phospho-serine 23 p53 by immunoblot analysis, cells were coinfected at a low MOI with an adenovirus encoding human p53 , which when phosphorylated can be detected by the anti-phospho-serine 20 antibody. Using this approach, we find that E2FI expression results in an increase in the phospho-serine 20 as well as the phospho-serine 15 forms of p53 in both wildtype and pI~RF-I- MEFs (Figure 3. 11 A and Figure 3.11 B).

E2F2 expression results in a slight increase in both the phospho-serine 15 and

I- phospho-serine 20 forms ofp53 in wildtype but not inpI~RF- MEFs (Figure 3.11 A and

Figure 3. 11 B). This inability ofE2F2 to affect the phosphorylation ofp53 in the absence ofpI~RF was also observed for the endogenous mouse p53 (Figure 3.7 and Figure 3.10).

The ability of E2F2 to affect p53 protein levels was, unlike E2FI , solely dependent on pI ~RF since both endogenous and exogenous p53 protein levels were unchanged when

I- E2F2 was expressed inpI~RF- MEFs while E2FI expression leads to an increase in p53

I- in both wildtype and pI~RF- MEFs (Figure 3. 10 and 3. 11 C). We note that the differential ability of E2FI and E2F2 to alter the ectopically expressed p53 in pI ~RF- " - - ......

Con.

br:1. :1 .. - .. lJ:Ili Li:iII .. ..

E2F1 . l::-o ...... k:J .DE2F

o DNA content

Figure 3.8. E2F2 does not induee apoptosis in wildtype MEFs.

Wildtype MEFs were infected with AdCon, AdE2FI , or AdE2F2 at MOIs of 100 300, or

500. Cells were harvested for PI staining and flow cytometry at 72 hpi. Percentages represent cells with sub-2N DNA content. /- .., .. ~~~..

wt MEFs P 1 9ARF- MEFs

p19ARF ns- II fJIIT .r' iillllJ. L .

actin

ARF Figure 3.9. E2Fl and E2F2 induee pl9 protein aeeumulation.

, AdE2F2, or AdE2FI Wildtype or pI~RF- MEFs were infected with AdCon, AdE2FI at an MOI of 100. Cells were harvested and lysates generated at 24 hpi. Western blot AR protein. Actin blot is shown as analysis was performed to determine levels of p I9 protein loading control. /- .-

wt MEFs p' 9ARF- - MEFs

L..- NL. NL. Or: L..- NL. UJ UJ UJ UJ UJ p53 -

Ser'8 p53

actin

Figure 3.10. E2F2 requires p19ARF to induce p53 protein and alter levels of the phospho-serine 15 form of p53.

Wildtype or pI ~RF- MEFs were infected with AdCon, AdE2FI , AdE2F2, or AdE2FI e13 at an MOI of 100. Cells were harvested and lysates generated at 24 hpi. Western blot analysis was performed to determine levels of total p53 protein and levels of the phospho-serine 18 form of p53. Actin blot is shown as protein loading control. MEFs appears to be exaggerated relative to the endogenous protein in these experiments.

These results suggest that E2F2 utilizes the pI9 /Mdm2 pathway to affect p53 levels.

F. p53 phosphorylation contributes to E2Fl-mediated apoptosis

Having found a correlation between E2FI-induced phosphorylation of p53 and

apoptosis, we determined if these covalent modifications are required or contribute to

E2FI-mediated apoptosis. Studies of the DNA damage response have suggested that it is

unlikely that a single p53 modification is responsible for activating p53 dependent

apoptosis (48), so we initially addressed this question by using . a p53 mutant in which

most of the N- and C- terminal amino acid residues known to be phosphorylated upon

DNA damage have been changed to alanines. This mutant, p53N/C, has been shown to

activate expression of p2I , Mdm2 and Bax reporter constructs as effectively as wildtype

p53 and has been reported to induce apoptosis in response to certain stimuli (13). Mdm2

has also been shown to regulate the levels of p53N/C (13). Indeed, we find that p53N/C

protein accumulates to levels similar to wildtype p53 when expressed by recombinant

adenovirus transduction (Figure 3. 12 A). To measure apoptosis p53- MEFs were

transduced with AdE2FI or AdE2F2 and either wildtype Adp53 or Adp53N/C (Figure

12 B). Without the addition of p53 , levels of apoptosis in E2FI or E2F2 expressing

samples were similar to AdCon infected cells. Cells expressing both E2F I and wildtype

p53 underwent apoptosis. In contrast, apoptosis was greatly reduced in cells transduced

with E2FI and p53N/C encoding adenoviruses suggesting a contribution by these

,oJ" (+p53)

Ser15 pS3 wt MEFs

Ser15 pS3 - pl9ARF-

(+p53)

Ser20 pS 3 MEFs

Ser20 pS3 - pl9ARF-

(+p53)

wt MEFs actin -

p 1 9ARF- actin -

Figure 3.11. E2Fl induees p53 protein and levels of the phospho-serine 15 and phospho-serine 20 forms ofp53 in cells laekingpl

(A) Western blot analysis of the phospho-serine 15 form ofp53 in wildtype andpI~RF-

MEFs infected with AdCon, AdE2FI , or AdE2F2 and an MOI of 100. (B) Western blot

I- analysis of the phospho-serine 20 form of p53 in wildtype and p I ~RF- MEFs infected with AdCon, AdE2FI , or AdE2F2 and an MOI of 100. (C) Western blot analysis of total

I- p53 protein levels in wildtype and pI ~RF- MEFs infected with AdCon, AdE2FI , or

AdE2F2 and an MOI of 100. Cells were harvested and lysates generated at 24 hpi. Blots were sequentially probed and stripped. Actin blot is shown as protein loading control. potential phosphorylation sites to apoptosis. As expected, E2F2 did not induce apoptosis when co-expressed with either p53 or p53N/C.

We next examined the role of p53 kinases on apoptosis signaling in response to

E2FI expression by using caffeine, a potent inhibitor of several of these kinases. We find that there is a dose dependent decrease in apoptosis in E2FI expressing MEFs following treatment with increasing doses of caffeine (Figure 3. 13). Treatment of cultured cells with caffeine prior to ionizing radiation results in a delayed and attenuated accumulation of p53 protein (192). This defect in p53 accumulation is due to the inhibition ofthe ataxia telangiectasia mutated (Atm) and the Atm-Rad3-related (Atr) kinases that phosphorylate p53 on serine 15. Inhibition of Atm and Atr wil also block subsequent signals that

activate the checkpoint kinases ChkI and Chk2 and perhaps other kinases that phosphorylate p53 on serine 20 (29 , 348). The observations that blocking p53

phosphorylation either by mutating potential phosphorylation sites on p53 or treatment

with the p53 kinase inhibitor, caffeine , can significantly compromise apoptosis

demonstrates that p53 phosphorylation is a critical step in E2FI mediated apoptosis. '" " .. ..",. ,. ..."

p53-1- MEFS Adp53 Adp53N/C

p53 -%SN4 actin \81:.. lIn' UII.AiII,

+ wt p53 + p53N/C

8% .. 0% .00 Con '"

E2F1

-- :2

E2F2 ""

Figure 3.12. p53 phosphorylation correlates with E2Fl-indueed apoptosis.

(A) p5T MEFs were infected with AdpS3 or Adp53N/C at MOIs of 10, 20 , or SO. Cells were harvested and lysates generated at 24 hpi. Western blot analysis was performed to detect total p53 protein levels. Actin blot is shown as protein loading control. (B)

Analysis of apoptosis inp5T MEFs infected with AdCon, AdE2FI , or AdE2F2 at an

MOI of 500 and coinfected with AdpS3 or AdpS3N/C at an MOI of 20. Cells were harvested for PI staining and flow cytometry at 72 hpi. Percentages represent cells with sub-2N DNA content. Con E2F1

Caffeine

Figure 3.13. Caffeine inhibits E2Fl-induced apoptosis.

Analysis of apoptosis in wildtype MEFs infected with AdCon or AdE2FI at an MOI

SOO. Caffeine was added at the indicated doses to the culture medium immediately following viral infection. Cells were harvested and apoptosis detected at 72 hpi. Error bars represent standard deviation of experiment performed in triplicate. /- /-

G. Diseussion

The signaling mechanism by which the RB/E2F pathway in general, and E2FI AR specifically, communicates with pS 3 has been thought to involve the p I9 /Mdm2 AR and Mdm2 are required for the majority of p53 pathway. Indeed, we find that pI9

protein accumulation observed following E2FI expression. However, contrar to the role

of the pI9AR pathway in p53 accumulation, we find that E2FI induces apoptosis in

14). It is unlikely that E2FI induction ofp73 MEFs lackingpI~RF (see model, Figure 3.

p I ~RF- is directly responsible for the apoptosis observed in MEFs since we show that

bothpI~RF andp53. These results are E2FI failed to induce apoptosis in MEFs lacking

p I ~RF in contrast to earlier studies which concluded that is important for E2F I-mediated

(409) and Tolbert et.al. apoptosis (73 , 463). However , reports published by Tsai et.al.

Rb (401) show thatpI~RF was not required for apoptosis in deficient mouse embryos or

following Rb inactivation by transgenic expression of a fragment of the SV -40 large- T

antigen, respectively. Likewise , a report published by Russell et. al. showed that crossing

an E2FI transgenic mouse into apI~RF- background does not reduce apoptosis caused

by expression of the E2F I trans gene (342). Thus, studies using several different models

arrive at the conclusion that the apoptosis associated with deregulated E2F activity does

not require pI

It is conceivable that forcing cells into S phase by ectopic E2F expression can

induce the apoptotic response. However, it is not likely to simply be ectopic S phase

induction following E2F expression that triggers apoptosis, since both E2F I and E2F2

can induce S phase at similar efficiencies (76), and we find that E2F2 does not induce apoptosis in MEFs. Moreover, there does not appear to be a specific phase of the cell cycle in which E2FI induces apoptosis (75, 211). Our findings that ectopic E2FI , but not

E2F2 , expression results in increased p53 phosphorylation in the absence of p I ~RF and that this covalent modification ofp53 contributes to E2FI-mediated apoptosis, suggests that E2FI may activate a cellular response similar to DNA damage.

The observation that caffeine inhibits E2F I-mediated apoptosis suggests that the

action of one or more p53 kinases is likely to be important for E2FI signaling. Signaling

cascades that activate protein kinases responsible for phosphorylating N-terminal

residues on p53 upon DNA damage are well documented (10). Candidate kinases include

ataxia telangiectasia mutated (Atm), the Atm-Rad3-related protein (Atr), ChkI and Chk.

Atm and Atr phosphorylate p53 at serine 15 as well as activate the Chk kinases by

phosphorylation (20 217 238 259). Active Chk kinases can then phosphorylate p53

at serine 15 and serine 20 (370).

Recently, Atm/Atr has been shown to phosphorylate the N-terminus ofE2FI , but

not E2F2 or E2F3 (235). This observation, together with the data presented here leads us

to speculate that ectopic E2FI expression or activation of endogenous E2FI upon

phosphorylation by Atm, leads to increases in the activity and, perhaps, levels of one or

more of the p53 kinases, which then phosphorylate p53 and promote apoptosis. Given

that the E2FI DNA binding mutant, E2FI 13, did not induce p53 phosphorylation or

apoptosis, transcriptional activation of one or more p53 kinases might provide a

mechanism for E2FI activation ofthis pathway. However, overexpression of Atm/Atr .. -

Rb / E2F1 E2F1 Kinase(s) P53 Apoptosis

p19ARF Mdm2 p53 p21 E2F1

E2F2 E2F3 S Phase genes Phase

Figure 3.14. Model ofE2Fl-indueed apoptosis and p53 aeeumulation.

E2FI , E2F2 , and to a lesser extent, E2F3 can signal p53 accumulation through the pI9AR/Mdm2 pathway. p53 phosphorylation contributes to E2FI-mediated apoptosis and AR may function to attenuate E2F proliferation- can occur in the absence of p I ~RF P I9 promoting activity as shown by the dashed lines. Activation of the p I9AR /Mdm2 pathway may increase levels of p53 , contributing substrate to the p53 kinases activated by E2FI , as shown by the dashed arrow. ChkI , or Chk2 kinases has been found to be insufficient for their activation (238, 259).

Therefore, an additional signal(s) may be necessar to stimulate their activity.

It is conceivable that E2FI , E2F2 , or E2F3 could signal throughpI~RF/Mdm2 increase p53 protein levels and that the increased pools of p53 would provide more substrate for the p53 kinases activated by E2FI. Thus , the pI ~RF/Mdm2 pathway may act as both an attenuator of proliferation by targeting E2F family members for degradation and as an amplifier of a DNA damage signal by increasing pools of p53 available for phosphory lation. The decision to undergo growth arrest or apoptosis would then depend on the cellular context or extent of DNA damage.

Our data implicates p53 phosphorylation as a key step in E2FI-mediated p53- dependent apoptosis. These observations raise the possibility that E2FI signaling and

DNA damage response pathways may converge and involve the same or related kinases to activate p53. Alternatively, E2FI may contribute to or be a component of the DNA damage pathway. Given these possibilities, it is possible that a role of E2FI may be to amplify DNA damage signals resulting in p53-mediated apoptosis. CHAPTER IV

APOPTOSIS ASSOCIATED WITH DEREGULA TED E2F ACTIVITY IS DEPENDENT ON E2Fl AND Atm/Nbsl/Chk2 A. Introduetion

Having found that E2FI can induce p53-dependent apoptosis in the absence of pI~RF we wanted to determine the pathway(s) by which E2FI activates p53 to induce cell death. The findings presented in chapter III demonstrate a requirement for covalent modification of p53 in the E2FI apoptosis pathway, and that the p53kinase(s) activated by E2FI are sensitive to caffeine. The apoptosis program initiated by DNA damage also correlates with p53 modification, with a subset of DNA damage responsive p53 kinases being sensitive to caffeine (10). Therefore, we hypothesized that E2FI may activate one or more DNA damage response kinase cascades resulting in p53 activation and apoptosis.

Kinases that have been shown to phosphorylate the N-terminus ofp53 in response to DNA damage include ataxia telangiectasia mutated (Atm), the Atm-Rad3-related protein (Atr), and the human checkpoint kinases ChkI and Chk2. Atm and Atr

phosphorylate p53 at serine 15 as well as activate the Chk kinases by phosphorylation

(20 217 238 259). Active Chk kinases can then furher phosphorylate p53 at serine

15 and serine 20 (370). Activation of downstream kinase activation by Atm can also

require Nbsl (40, 234, 460). To address which kinase(s) may be involved in E2FI-

mediated p53 activation and apoptosis, we used primary human fibroblasts from patients

with ataxia telangiectasia and Nijmegen breakage syndrome, lacking functional Atm and

Nbsl gene products, respectively. Using cells from these patients, we tested the

hypothesis that E2FI and deregulation ofRb family members by expression of the HPV-

16 E7 protein induces apoptosis through activation of a pathway that shares some

similarities to the apoptosis program induced by DNA damage. B. Roles for Atm and Nbsl in apoptosis induction

Atm kinase activity is often induced in response to cellular stress leading to phosphorylation of many substrates, including serine 15 on p53. Given that ectopic E2FI expression also results in phosphorylation of serine 15 on p53 (340), we determined whether Atm was required for E2FI-mediated apoptosis and p53 phosphorylation. We found that E2Fl-mediated apoptosis was compromised in fibroblasts isolated from an AT patient ectopically expressing E2FI (Figure 4. 1). Similar results were obtained from AT fibroblasts isolated from a different donor (data not shown). In addition, apoptosis was

I- also reduced following expression of E2FI in atm MEFs (Figure 4.2). The apoptosis observed in fibroblasts ectopically expressing E2FI appears to be specific to E2FI because the related E2F family member, E2F2, was unable to induce apoptosis (Figure

1).

We next examined the ability ofE2FI and E2F2 to induce the phosphorylation of serine 15 and serine 20 residues on p53 in the absence of Atm. We found that in cells lacking Atm, both E2FI and E2F2 were still able to induce total p53 protein levels AR (340). However (Figure 4.3) likely due to induction of p I4 , phosphorylation of p53 at serine 15 was reduced and phosphorylation at serine 20 was absent in AT cells following

E2FI expression (Figure 4.3). An increase in the phospho-serine 15 form ofp53 was also observed following E2F2 expression in normal, but not AT, cells (Figure 4.3). Because

E2F2 expression led to lower levels of the phospho-serine 15 form of p53 than E2F I , it was possible that E2F2 was not inducing apoptosis because it is not as efficient at c: 8

Con E2F1 E2F2 Con E2F1 E2F2 Con E2F1 E2F2 Normal NBS

Figure 4.1. E2Fl-indueed apoptosis is redueed in human fibroblasts laeking funetional Atm or Nbs1. , and NBS Apoptosis analysis in normal human dermal fibroblasts, AT fibroblasts (AdE2Fl), fibroblasts. Cells were infected with recombinant adenovirus encoding E2FI

E2F2 (AdE2F2), or a control virus (AdCon) at an MOI of 1000. Cells were harvested and

represent standard apoptosis detected at 96 hours post infection (hpi). Error bars

deviation of experiment performed in triplicate. Con E2F1 Con E2F1 afm

Figure 4.2. E2Fl-indueed apoptosis is redueed in mouse embryo fibroblasts laeking atm.

I- Apoptosis induction in wildtype and atm MEFs. Cells were infected with AdE2FI or

AdCon at an MOI of 500. Cells were harvested and apoptosis detected at 72 hpi. Error bars represent standard deviation of experiment performed in triplicate. Normal NBS

Con E2F1 E2F2 Con E2F1 E2F2 Con E2F1 E2F2

p53

actin

Normal NBS

Con E2F1 E2F2 Con E2F1 E2F2 Con E2F1 E2F2

Ser15

actin

Ser20

actin

Figure 4.3. E2Fl and E2F2 require Atm and Nbsl to effieiently induee phosphorylation of p53.

Western blot analysis of p53 protein levels following infection with AdCon, AdE2FI

AdE2F2 at an MOI of 1000. Cells were harvested and lysates generated at 24 hpi. The phospho-serine 15 and phospho-serine 20 forms of p53 were detected with polyc1onal antisera specific for each modification. Actin blots are shown as protein loading controls. inducing Atm activity as E2F1. To control for the differences in activation by E2FI and

E2F2 of the kinase(s) that phosphorylate p53 on serine 15, we expressed E2F2 at doses

that resulted in levels of serine 15 phosphorylation and p53 accumulation that were

similar to levels observed following E2FI expression (Figure 4.6). Even at these elevated

doses, E2F2 still did not induce apoptosis (Figure 4.5) or phosphorylation of p53 at serine

20 (Figure 4.6). Because E2F2 can also activate Atm resulting in p53 phosphorylation at

serine 15 in the absence of apoptosis, these results suggest that while E2FI requires Atm

. to signal apoptosis, Atm activation is not the commitment step for apoptosis induction.

In addition to directly phosphorylating p53 on serine 15 (20 , 43), Atm also

activates other kinases that lead to p53 phosphorylation on serine 20 (53 , 155 259 370

443). Since downstream kinase activation by Atm can require Nbsl (40 234 460), we

asked whether functional Nbsl protein was necessary for E2FI-induced apoptosis. We

found that E2FI-induced apoptosis was compromised in fibroblasts from NBS patients

similar to the reduction observed in AT cells (Figure 4. 1). Although expression of E2FI

was found to be slightly lower in AT cells than in normal cells (Figure 4.4), increased

amounts of E2F I stil did not induce apoptosis in AT cells (data not shown). Multiple

forms of both endogenous and exogenously expressed E2FI are routinely observed by

western blot, likely due to posttranslational modifications associated with E2F

regulation (212 252 253 310). It remains unclear as to what patterns ofposttranslational

modifications contribute to the observed altered mobility of E2FI. Although E2FI was

able to induce total p53 protein levels in NBS cells (Figure 4.3), we observed a modest Con E2F1

E2F1

actin

Con E2F2

E2F2

actin

Figure 4.4. AdE2Fl and AdE2F2 are expressed to similar levels in normal, AT, and

NBS fibroblasts.

Normal, AT, and NBS fibroblasts were mock infected or infected with a low dose (MOI of 100) of either E2FI or E2F2. Cells were harvested and lysates generated at 24 hpi and levels ofE2FI or E2F2 were compared by western blot analysis. Actin blots are shown as protein loading controls. Con E2F1 E2F2

Figure 4.5. Inereased doses of AdE2F2 does not lead to apoptosis.

Normal human fibroblasts were infected with AdCon, AdE2FI at an MOI of 1000, or increasing doses of AdE2F2 (MOI of 1000, 1500, 2000, or 2500). Cells were harvested and apoptosis detected at 96 hpi. Error bars represent standard deviation of experiment performed in triplicate. p53

actin

Ser15

actin

Ser20

Figure 4.6. Increased doses of AdE2F2 lead to inereased levels of phospho-serine 15 p53 but not phospho-serine 20 p53.

Normal human fibroblasts were infected with AdCon, AdE2FI at an MOI of 1000, or increasing doses of AdE2F2 (MOI of 1000, 1500, 2000, or 2500). Cells were harvested and lysates generated at 24 hpi. Cellular lysates were analyzed for levels of p53 and the phospho-serine 15 and phospho-serine 20 forms ofp53. Actin blots are shown as protein loading controls. decrease in the levels of the phospho-serine 15 form and a large decrease in the levels of the phospho-serine 20 form ofp53 in NBS cells following E2FI expression (Figure 4. 3), demonstrating that functional Nbsl protein is required for E2FI-mediated apoptosis and for signaling p53 phosphorylation at the serine 20 residue. Ectopic E2F2 expression found to be similar in all three cell types (Figure 4.4), failed to induce apoptosis in NBS cells (Figure 4. 1) but did cause an increase in both total p53 levels and the phospho- serine 15 form of p53 (Figure 4.3). These results are consistent with a mechanism whereby E2F2 alters the phospho-serine 15 form of p53 in NBS cells through its ability to activate Atm.

c. Chk2 is required for E2Fl-mediated apoptosis

Given that phosphorylation of p53 on serine 20 correlates with apoptosis, we proceeded to use this as a marker to identify any additional kinase(s) that may contribute to E2FI-mediated apoptosis. Among the Atm-induced kinases that require functional

Nbsl protein for activation and that directly phosphorylate p53 on serine 20, is the human

checkpoint kinase Chk2 (40, 234, 370). To examine the role of Chk2 in E2FI-induced

apoptosis, we coexpressed E2FI with a kinase defective form of Chk2 (DN-Chk2) to

inhibit Chk2 kinase activity in fibroblasts. We observed a reduction in apoptosis when

E2FI was coexpressed with DN-Chk. Apoptosis levels did not appreciably change with

a dominant negative form of ChkI (DN-ChkI) (Figure 4.7), another DNA damage

responsive kinase that is capable of phosphorylating p53 on serine 20 (370). Expression c 50

U: 30

CD 20

DN- DN- DN- DN- Con E2F1

Figure 4.7. Dominant negative Chk2, but not dominant negative Chkl reduees

E2Fl-induced apoptosis.

Normal human fibroblasts were infected with AdCon or AdE2FI at an MOI of 1000 and

coinfected with a recombinant adenovirus encoding dominant negative ChkI (DN-ChkI)

or dominant negative Chk2 (DN-Chk2) at an MOI of 1000. Cells were harvested and

apoptosis detected at 96 hpi. Error bars represent standard deviation of experiment

performed in triplicate. siCon siChk2a siChk2b Time: 24 48 24 48 24 48 Chk2

5 60

Con E2F1 E2F2 Con E2F1 E2F2 siCon siChk2b

Figure 4.8. siRNA targeting of Chk2 inhibits E2Fl-indueed apoptosis.

(A) HEL fibroblasts were transfected with siChka, siChkb, or a control siRNA (siCon).

Cells were harvested and lysates generated at 48 hours post transfection. Actin blot is shown as protein loading control. (B) HEL fibroblasts were transfected with siChkb or siCon 24 hours prior to infection with AdCon, AdE2FI , or AdE2F2 at an MOI of 250.

Cells were harvested and apoptosis detected at 96 hpi. Error bars represent standard deviation of experiment performed in triplicate. . '- -

ofDN-ChkI or DN-Chk alone did not alter levels ofE2FI protein (data not shown). To

confirm the involvement ofChk in E2FI-mediated apoptosis, we used siRNAs to reduce

the levels of Chk2 in cells (Figure 4. 8 A). We observed a reduction in apoptosis

following E2FI expression in cells transfected with siChkb (Figure 4.8 B), and no effect

of this siRNA following expression ofE2F2 (Figure 4. 8 B). Similar results were obtained

using siChka (data not shown).

We next determined the involvement of Chk in E2F 1- induced pS 3 accumulation

and modification. Since Chk directly phosphorylates p53 on serine 20, we asked ifDN-

Chk2 expression or Chk2 siRNA transfection could block E2FI-induced p53

phosphorylation. We found that DN-Chk2 expression and the Chk2 siRNA reduced the

levels of the phospho-serine 20 form ofp53 following E2FI expression but had no effect

on either total p53 levels or the levels of the phospho-serine 15 form of p53 (Figure 4.

and 4. 10). Coexpression of DN-ChkI was unable to inhibit E2FI-induced p53

accumulation and had only a modest effect on p53 phosphorylation (Figure 4.9).

D. E2Fl specifically induees Chk2 expression

We have shown that E2F I requires Atm, Nbs I , and Chk2 to efficiently induce

apoptosis. However, E2F2 is also able to activate Atm resulting in phosphorylation of

p53 at serine 15 but does so without inducing apoptosis. We confirmed the activation

state of Atm using an antibody that recognizes a modified form of Atm observed

following DNA damage that correlates with Atm activation (17). Expression of either

E2F I or E2F2 led to an increase in the levels of the phospho-serine 1981 form of Atm . . "

Con E2F1

p53

actin III:

Ser15

actin

Ser20

actin

Figure 4.9. DN-Chk2, but not DN-Chkl, inhibits phosphorylation of p53 at serine

20.

Normal human fibroblasts were infected with AdCon or AdE2FI at an MOI of 1000 and coinfected with DN-ChkI or DN-Chk2 at an MOI of 1000. Cells were harvested and lysates generated at 24 hpi. Western blot analysis was performed to determine levels of p53 protein and levels of the phospho-serine 15 and phospho-serine 20 forms of p53.

Actin blots are shown as protein loading controls. Con E2F1

en . p53

actin

Ser15

actin

Ser20

actin

Chk2

actin

Figure 4.10. siRNA targeting ofChk2 inhibits E2Fl-induced phosphorylation ofp53 at serine 20.

HEL fibroblasts were transfected with siCon or siChk2. Twenty four hours post transfection, cells were infected with AdCon or AdE2FI at an MOI of 250. Cells were harvested and lysates generated at 24 hpi. Western blot analysis was performed to determine levels of p53 protein, levels of the phospho-serine 15 and phospho-serine 20 forms of p53 , and levels of Chk2 protein. Actin blots are shown as protein loading controls. Normal NBS

Con E2F1 E2F2 Con E2F1 E2F2 Con E2F1 E2F2

Chk2

actin

Chk1

actin

Figure 4.11. Expression of E2Fl, not E2F2, leads to inereased Chk2 protein levels

normal AT, and NBS eells.

Normal human fibroblasts, AT fibroblasts, and NBS fibroblasts were infected with

AdCon, AdE2Fl , or AdE2F2 at an MOI of 1000. Cells were harvested and lysates generated at 24 hpi. Western blot analysis was done to determine levels of ChkI and

Chk2 protein. Actin blots are shown as protein loading controls. chk2

actin

p53

actin

Ser15

acUn

Ser20 Con E2F1 e132 actin

Figure 4. 12. Expression of E2Fl 132 fails to induee apoptosis, inerease Chk2 protein levels or induce phosphorylation of p53.

(A) Normal human fibroblasts were infected with AdCon, AdE2FI , or AdE2FI 13 at an

MOI of 1000. Cells were harvested and apoptosis detected at 96 hpi. Error bars represent standard deviation of experiment performed in triplicate. (B) Normal human fibroblasts were infected with AdCon, AdE2FI , or AdE2FI 13 at an MOI of 1000. Cells were harvested and lysates generated at 24 hpi. Western blot analysis was done to determine levels of Chk2 protein or levels of (C) p53 protein and the phospho-serine 15 and phospho-serine 20 forms of p53 . Actin blots are shown as protein loading controls. while leaving the total Atm protein levels unchanged (Figure 4. 12). The difference between E2F I and E2F2 signaling appears to be the ability of E2F I to stimulate Chk2 activity, which results in an increase in the phospho-serine 20 form of p53 and correlates with E2FI-induced apoptosis. Because Atm activation is upstream of Chk2 in signaling to p53 , E2FI expression must have an additional effect(s) downstream or independent of

Atm that is specific to E2FI for apoptosis induction. We found that expression ofE2FI but not E2F2, led to an increase in the levels of Chk2 protein, and this increase occurred in the absence of Atm or functional Nbsl (Figure 4. 11). E2FI expression also results in accumulation of the phospho-threonine 68 form ofChk (data not shown), a modification observed following DNA damage that may be associated with Chk2 activation (259

264). The phospho-threonine 68 modification of Chk may not be a reliable marker of

Chk2 activation (6, 356, 439). Instead, we examined the Chk substrate p53 serine 20 residue as a marker for Chk activation. Whle expression of E2F I resulted in an increase in Chk protein levels in the absence of functional Atm or Nbs I (Figure 4. 11), we did not observe an increase in the phospho-serine 20 form of p53 in these cells (Figure 4. 3).

Induction and activation of Chk2 by E2FI appeared to be specific because E2FI expression did not result in an increase in ChkI protein (Figure 4. 11) or in an increase in the phospho-serine 345 form of ChkI (data not shown). Chk2 protein accumulation

appeared to result from an increase in Chk2 mRNA levels following E2FI expression

(Figure 4. 13 A). E2F2 expression did not lead to an increase in Chk2 protein levels

(Figure 4. 11) or Chk2 mRNA (Figure 4. 13 A). Expression of an E2FI DNA binding Atm Ser1981 Atm

Figure 4.13. Expression of E2F! or E2F2 leads to an increase in the phospho-serine

1981 form of Atm while leaving total Atm protein levels unehanged.

Normal human fibroblasts were infected with AdCon, AdE2FI , or AdE2F2 at an MOI of

1000. Cells were harvested and lysates generated at 24 hpi. Western blot analysis was performed to determine levels of Atm and levels of the phospho-serine 1981 form of

Atm. ..

L. () WC\

Chk2

GAPDH

Atm

GAPDH

Figure 4. 14. E2Fl, not E2F2, leads to an inerease in Chk2 mRNA levels but not Atm mRNA levels.

(A) Northern blot analysis for Chk2 mRNA isolated from normal human fibroblasts infected with AdCon, AdE2FI , or AdE2F2 at an MOI of 1000. GAPDH shown as

loading control. (B) Northern blot analysis for Atm mRNA isolated from normal human fibroblasts and infected as in (A). GAPDH shown as loading control. mutant, E2FI 13, did not lead to an increase in Chk2 protein levels over control cells

(Figure 4. 12) suggesting that Chk may be induced by a transcriptional mechanism. We also did not detect apoptosis , p53 accumulation, or p53 phosphorylation following expression of E2FIe13 (Figure 4. 12). Since neither E2FI nor E2F2 induced the

expression of Atm mRNA (Figure 4. 13 B), induction of Chk2 expression appears to be specific to E2F I and correlates with the induction of apoptosis.

E. Chk2 and E2F eooperate in apoptosis induction

Given that the induction of Chk2 mRNA and protein levels following expression of E2FI is associated with E2FI-specific apoptosis, we next asked whether Chk2 cooperates with E2FI to signal apoptosis. We found that coexpression of Chk2 with a reduced amount of E2F I encoding virus resulted in enhanced levels of apoptosis whereas expression of Chk alone had a nominal effect on apoptosis levels (Figure 4. 14).

Thus, it appears that Chk is limiting for E2FI-induced apoptosis. This observation raises the possibility that the different abilities of E2FI and E2F2 to activate the apoptosis

program lies in the capacity of E2F I to induce Chk2 expression. Given this possibility,

we determined if Chk2 could cooperate with E2F2 to induce apoptosis if provided

trans . We found that co expression of Chk2 with E2F2 permitted E2F2 to induce apoptosis (Figure 4. 14) and led to an increase in the phospho-serine 20 form of p53

(Figure 4. 15). Coexpression ofE2FI or E2F2 with ChkI had no effect on apoptosis (data not shown). The sudden onset of serine 20 phosphorylation at the point where cells are E2F1 E2F2 Con 500

Figure 4.15. E2Fl and E2F2 cooperate with Chk2 in apoptosis induction.

Normal human fibroblasts were infected with AdCon, AdE2Fl , or AdE2F2 at an MOI of

SOO and coinfected with the indicated doses of AdChk2. Cells were harvested and apoptosis detected at 96 hpi. Error bars represent standard deviation of experiment performed in triplicate. g g

Con E2F1 E2F2 000 000 000 a a a 00000 Chk2 a .. L! .. C' L! a .. L! .. C' L! p53

actin

Ser20

actin

Figure 4.16. Chk2 eooperates with E2F2 to induce phosphorylation of p53 at serine

20.

Normal human fibroblasts were infected with AdCon, AdE2FI , or AdE2F2 at an MOI of

500 and co infected with the indicated dose of AdChk2. Cells were harvested and lysates generated at 24 hpi. Western blot analysis was performed to determine levels of total p53 protein and levels of the phospho-serine 20 form ofp53. Actin blots are shown as protein loading controls. undergoing apoptosis in response to coexpression of E2F2 and Chk2 may mean that

additional events are required to lead to phosphorylation ofp53 through Chk2. These data

suggest that an E2FI-specific increase in Chk2 expression is essential for p53 activation

and apoptosis induction.

F. Apoptosis induetion by HPV-l6 E7 is dependent on E2Fl and Atm/Nbsl/Chk2

We next examined the role of Chk2 in apoptosis resulting from deregulation of endogenous E2F activity. The human papiloma virus (HPV) type 16 E7 protein binds to and inactivates Rb family members resulting in the release of Rb-associated factors including E2F proteins (277). We found that expression of the HPV- I6 E7 protein resulted in apoptosis induction in human fibroblasts plated at low density (Figure 4. 16).

Similar to the apoptosis observed following ectopic E2F I expression, apoptosis resulting from Rb family inactivation by HPV- I6 E7 required functional Atm and Nbsl proteins

(Figure 4. 17). Since Rb inactivation by E7 results in release of five E2F family members from Rb proteins, we used siRNAs targeted to E2FI to address the requirement for E2FI in E7 induced apoptosis. We screened three siRNAs targeted to E2FI for their ability to inhibit E2FI expression (Figure 4. 18 A). siE2FIc was used for the remainder of the experiments shown, but similar results were obtained with the other E2FI siRNAs (data not shown). We found that pre-treatment of cells with siE2F I blocked the ability of E7 to induce apoptosis (Figure 4. 18 B and data not shown). Having observed that E7 induces apoptosis through E2F! , we next determined ifE7-induced apoptosis requires Chk. We found that reducing Chk1evels with an siRNA decreased the ability ofE7 to induce g, 12

Q) 4

Con E2F1

Figure 4.17. HPV-16 E7 induees apoptosis in human fibroblasts.

HEL fibroblasts were infected with AdE2FI at an MOI of250, or AdCon and AdE7 at an

MOI of 1000. Cells were harvested and apoptosis detected at 96 hpi. Error bars represent

standard deviation of experiment performed in triplicate. Con Con Con Normal NBS

Figure 4. 18. HPV-16 E7-induced apoptosis is reduced in eells lacking funetional

Atm or Nbs1.

Normal, AT, and NBS fibroblasts were infected with AdCon or AdE7 at an MOI of 1000.

Cells were harvested and apoptosis detected at 96 hpi. Error bars represent standard deviation of experiment performed in triplicate. E2F1

actin

Con Con Con siCon siE2F1 c siChk2b

Figure 4.19. siRNA targeting ofE2F1 or Chk2 inhibits E7-indueed apoptosis.

(A) Western blot analysis ofE2FI protein levels in cells transfected with siCon, siE2FIa siE2FI b, or siE2FIc. Cells were harvested and lysates generated at 48 hours post transfection. Actin blot is shown as protein loading control. (B) Analysis of apoptosis in cells transfected with the marked siRNA 24 hours prior to infection with AdCon or AdE7 at an MOI of 1000. Cells were harvested and apoptosis detected at 96 hpi. Error bars represent standard deviation of experiment performed in triplicate. 1;'

apoptosis (Figure 4. 18 B). To confirm the requirement of and determine the specificity

for E2FI in E7-mediated apoptosis, we used siRNAs targeted to E2FI , E2F2 or E2F3

(Figure 4. 19). siE2F2a and siE2F3a were used for the remainder of the experiments

shown, but similar results were obtained with the other E2F2 and E2F3 siRNAs (data not

shown). We found that neither siE2F2 nor siE2F3 were able to block apoptosis induced by E7 expression (Figure 4.20). These data suggest that apoptosis resulting from

deregulation of endogenous E2F activity occurs specifically through E2FI and the

Atmlbs/Chk pathway.

G. HPV- 16 E7 induees Chk2 expression and p53 Modifieation

We next asked ifE7 expression induces modifications to p53 similar to E2F1. We

found that E7 expression resulted in an increase in p53 levels, and the levels of the

phospho-serine 15 and phospho-serine 20 forms ofp53 (Figure 4.21). Additionally, Chk

protein levels were elevated following either E2FI or E7 expression (Figure 4.22 A).

phospho-serine 1981 Similar to E2F I , expression of E7 resulted in an increase in the

form of Atm while leaving total Atm protein levels unchanged (Figure 4.22 B).

Expression of E7 also resulted in an increase in the phospho-threonine 68 form of Chk2

(data not shown). These observations suggest that E7 is able to activate Atm kinase

activity resulting in an increase in active Chk kinase.

We next determined whether E2FI was required for E7 to increase Chk and p53

levels, and modifications to p53. We found that E2FI is not required for much of the

observed increase in p53 or the phospho-serine 15 form ofp53 following E7 expression 100

E2F2

actin

ro

E2F3

actin

Figure 4.20. siRNA knockdown of E2F2 and E2F3. siRNAs targeted to E2F2 or E2F3 were transfected into HEL fibroblasts. Cells were harvested and lysates generated at 48 hours post transfection. Western blot analysis was performed to determine effciency of siRNA targeting of E2F2 or E2F3. Actin blots are shown as protein loading controls. " ' . ,- ;.

101

Con Con E7 Con E7 Con E7 siCon siE2F1c siE2F2a siE2F3a

Figure 4.21. HPV-l6 E7 requires E2Fl but not E2F2 or E2F3 to induce apoptosis.

HEL fibroblasts were transfected with the marked siRNA 24 hours prior to infection with

AdCon or AdE7 at an MOI of 1000. Cells were harvested and apoptosis detected at 96

hpi. Error bars represent standard deviation of experiment performed in triplicate. 102

Con E2F1 p53

actin

Ser15

actin

Ser20

actin

Figure 4.22. HPV-16 E7 expression results in p53 accumulation and modifeation similar to those observed following expression of E2F1.

HEL fibroblasts were infected with AdE2FI at an MOI of250, or AdCon and AdE7 at an

MOI of 1000. Cells were harvested and lysates generated at 24 hpi. Western blot analysis was performed to determine the levels of total p53 protein and the levels of the phospho- serine 15 and phospho-serine 20 forms ofp53. Actin blots are shown as protein loading controls. 103

(Figure 4.23). This increase in p53 protein was likely due to activation of the

pI4 /Mdm2 pathway by E2F2 and/or E2F3, while the increase in the phospho-serine 15 form of p53 is likely due to the ability of E2F2 , and possibly other E2Fs, to activate

Atm (Figure 4. 12 and Figure 4.22 B). However, we found that E2Fl was required for E7 to induce the phosphorylation of p53 at serine 20 and to increase the levels of Chk2 protein (Figure 4.23). Additionally, we found that E7 expression resulted in an increase in

Chk2 mRNA levels (Figure 4.24) and this could be attributed to E2FI (Figure 4. 24).

Having found that E2F I is required for E7 to induce C h k 2 expression and

phosphorylation of p53 at serine 20, we next determined if Chk2 was required for E7 to induce this modification. We found that an siRNA directed against Chk2 was able to

block E7-induced phosphorylation of p53 at serine 20, while total p53 levels and the levels of the phospho-serine 15 form of p53 remained unaffected by addition of this siRNA (Figure 4.23). These results demonstrate a requirement for E2FI in Chk2 induction and kinase activation following Rb inactivation by HPV - 16 E7 as measured by an increase in the phospho-serine 20 form of p53. Taken together , apoptosis resulting from inactivation of Rb family members is dependent specifically on E2F 1 and its ability to induce Chk2 expression. 104

Con E2F1

Chk2

actin

Con E2F1

Atm

Ser1981 Atm

Figure 4.23. HPV-16 E7 expression results in an inerease in Chk2 protein levels and levels of the phospho-serine 1981 form of Atm, while leaving total Atm protein levels unehanged.

(A) HEL fibroblasts were infected with AdE2FI at an MOI of 250, or AdCon and AdE7 at an MOI of 1000. Cells were harvested and lysates generated at 24 hpi. Western blot analysis was performed to determine Chk protein levels. Actin blot is shown as protein loading control. (B) HEL fibroblasts were infected as in (A). Cells were harvested and lysates generated at 24 hpi. Western blot analysis was performed to determine Atm protein levels and levels of the phospho-serine 1981 form of Atm. ... . ,.

105

Con

tJ tJ

00 00

p53

E2F1 \':)i n 'fi"-

actin - llij l'tm

Ser20

actin

Ser15

Chk2

actin LJ, rJi

Figure 4.24. HPV-l6 E7 requires E2Fl and Chk2 to inerease the levels of the phospho-serine 20 form of p53.

HEL fibroblasts were transfected with the marked siRNA 24 hours prior to infection with

AdCon or AdE7 at an MOI of 1000. Cells were harested and lysates generated at 24 hpi.

Western blot analysis was performed to determine the levels of total p53, the levels of the phospho-serine 15 and phospho-serine 20 forms of p53 , and levels of Chk2. Actin blots are shown as protein loading controls. + '

106

c:c:0 u.N 00 0 00 ' c: u. c: + o N I' o I'UJ

Chk2

GAPDH

Chk2. Figure 4.25. HPV- l6 E7 requires E2Fl to induee

HEL fibroblasts were untransfected or transfected with the marked siRNA 24 hours prior to infection with AdE2FI at an MOI of 250, or AdCon and AdE7 at an MOI of 1000. Chk2 RNA was isolated at 24 hpi. Northern blot analysis was done to determine levels of

RNA. GAPDH shown as loading control. .:'

107

H. Diseussion

In this chapter we describe a pathway involving the E2F I-specific induction of

Chk2 expression that links loss of proliferation control to an apoptotic pathway with

some similarity to the apoptosis pathway induced by DNA double strand breaks. Unlike

the DNA damage signals that activate p53 in the absence of de novo gene expression

(178), E2F I-mediated apoptosis requires induction of Chk2 expression and possibly other

components of apoptosis signaling (283) to fully activate p53 and kil cells. The results

presented here suggest that deregulated E2F 1 function induces apoptosis by activation of

an AtmlNbsllChk2/p53 pathway following disruptions in the Rb/E2F proliferation

pathway (Figure 4.25).

To model deregulation ofE2F proteins, we expressed the HPV- I6 E7 protein, and

determined the requirement for E2F family members and the Atmlbs lIChk pathway

for signaling to p53 and apoptosis induction. It has been suggested that E2FI is

resonsible for a portion of the phenotypes associated with E7 expression, including

disruption of normal cell cycle control , interfering with cellular differentiation, and

apoptosis induction (54 , 286). Supporting the involvement of E2FI in E7-induced

apoptosis, transgenic mice that express E7 in the lens exhibit increased apoptosis in the

fiber cell compartment that is dependent on E2FI (261 , 304). The results presented here

suggest that apoptosis induction by HPV- I6 E7-mediated Rb family inactivation in

primary human fibroblasts relies primarily on the ability of E7 to activate E2FI and the

I.; AtmlbsllChk/p53 pathway (Figure 4.25).

..1 108

Viral Oncoprotein Inactivation

Rb/E2F E2F1 Chk2 Chk2 p53 Apoptosis

Atm/Nbs1

Figure 4.26. Model of p53 activation following deregulation of E2F1.

Deregulation of the Rb/E2F proliferation pathway results in activation of an apoptotic pathway with some similarities to the apoptosis pathway induced by DNA double strand breaks. Apoptosis induction associated with deregulated E2F activity is dependent on

E2FI and the ability ofE2FI to induce Chk2 and to signal through Atmlbsi resulting in activation of p53 and apoptosis. 109

In this study, we examined phosphorylation of the serine 20 residue on p53 as a marker for Chk2 activation. In normal, AT, and NBS cells, while we observed an increase in total p53 levels, only in normal cells did we observe an increase in the phospho-serine 20 form of p53 , a substrate for active Chk2 kinase. Inhibition of Chk2 activity by a dominant negative construct or by siRNA targeting resulted in a failure to phosphorylate the serine 20 residue following expression of either E2FI or E7 demonstrating the specificity of this modification by Chk.

While we describe the involvement ofE2FI in apoptosis resulting from Rb family inactivation, E2F I also plays a role in apoptosis induction following exposure of cells to

DNA damaging agents (235). We speculate that activation of E2FI by DNA damage

ARF leads to increased pI4 expression resulting in increased pools of p53 protein. E2FI is

also able to activate Atm kinase activity and induce Chk2 expression leading to increased p53 activation and E2FI activity. E2FI activation following DNA damage would therefore act to amplify DNA damage signals converging at p53 to result in apoptosis. 110

CHAPTER V

E2Fl INDUCES Mrell FOCI FORMATION AND BLOCKS CELL CYCLE PROGRESSION IN HUMAN FIBROBLASTS 111

A. Introduetion , we next determined what Having found that Chk2 induction is specific to E2FI upstream signals are specifically activated by E2F1. Activation of p53 following DNA , such as Atm, Atr, and damage involves both activation of DNA damage response kinases , 285 , 372). Chk2, as well as relocalization of DNA damage recognition proteins (I

Among the DNA damage recognition proteins that relocalize to sites of DNA breaks are

Mre11 , Rad50, and NbsI , components of the MRN DNA damage recognition complex

(47 285). Activation of certain S phase cell cycle checkpoints also requires the activity of

Atm (373). The Atm triggered S phase checkpoint requires, among other proteins

functional Nbs I , a component of the MRN complex that is also required for E2F

induced apoptosis and recognition of certain forms of DNA damage (412). In addition to

MRN relocalization following DNA damage, the DNA damage sensors 53BPI and

yH2AX also relocalize to sites at or near DNA breaks (205 , 217, 426). Because of the , we requirement for DNA damage response proteins for E2FI-induced apoptosis

hypothesized that E2FI may induce relocalization of some or all ofthese DNA sensors. 112

B. E2Fl specifieally eauses MRN reloealization

The MRN complex functions in checkpoint signaling by relocalization to discrete , we nuclear foci following certain types of DNA damage (47, 66, 255 , 285). Therefore

examined the cellular localization of the MRN complex following expression of E2F

proteins by examining the Mre II component of the complex. We found that expression

of E2F I induces relocalization of Mre II to discrete nuclear foci, while no foci were

observed following expression of E2F3a (Figure 5. 1 A). MreII foci were observed in

98% of cells following expression of E2FI , but at levels similar to control following

expression ofE2F3a (Figure 5.1 B). Formation of Mrel I foci correlated with an increase

in the phospho-serine 20 form of p53 (Figure 5. 2) and with apoptosis induction (Figure

3). Expression of E2F3a resulted in an increase in the phospho-serine 15 form of p53

(Figure 5.2), a target of the Atm kinase, but not with an increase in the phospho-serine 20

(Figure 5.2) form ofp53 or with apoptosis induction (Figure 5. 3).

C. E2Fl induees irradiation indueed foei (IRIF)-tike formation

The formation of MRN foci has been described under a number of cellular

conditions (241 , 254, 255 , 285 , 437). To determine the type of MRN foci formed

following expression of E2F I , we sought to further characterize the Mre II foci. MRN

foci formation requires the Nbs I component of the complex (47). Similarly, we found

that E2FI-induced Mrell foci fail to form in cells lacking Nbsl (Figure 5.4). DNA

damage foci have also been shown to form independently of Atm (267). Likewise, we ...

113

DAPI Mre11 Merge % Cells Con Virus MOl with Foci Con 2500 ..1%

E2F1 1000 98% E2F1 E2F3a 1000 ..1% E2F3a 1500 ..1% E2F3a 2000 ..1% E2F3 E2F3a 2500 ..1%

Figure 5.1. E2F! specifcally induees Mrell foci formation.

(A) Normal human dermal fibroblasts were infected with AdCon, AdE2FI , or AdE2F3a at an MOl of 1000. Cells were fixed and stained for MreII at 24 hpi. (B) Normal human dermal fibroblasts were infected with AdCon, AdE2FI , or AdE2F3a at the indicated

MOls. Cells were fixed and stained for MreII at 24 hpi. Cells were counted to determine the percentage of cells with MreII foci. A minimum of300 cells per well was counted. 114

p53

actin

Ser15

actin

Ser20

actin

Figure 5.2. Expression of E2Fl, but not E2F3a, results in an increase in the phospho-serine 20 form of p53.

Normal human dermal fibroblasts were infected with AdCon or AdE2FI at an MOI

1000, or AdE2F3a at MOIs of 1000, 1500, 2000, or 2500. Cells were harvested and lysates generated at 24 hpi. Western blot analysis was done to determine levels of total p53 protein, and levels of the phospho-serine 15 and phospho-serine 20 forms of p53.

Actin blots are shown as protein loading controls. 115

Con

Figure 5.3. E2Fl, but not E2F3a, induces apoptosis in human fibroblasts.

Normal human dermal fibroblasts were infected with AdCon or AdE2FI at an MOI

1000, or AdE2F3a at MOIs of 1000, 1500 , 2000, or 2500. Cells were harvested and apoptosis detected at 96 hpi. Error bars represent standard deviation of experiment performed in triplicate......

116

Wildtype NBS DAPI Mre11 Merge DAPI Mre11 Merge

con...

E2F1

Figure 5.4. E2Fl fails to induces Mrell foci in NBS cells.

Normal human dermal fibroblasts or NBS fibroblasts were infected with AdCon or

AdE2FI at an MOI of 1000. Cells were fixed and stained for MreII at 24 hpi. 117

found that E2F I is able to induce the formation Mre II foci in cells lacking Atm (Figure

5). To confirm the involvement of Nbsl in the MreII foci observed following expression of E2F I , we examined the colocalization of Nbs I and Mre II. We found that

Nbsl and MreII co localize to nuclear foci following expression ofE2FI (Figure 5.6 A).

In addition to relocalization of the MRN complex, DNA damage also induces relocalization of other DNA damage response proteins, including yH2AX and 53BPI (9,

205 338 353 426). We found that expression ofE2FI induces relocalization ofyH2AX and 53BPI to nuclear foci, and this correlates with the relocalization of MreII foci

(Figure 5.6 B and Figure 5.6 C). Taken together, these results suggest that ectopic expression of E2F I induces the formation of foci similar to the irradiation induced foci

(IRIF) seen following DNA damage.

MRN complex relocalization has also been observed during normal S phase progression, when no extrinsic DNA damaging agent has been present (254). Since E2FI and E2F3a have been described to induce S phase in rodent fibroblasts (76, 183, 211), we determined if the observed foci could be attributed to S phase progression. A hallmark of the reported S phase MRN foci is their localization at or near sites of BrdU incorporation.

When we examined the subset of cells that incorporate BrdU following expression of

E2FI , we found no colocalization of Mre11 and BrdU (Figure 5.7). Indeed, we oberved

very few BrdU positive cells following expression of E2F1. The rare cells that show

BrdU incorporation following expression of E2FI show no co localization of Mrell and

BrdU (Figure 5.7 bottom panels)......

118

Wildtype DAPI Mre11 Merge DAPI Mre11 Merge

Con

E2F1

Figure 5.5. E2Fl induces Mrell foci formation in AT cells.

Normal human dermal fibroblasts or NBS fibroblasts were infected with AdCon or

AdE2FI at an MOl of 1000. Cells were fixed and stained for Mrell at 24 hpi......

119

eon ..1I1i

E2F1

eon

E2F1

eon ..8.

E2F1

Figure 5.6. E2Fl induces relocalization of DNA damage response proteins.

Normal human dermal fibroblasts were infected with AdCon or AdE2FI at an MOl of

1000. Cells were fixed and stained for Mrell and Nbsl (A), MreII and yH2AX (B), or

MreII and 53BPI (C) at 24 hpi. Merged images are also shown......

120

DAPI BrdU Mre11 Merge

Con

E2F1

E2F1

Figure 5.7. Expression of E2Fl induces Mrell foci that do not correlate with BrdU incorporation. an MOI of Normal human dermal fibroblasts were infected with AdCon or AdE2FI at

1000. Cells were fixed and stained for Mrell and BrdU at 24 hpi. Merged images are also shown...... '- ,

121

DAPI Mre11 Merge

con

E2F1

E2F1 283

Figure 5.8. E2Flt-283 induces Mrell foci formation similar to full length E2Fl.

Normal human dermal fibroblasts were infected with AdCon, AdE2FI , or AdE2Fb-283 at an MOl of 1000. Cells were fixed and stained for MreII at 24 hpi. 122

DAPI M re 11 Merge

Con

E2F1

E2F1

Figure 5.9. E2Fl 132 induces Mrell foci formation similar to E2Fl.

Normal human dermal fibroblasts were infected with AdCon, AdE2FI , or AdE2FII-283 at an MOI of 1000. Cells were fixed and stained for MreII at 24 hpi. 123

It is possible that an interaction between E2FI and Nbsl (254) may provide a mechanism for the observed MRN relocalization following expression of E2FI. The MRN complex has also been shown to relocalize to sites that contain E2F binding elements near origins of DNA replication (254). To examine whether the interaction of E2FI and Nbsl is required for MRN relocalization, we used an E2FI mutant, E2FlJ-283, that lacks the Nbsl interaction domain (254). We found that expression of E2FlJ-283 results in MRN relocalization similar to expression of full length E2FI (Figure 5. 8). Additionally, expression of a DNA binding mutant of E2FI , E2FIe13, was found to induce relocalization of Mrel I (Figure 5.9), suggesting that E2FI is not responsible for targeting the MRN complex to specific sites on DNA. Taken together, these results suggest that expression of E2F I induces foci consistent with IRIF, and not foci correlative of S phase induction.

D. E2Fl blocks eell eycle progression in human fibroblasts, roles for Nbsl and

53BPl

The observed formation ofMRN foci following expression ofE2FI suggests that

E2FI may be activating a cell cycle checkpoint. However, the importance of E2FI and

E2F3a in promoting the transition from G to S phase of the cell cycle has been well documented. Ectopic expression of E2FI or E2F3a in rodent fibroblasts has been shown to promote S phase entry, even under seru deprivation (76, 183 210). However, ectopic

expression of E2FI in human cells has been found to be insufficient for S phase entry,

and can lead to senescence in normal human fibroblasts (83 , 240, 265 379). Following '#

124

Q) :e :J L. J!

Con E2F1 E2F3a

Figure 5.10. E2Fl, butnot E2F3a, bloeks cell eycle progression.

Serum starved HEL fibroblasts were infected with AdCon, AdE2FI , or AdE2F3a at an

MOI of 250. Immediately following infection, cells were returned to normal growth medium. Cells were fixed and stained for BrdU incorporation at 24 hpi. A minimum of

300 cells per well was counted. 125

Con E2F1 E2F3a Ser1981 Atm

Figure 5.11. Expression of E2Fl and E2F3a results in increased levels of the phospho-serine 1981 form of Atm.

HEL fibroblasts were infected with AdCon, AdE2FI , or AdE2F3a at an MOI of 250.

Cells were harvested and lysates generated at 24 hpi. Western blot analysis was performed to determine the levels of the phospho-serine 1981 form of Atm. 126

DNA damage, relocalization of DNA damage response proteins, including the MRN complex and 53BPI , can lead to activation of a cell cycle checkpoint, blocking cell cycle progression (84, III , 185 384 394 423 450). We therefore tested the ability ofE2FI block cell cycle progression. When normal growth medium was added to starved cells at the time of E2F expression, we found that E2FI blocks cells from entering the cell cycle while expression of E2F3a allowed cell cycle progression as measured by BrdU incorporation (Figure 5.9). A block in cell cycle progression is commonly observed following DNA damage when a cell cycle checkpoint is activated to prevent replication of damaged DNA (49). Firing of an S phase checkpoint can involve activation of Atm and downstream signaling proteins (191). We found that expression ofE2FI or E2F3a able to activate Atm, as measured by an increase in the phospho-serine 1981 form of Atm

(Figure 5.10).

Since Atm activation was not unique to E2FI and did not correlate with the observed block in cell cycle progression, we examined the roles of DNA damage signaling proteins Nbsl and 53BPI in preventing cell cycle progression. Nbsl and 53BPI are important in the Atm-dependent S phase checkpoint, and loss of either Nbsl or

53BPI hypersensitizes cells to ionizing radiation and abrogates activation of DNA damage induced cell cycle checkpoints (66, 84, 111 423 427). To determine the roles of

Nbsl and 53BPI in blocking cell cycle progression following expression of E2FI , we used siRNAs to reduce the levels of Nbsl or 53BPI in cells (Figure 5. 11 A). We found that transfection of cells with siNbsi or si53BPI prior to expression of E2FI partially alleviated the block in cell cycle progression (Figure 5. 11 B). Transfection of siNbsi and : '

127

53BP1 Nbs1

actin : actin

siNbs1 a siC on si53BP1 siNbs1 a siCon si53BP1

Con E2F1

Figure 5.12. Roles for Nbsl and 53BPI in blocking eell eycle progression.

(A) HEL fibroblasts were transfected with siRNAs targeted to 53BPI or Nbs1. Cells were

harvested and lysates generated at 48 hpi. Western blot analysis was performed to

determine levels of 53BPI or Nbsl protein. (B) HEL fibroblasts were transfected with

marked siRNAs, serum deprived for 48 hours, and infected with AdCon or AdE2FI at an

MOI of 250. Immediately following infection, cells were returned to normal growth

mediurh Cells were fixed and stained for BrdU incorporation at 24 hpi. A minimum of

300 cells per well was counted. ;,) \) ') " \) ') ,, \)

128

Con E2F1

co ,, co ') co '\') \) 10 ') hpi: \) b- co ,, \) b-co '\')

p21

actin

Figure 5.13. Expression of E2Flieads to aecumulation of p21 protein.

HEL fibroblasts were infected with AdCon or AdE2FI at an MOl of 250. Cells were harvested and lysates generated at indicated hpi. Western blot analysis was performed to

determine levels of p2I protein. Actin blot is shown as protein loading control. ..;,-, " () '"

129

w "en O p21

actin !J'Jlr"'-il"._w

siCon sip21 a siCon sip21a Con E2F1

Figure 5.14. Role for p21 in blocking eell eycle progression.

(A) HEL fibroblasts were transfected with siRNAs targeted to p2I. Cells were harvested

and lysates generated at 48 hpi. Western blot analysis was performed to determine levels

of p2I protein. (B) HEL fibroblasts were transfected with marked siRNAs, serum

deprived for 48 hours, and infected with AdCon or AdE2FI at an MOI of 250.

Immediately following infection, cells were returned to normal growth medium. Cells

were fixed and stained for BrdU incorporation at 24 hpi. A minimum of 300 cells per

well was counted. "- (; " ".""" """' ;::

130

Can E2F1 C Cr r: (' o 1.a.U 0 w Ow " p53 p21 " ,

actin lli actin F"'-"" Q) CJ :J CJ Q) ef

siCon sip53 siCon sip53 Con E2F1

Figure 5.15. Role for p53 in blocking eell cycle progression and p21 aeeumulation.

(A) HEL fibroblasts were transfected with an siRNA targeted to p53. Cells were

harvested and lysates generated at 48 hpi. Western blot analysis was performed to

determine levels of p53 protein. (B) Cells were transfected with marked siRNAs, serum

deprived for 48 hours, and infected with AdCon or AdE2FI at an MOI of 250.

Immediately following infection, cells were returned to normal growth medium. Cells were fixed and stained for BrdU incorporation at 24 hpi. A minimum of 300 cells per

well was counted. (C) Cells were transfected with the marked siRNA 24 hours prior to

infection with AdCon or AdE2Fl at an MOI of 250. Cells were harvested and lysates

generated at 24 hpi. Western blot analysis was performed to determine levels of p2I protein. -=,'.

131

si53BPI together showed no effect on cell cycle progression above transfection of the

single siRNAs (data not shown). This data suggets that Nbsl and 53BPI playa role in

blocking cell cycle progression following expression of E2F I.

E. Roles for p53 and p21 in E2Fl-indueed eell cycle bloek

In the DNA damage response pathway, Nbsl and 53BPI function upstream of

p53 activation(185, 423). We therefore determined ifp53 played a role in the block in

cell cycle progression. We found that treatment of cells with sip53 prior to expression of

E2FI partially alleviated the observed block in cell cycle progression (Figure 5.14 B).

Activation of p53 following certain forms DNA damage can result in activation of a p2I-

dependent cell cycle checkpoint (23 98). We therefore determined ifp2I played a

role in the block in cell cycle progression observed following expression of E2F1. We

found that expression of E2F I in randomly cycling cells resulted in a rapid accumulation

ofp2I protein (Figure 5. 12). Additionally, we found that reducing p2I protein levels in

cells with an siRNA targeted to p2I (Figure 5.13 A) partially alleviated the block in cell

cycle progression observed following expression of E2F I (Figure 5. 13 B). Treatment of

cells with sip21 together with siNbsi and/or si53BPI showed no effect on cell cycle

p2I is a known transcriptional progression above sip2I alone (data not shown). Since

target of p53 (97, 98), we determined if p53 was responsible for the observed induction 14 A) ofp21. We found that treatment of cells with an siRNA targeted to p53 (Figure 5.

reduced the ability ofE2FI to promote accumlation ofp2I (Figure 5.14 C). Treatment of

cells with sip53 together with sip2I , siNbsI , or si53BPI showed no effect on cell cycle 132

progression above sip2I alone (data not shown). Together, these observation suggest that expression of E2F I blocks cell cycle progression in part through its ability of activate p53 resulting in accumulation of p2I protein.

The observation that expression of E2F 1 resulted in a rapid accumulation of p2I protein and a block in cell cycle progression suggests that E2FI is activating a cell cycle checkpoint prior to the induction of apoptosis. To test whether checkpoint activation by

E2FI results in a delay in the apoptotic response, we reduced the levels of 53BPI or p2I in cells with siRNAs and examined the ability of E2FI to induce apoptosis. We found that treatment of cells with si53BPI or sip2I prior to expression of E2FI resulted in an accelerated induction of apoptosis (Figure 5. 15). Taken together, these results suggest that E2Fl is activating a cell cycle checkpoint that delays the induction of apoptosis. 133

70 -

-Can siCon E2F1 siCon C) 40 -Can si53BP1 c: E2F1 si53BP1

Q) 20 -Can sip21 E2F1 sip21 Q)

hpi: 24

Figure 5.16. Reduced levels of 53BP1 of p21 leads to enhanced E2F1-induced apoptosis.

HEL fibroblasts were transfected with the marked siRNA 24 hours prior to infection with

AdCon or AdE2FI at an MOl of 250. Cells were harvested and apoptosis detected at indicated times following infection. Error bars represent standard deviation of experiment performed in trplicate. 134

E. Discussion

In this chapter we describe E2FI-induced relocalization of the DNA damage recognition MRN complex into discrete nuclear foci characteristic of foci observed

following DNA damage. MreII relocalization is found to correlate with relocalization of

the DNA damage response proteins yH2AX and 53BP1. We also find that expression of

E2F I results in a block in cell cycle progression, at least partially dependent on activation

of a cell cycle checkpoint pathway involving NbsI , 53BPI , p53, and p21. Expression of

E2FI results in an increase in p2I protein levels that correlates with the observed block in

cell cycle progression. These data suggest that initially following expression of E2F I , a

growth arrest pathway consisiting ofNbsI , 53BPI , and p2I predominates (Figure 5.

left). The finding that reducing p2I and 53BPI protein levels accelerates apoptosis

suggests that continued expression of E2F I or inactivation of this cell cycle checkpoint

pathway favors activation ofthe apoptosis pathway (Figure 5. 16 right)

Our data show that formation of Mre II-containing foci following expression of

E2FI correlates with relocalization of the DNA damage signaling proteins yH2AX and

53BP1. However, we observe incomplete overlap of these proteins. Colocalization ofthe

DNA damage response proteins MreII and yH2AX has been observed following y-

irradiation (51 , 309). However, these studies examined foci formation at later time points

following y-irradiation when the majority of lesions have been repaired (377). Using a

chromatin extraction protocol, Petrini and colleagues found that MreII and yH2AX foci

do not colocalize, but remain adjacent throughout the course of DNA repair (267, 268).

This is not surprizing given that y H2AX foci can be observed not only at sites of ..

135

Initial E2F1 Activation Sustained E2F1 Activity

E2F1 E2F1

Nbs1 53BP1 Nbs1 53BP1

p53 p53 .. p21

GfO\N\n GroWth p.."ffes\ Arrest

Figure 5.17. Model ofE2Fl-indueed apoptosis and growth arrest.

(Left) Initial expression ofE2FIleads to a block in cell cycle progression through NbsI

53BPI , p53 , and p21. (Right) Continued expression ofE2FIleads to activation of the

apoptosis program. Reduction of 53BPI or p2I protein levels acts to reduce the ability of

E2F I to promote growth arrest and favors the apoptosis pathway. 136

damaged DNA, but also over large portions of a chromosome surrounding DNA damage

(337). Therefore, our data showing incomplete overlap ofE2FI-induced foci is consistent with foci observed following y-irradiation. The question remains as to why DNA damage-like foci form following expression of E2F1. We speculate that ectopic E2FI activity may lead to DNA breaks either directly or as a result of slowing or preventing

DNA repair. The MRN complex and other DNA damage signaling proteins may then recognize these breaks in a manner similar to the DNA damage response, and initiate a signaling pathway resulting in activation of a cell cycle chekpoint or apoptosis.

Our data describe a partial alleviation of the E2FI-induced block in cell cycle progression by siRNA targeting of the p21 checkpoint pathway. While it appears that the p21 pathway is only partially required for this checkpoint, we cannot rule out the possibility that, due to incomplete knockdown in gene expression, that the observed checkpoint is totally dependent on the p2I pathway. However, this seems unlikely due to the undetectable levels of p2I protein remaining after siRNA treatment, and only a modest alleviation of the cell cycle block. It remains unclear as to what additional components may be required for mediating this block. It is possible that p I4AR may be contributing to the observed block in cell cycle progression. pI4ARF has been found to playa role in E2FI-induced senescence (83). However, the role of pI4AR in the E2FI- induced senescence pathway is dependent on p53 function (83). We observe only a partial increase in cell cycle progression following treatment of cells with sip53 suggesting that an additional pathway(s) that are p53 independent may be contributing to the observed growth arrest. One such pathway may involve activation of the cyclin 137

dependent kinase inhibitor pI6ink4a. pI6 plays roles in both growth arrest and senescence by inhibiting Rb phosphorylation (7, 189, 381). The potential role of pI6 in E2FI- mediated growth arrest remains to be determined. :.

138

CHAPTER VI

DISCUSSION

I:i, 139

A. Thesis overview

The goal of this study was to determine the pathway(s) by which deregulation of the Rb growth control pathway results in p53-dependent apoptosis. It had been well established that inactivation of Rb by mutation, gene deletion, or binding by viral oncoproteins results in apoptosis that is often p53- dependent. Mouse models and in vitro evidence suggest that E2FI is the primary mediator linking Rb inactivation to p53 activation and apoptosis (246, 305, 316 408). The prevailing hypothesis ofE2FI-induced apoptosis proposed a central role for the pI9 /Mdm2 pathway in activation ofp53 and apoptosis following deregulation of cellular growth control (25 , 189).

The data presented in this thesis demonstrate that pI ~RF is not required for p53- dependent apoptosis following expression ofE2F1. Instead, we found that E2FI-induced apoptosis requires covalent modifications of p53 by a caffeine sensitive kinase(s). The

DNA damage kinases Atm and Chk2, both of which are sensitive to caffeine, as well as the DNA damage response protein Nbs I are required for E2F I and HPV - 16 E7 to induce

apoptosis. Upregulation of Chk2 is specific to E2FI , and correlates with p53 activation and apoptosis. The data also show that E2FI specifically induces the relocalization of

DNA damage recognition and response proteins including MreII , NbsI , 53BPI , and yH2AX prior to apoptosis. The relocalization of DNA damage sensors correlates with a block in cell cycle progression that is mediated, in part, by activation of a p2I-dependent cell cycle checkpoint. These result suggest that deregulation of E2F I results in activation

of a cell cycle checkpoint prior to the induction of apoptosis. I' '

140

B. E2Fl is the eentral mediator of apoptosis following loss of Rb growth eontrol.

E2F I has been reported to play dual roles as both an oncogene and a tumor suppressor. It has been proposed that the ability of E2F I to act as a tumor suppressor is suppressive mainly due to activation of the apoptosis program (408). The tumor properties of E2FI are revealed in mice lacking E2FI which exhbit defects in apoptosis and display an increase in tumor formation (113 , 449). The role of E2FI in the DNA damage pathway, discussed below, also support the role ofE2FI as a tumor suppressor.

The role of E2FI as an oncogene have been attributed mainly to the ability of E2FI to promote re-entry into the cell cycle. Overexpression of E2F I and activated Ras

in vivo can also primar cells can result in transformation (182). Overexpression ofE2FI

result in an increase in tumor formation and loss of E2F I leads to reduction in tumor

growth in Rb +/- mice (317, 448).

Our data support the roles of E2FI as both an oncogene and a tumor suppressor.

Overexpression of E2FI or inactivation of Rb by the HPV- I6 E7 protein results in

apoptosis, consistant with the role of elevated levels of E2F I acting to suppress

tumorogenesis. Specifically, the data in chapter IV demonstrate that E2FI , and not the

other activator E2F family members, acts as the central mediator of apoptosis induction

in response of loss of Rb growth control. However, our studies also indicate how E2FI

may also function as an oncogene. Overexpression ofE2FI in human fibroblasts leads to

relocalization of DNA damage recognition and response proteins , suggesting that

elevated E2FI activity may lead to an increase in mutation rate and/or genomic

instability, possibly by an accumulation of DNA breaks. 141

It appears as though p53 also plays dual roles in the E2FI signaling pathways.

Accumulation of p2I protein following expression of E2F I is likely due to

transactivation of p2I expression by p53. Indeed, targeting of p53 with an siRNA reduced the ability of E2FI to increase p2I protein levels. Induction of apoptosis by

E2FI also requires activation of p53 through covalent modification by Atm, Chk2, and possibly additional kinases. It remains to be determined if there are differences in p53 activation by E2FI that mediate either a growth arrest or apoptosis.

It is concievable that in normal cells, E2F I has the ability to respond to loss proper growth control and induce a block in cell cycle progression associated with redistribution of DNA damage repair molecules. If proper cell cycle regulation is not returned, sustained E2FI activity would then promote apoptosis. In human cells, the inactivation ofRb by cellular or viral oncoproteins would lead to increased E2FI activity.

E2F I would then activate p2I resulting in activation of cell cycle checkpoint to prevent inappropriate proliferation and tumorogenesis. Activation of p2I could further inhibit

E2FI activity by blocking cellular cyclin-Cdk activt and additional Rb phosphorylation.

However, sustained E2FI activity may bypass the cell cycle arrest, and promote

apoptosis. Therefore, E2F I may act as a transducer of signals resulting from deregulation

of Rb growth control, promoting a block in cell cycle progression to allow for restoration

of proper cell cycle regulation or apoptosis to eliminate the potential for subsequent

transformation of cells. It remains unclear as to why E2F I blocks cell cycle progression

in human fibroblasts but is able to induce S phase in rodent cells, even in the presence of

increased p2I protein levels (77). It is possible that rodent E2FI fails to activate the p53- 142

independent checkpoint pathway suggested in Chapter V. This would be consistent with the observations that rodent fibroblasts fail to undergo senescence as efficiently as human fibroblasts (346, 376).

The importance of the E2F family, and E2FI specifically, in responding to loss of

E2F appropriate cellular proliferation control suggests that genes may be an attractive target for mutation in tumorogenesis. However, while mutations in the Rb growth control pathway are common in tumors, mutations in E2F family members are rarely found in

human tumors. E2F expression has been reported to be either reduced or amplified in some different tumor types (99 106 110 157 177). However, the infrequency ofE2FI mutations identified in human tumors supports the dual roles of E2FI demonstrated our findings. We speculate that mutations resulting in loss of E2F function would be disadvantageous for tumor formation possibly due to a loss in proliferative capacity.

Amplification of E2FI would be similarly disadvantageous due to the ability of E2FI to induce apoptosis when deregulated.

c. Pathways tinking E2F! to p53

1. Role of ARF in E2F signating

Our data presented in chapter III clearly show that distinct pathways mediate p53 accumulation and p53-dependent apoptosis following ecotopic expression of E2FI in rodent fibroblasts. However, in MEFs, mouse models, and human cells pI ~RF AR is dispensible for apoptosis induction by E2FI (236, 340, 342 401 , 409). While pI9 AR not required for E2F I-induced apoptosis, it is likely that activation of the p I9 /Mdm2 ,j .

143

AR in negative pathway influences E2FI signaling. Evidence points to a role for pI9 AR and human pI4AR both physically regulation of cellular growth control. Mouse pI9

interact with E2FI , inhibit E2FI transactivation function, and promote its degradation ARF , expression ofpI9 is able to inhibit (107 251 256). In addition to inhibition ofE2FI

deregulated proliferation associated with expression of the cellular oncoproteins Ras and AR also Myc as well as the viral oncoproteins EIA and E7 (73 , 303 , 306, 463). pI9

physically interacts with DPI , the hertodimeric E2F binding partner (68). The interaction AR and D P , suggesting that between p I9 I is inhibited by coexpression of D P I and E2F I negative DPI-E2FI complexes that exist during the G1- S boundary are resistant to AR (68). Taken together ectopic regulation by p I9 , induction of p I9 , either by

expression of E2F or expression of HPV - 16 E7, would provide negative feedback on the

growth promoting activity ofE2FI.

However, there is a species-specific difference in the activity of mouse pI9 AR in regards to p53 stabilization and cellular growth control. The results and human pI4

presented in chapter III describe the majority of p53 accumulation following expression

of E2FI is dependent on pI9 , while in human fibroblasts, using an siRNA to target (236). The pI4AR had little effect on p53 stabilization following expression of E2FI

human I4AR protein lacks the C-terminal fort amino acids that are present in the mouse

pI9AR protein (25). These C-terminal amino acids have been proposed to be important AR and the peroxisomal targeting chaperone protein for an interaction between p I9 AR may reduce the growth suppressive PexI9p (390). The interaction ofPexI9p with pI9 AR in mouse cells by sequestering pI9AR in the cytoplasm (421). Human activity ofpI9 144

ARF pI4 lacks the C-terminal PexI9p interaction domain, and so has been proposed to

have enhanced growth suppressive properties (421), possibly through mechanisms

independent of p53 (46, 428)

The data presented in chapter V describe a block in cell cycle progression

following expression of E2FI in normal human fibroblasts. The block in cell cycle

progression is due, in part, to the activation of a NbsI- and 53BPI-dependent checkpoint

pathway. Activation of this pathway results in a rapid induction ofp2I protein following

expression of E2F1. Due to the enhanced growth suppressive properties of human pI4 , it is concievable that induction of pI4AR protein following expression of E2FI

could also contribute to the S phase block that is observed in normal human fibroblasts and not in rodent fibroblasts.

The cyclin-dependent kinase inhibitor p I6ink4a may also playa role in the block in cell cycle progression observed following expression of E2F I. p 16 prevents activation of

Cdk4/6, which is thought to be an essential step in Rb inactivation and crucial for the entry into S phase (108, 156). Upregulation ofpI6 protein levels by E2FI would act to maintain Rb in its growth inhibitory state, blocking additional E2F activity and preventing the transition from G to S phase. The importance of the p 16 pathway in controlling deregulated proliferation is suggested by the finding that most, if not all human tumors contain mutations in the cyc1in D/p I6/Rb pathway or in proteins that control this pathway (366). Similar to the function proposed above for ARF, pI6 may also provide negative feedback on the growth promoting activity of E2F I. 145

2. IRIF -like foei formation

Data presented in chapter IV and chapter V suggest that ectopic expression of

E2FI or Rb inactivation by HPV- I6 E7 activates an apoptotis program with similarities to the apoptosis pathway activated by DNA double strand breaks. Both expression of

E2FI and DNA double strand breaks result in relocalization of DNA damage repair and

checkpoint signaling proteins to discrete nuclear foci. In response to y-irradiation or

certain genotoxic agents, proteins, including yH2AX, 53BPI , and the MRN complex

relocalize to sites of actual DNA breaks. MRN and yH2AX were though to be responsible

for the initial recognition of DNA breaks, with the MRN complex functioning to initiate

repair and yH2AX functioning to recruit other DNA repair factors as well as 53BPI to

activate a cell cycle checkpoint. However, recent evidence suggests that the mediator of

DNA damage checkpoint protein I (Mdcl) is required for MRN, yH2AX, and 53BPI foci

formation (127 , 242, 271 , 385, 445) as well as for effcient Chk activation following y-

irradiation (242 , 311 , 445). Inhibiting Mdc I expression with an siRNA results in

decreased p53 stabilization and a failure to activate a cell cycle checkpoint following

irradiation (127 242 385).

The mechanism of MRN, yH2AX, and 53BPI foci formation following

expression of E2F I remains unclear. It has been proposed that E2F I may target the MRN

complex to DNA by virtue of an interaction with the Nbsl component of the complex

(254). This scenerio seems unlikely due to the foci formation observed following

expression of E2F 1 283, an E2F I mutant lacking the Nbs I interaction domain.

Additionally, no interactions between E2FI and either yH2AX or 53BPI have been 146

described. It is possible that E2FI could direct MRN, yH2AX, and 53BPI foci formation through interaction with Mdc I , if such an interaction exists. Another possibility of how

E2FI induces DNA damage foci is that ectopic expression ofE2FI prevents the repair of

DNA damage that may arise during replication or induces DNA damage by an unown mechanism prior to activation of the apoptosis program. E2FI-induced DNA damage

would then recruit DNA damage repair and checkpoint signaling proteins resulting in p53

activation and either apoptosis or the growth arrest described in chapter V.

The possibility exists that expression of E2FI could result in DNA breaks

indirectly through the induction of reactive oxygen species (ROS). Accumulation ofROS

can lead to the induction of apoptosis through several mechanisms, including direct

Myc caspase activation (41). Expression of the oncogene results in ROS accumulation

that contributes to the DNA breaks and MreII relocalization observed following

expression of c-Myc (413). Together with the observations that expression of E2FI can

induce ROS accumulation and that c-Myc induced apoptosis is compromised in E2Fr

MEFs suggests that c-Myc induced accumulation ofROS, and subsequent DNA damage

may be a consequence of E2FI activation (228, 397). Therefore, it is possible that ROS-

induced DNA damage contributes to the observed relocalization of DNA damage sensors

following expression of E2F!. The failure of expression of E2F3a to result in

relocalization of DNA damage sensors may then be due to a failure to cause ROS

accumulation. The contribution of ROS accumulation to E2FI-induced apoptosis, and

E2F signaling, remains to be determined. 147

The findings in chapter V that expression of E2F I in human cells results in a block in cell cycle progression coupled with the ability of E2FI to induce DNA replication in rodent cells, suggests that E2FI may be activating a DNA replication fork arrest. Replication fork arrest occurs in response to perturbations in many aspects of

DNA replication including insufficient pools of dNTPs, polymerase inhibition, DNA modifications, or DNA breaks that may arise during replication (300). It is possible that constitutive expression of E2FI induces DNA damage as a consequence of stalled replication forks, and results in recruitment of recognition and repair complexes. This seems unlikely, as E2F3a induces inappropriate S phase but fails to induce foci formation or checkpoint activation. Additionally, stalled replication forks activate p53 through an

Atr/ChkI-dependent pathway (22), contrary to the Atm/Chk2 pathway required for E2FI to activate p53.

3. Role of Atm in E2F signating

Atm serves as the primary link between DNA damage sensors and activation of cell cycle regulatory proteins. In undamaged cells, Atm exists as an inactive dimer.

Rapidly following DNA double strand breaks Atm undergoes intermolecular autophosphorylation at the serine 1981 residue resulting in dimer dissociation and an increase in Atm kinase activity (17). Atm phosphorylation is thought to be triggered by chromatin changes associated with an influx of DNA repair factors to sites of DNA damage (17). It remains unclear as to how Atm is activated following expression of

E2FI , E2F2, E2F3a, or HPV- I6 E7. We speculate that expression ofE2F proteins or E7 148

results in chromatin changes associated with induction of S phase or activation of DNA damage response proteins. In the case of HPV - 16 E7, activation of Atm may be a result of chromosomal structural changes and DNA breaks that occur following E7 expression

(86). However, since activation of Atm by E2FI results in apoptosis, we canot rule out the possibility that the different E2F family members activate Atm by distinct mechanisms.

Interestingly, Atm was found not to be required for apoptosis resulting from Rb inactivation in murine brain choroid plexus epithelium (233). In this model, apoptosis resulting from Rb inactivation is also dependent on E2FI and p53 , and occurs in the

absence of pI ~RF (408 , 409). While it is not apparent why this apoptosis is Atm- independent in the choroid plexus epithelium , p53-dependent apoptosis that is Atm- independent has been described in certain cell types (21 , 151). Alternatively, these observations suggest that there may be a species-specific bias for Atm in the E2FI-

I- mediated apoptosis pathway. Indeed, the reduction in apoptosis observed in atm MEFs is not as dramatic as that seen in human dermal fibroblasts following E2FI expression.

We speculate that in the murine system other signaling pathways such as Atr/ChkI may

compensate for the loss of Atm, whereas a more stringent requirement for Atm in human

dermal fibroblasts is observed. Therefore, there may be both cell-type and species-

specific requirements for Atm in apoptosis induction, and it is possible that other Atm-

related kinases may compensate for the loss of Atm function in some cells. --

149

viral oncoproteins Rb Inactivation DNA damage

E2F3a Rb/E2F E2F2 S Phase -- L E2F1

;iF

Apoptosis Growth Arrest

Figure 6.1. Model of E2F signaling network.

Inactivation of Rb durng a normal cell cycle leads to activation of E2FI , E2F2, and

E2F3a resulting in cell cycle progression (shown in green). Inapproprite Rb inactivation by mutation or viral oncoproteins can lead to activation of two distinct, E2FI-specific pathways. The apoptosis pathway (shown in red) signals to p53 through NbsI , Atm, and Chk resulting in p53 covalent modification and apoptosis. The checkpoint pathway

(shown in blue) signals, in part, through NbsI , 53BPI , p53, and p21. The checkpoint pathway also involves a signaling pathway independent ofp53 and p2I through an, as of yet, undefined mediator. AR may playa role in E2FI signaling by increasing pools of p53 protein. 150

4. Aetivation of p53 for the induction We have defined a requirement for covalent modification ofp53 of apoptosis following expression of E2F1. Activation of p53 by covalent modification mouse) and serine 20 in specifically phosphorylation of serine 15 (serine 18 in the response to DNA damage, has been a topic of great interest due to the location of these residues within the Mdm2 interaction domain of p53. Mutational analysis of p53 sites serine 20, have been known to be covalently modified, including serine 15 and anyone of these sites for inconsistent regarding the requirements for phosphorylation at 441). The observation p53 dependent apoptosis in response to DNA damage (13 411

that expression of E2FI , 2, and 3a results in phosphorylation of p53 at the serine

residue suggests that some p53 modifications may occur as a result of ectopic induction

of S phase. Chromatin alterations that occur as a consequence of DNA replication may be 15 form ofp53. sufficient to activate Atm, leading to an increase in the phospho-serine

This initial modification of p53 at the serine 15 residue during the onset of S phase would

be insufficient for activation of the apoptosis program, but may prime p53 for additional , occur modifications should errors, including DNA damage or stalled replication forks

during DNA replication.

While our study examined the phosphorylation status of two N-terminal residues

on p53 known to be phosphorylated following DNA damage, we have not established

that either of these modifications are causal in p53 mediated apoptosis, nor have we ruled

out the involvement or importance ofmodification(s) to other residues on p53 following

E2FI expression. However, our finding that apoptosis is reduced when E2FI is 151

coexpressed with a p53 mutant lacking many of the known phosphorylation sites demonstrates a contribution by potential p53 phosphorylation targets to E2FI-induced apoptosis.

C. Model of E2F! apoptosis pathway

Expression ofE2FIleads to relocalization of DNA damage repair and recognition proteins, including the MRN complex and 53BPI , to discrete nuclear foci. The molecular events leading to relocalization of these components remains to be determined.

Activation of Atm by autophosphorylation is proposed to occur following relocalization of DNA damage sensors by, as yet, an unkown mechanism (17). Activated Atm is then able to phosphorylate p53 at the serine 15 residue. However, expression of E2F I , E2F2 and E2F3a results in Atm activation and phosphorylation of the serine 15 residue on p53.

Unique to E2FI is the ability to induce Chk2 mRNA and proteins levels. Chk is able to phosphorylate the serine 20 residue, and this modification is found to correlate with

E2FI-induced apoptosis. Chk2 may be limiting in this pathway, and when provided in trans Chk enhances E2FI-induced apoptosis and allows E2F2 to induce apoptosis. It is

ARF possible that E2FI , E2F2 , or E2F3a could also signal throughpI4 /Mdm2 to increase p53 protein levels and that the increased pools of p53 would provide more substrate for

Atm andChk (Figure 6. 1). 152

D. Summary

The dual functions of E2F I in checkpoint activation and apoptosis identified in this dissertation provide novel insight into the function of Rb/E2F growth control both in normal cells and in cells containing mutations in the Rb/E2F growth control pathway.

The genomic integrity of mamalian cells is subject to constant attack, either by external damaging agents, or internal mutagens such as errors in DNA replication. If mutations arise that compromise proper Rb-mediated growth control, our results suggest that E2FI may initiate a signaling cascade to activate a cell cycle checkpoint. If proper growth control cannot be restored, continued E2FI activity may lead to apoptosis, preventing an accumulation of mutations that may lead to transformation. The importance ofE2FI as a signal transducer of loss of proper Rb-mediated proliferation control can be observed in

mouse models. Loss of E2F I in Rb mouse models results in a decrease in S phase and a

decrease in apoptosis in the central nervous system and choroid plexus, suggesting an

important role for the E2FI apoptotic pathway in preventing tumor formation (305 408).

The obervation that adult mice lacking E2F I develop tumors (449) also suggests that the

E2F I signaling network is important in suppressing tumor formation.

However, our data may point to a role for an E2FI-specific checkpoint activation

E2F I fails in these mouse models. It is possible that loss of Rb function in mice lacking

to activate the E2FI-induced cell cycle checkpoint pathway described in Chapter V.

Since E2F3a, and possibly E2F2, cannot compensate for E2FI in this pathway, cells may

fail to properly block cell cycle progression and this may lead to additional mutations.

Compounding the loss of the E2F I growth control checkpoint would be the failure to /-

153

, loss of E2FI would lead to an activate the E2FI-induced apotosis pathway. Therefore inability to respond to loss of Rb-mediated growt control.

Although we have defined a pathway linking deregulated E2FI activity to p53 Atm/Chk2/p53 and apoptosis, integration of E2FI signaling and activation of the

I in apoptosis pathway also offers a mechanism for the proposed involvement of E2F resulting from DNA damage. Following treatment of cells with DNA damaging agents

E2FI protein accumulates (30, 158, 165 235 266 294), and is phosphorylated at an N- terminal Atm recognition sequence that is unique to E2FI among the E2F family members (235). This phosphorylation of E2FI is largely dependent on Atm and is required for efficient E2FI stabilization following DNA damage (235). Chk2 has also , and this been shown to phosphorylate and stabilize E2FI following DNA damage modification has been shown to be required for E2F I dependent apoptosis following

DNA damage by altering its promoter specificity (383). Additionally, DNA damage E2Fr mice (235), suggesting induced apoptosis is compromised in thymocytes from

that E2FI has multiple roles in DNA damage signaling. We speculate that activation of

ARF E2FI by DNA damage leads to increasedpI4 expression resulting in increased pools

induce Chk2 of p53 protein. E2FI is also able to activate Atm kinase activity and

expression leading to increased p53 activation and E2FI activity. E2FI activation

following DNA damage would therefore act to amplify DNA damage signals converging

at p53 to result in either a growth arrest or apoptosis (Figure 6. 1). 154

E. Future directions While we have made significant advances toward our understanding of E2FI

signaling, a number of questions require further exploration. How does expression of

E2FI result in relocalization of DNA damage response factors? If DNA damage foci

form following DNA damage, what is causing the DNA damage following expression of

E2FI? The finding that E2FI activates both a cell cycle checkpoint and an apoptotic

response suggests that there may be a molecular switch between the divergent outcomes.

How the cell decides which pathway to favor remains unclear. A divergence in E2FI

function was also found between human and rodent cells. It remains to be determined

why E2FI can induce S phase in rodent cells, but blocks cell cycle progression in human

fibroblasts. Given the additional mechanisms of proliferation and checkpoint control

described in this dissertation, E2FI may function in multiple pathways in the cellular

response to DNA damage.

Lastly, the exact mechanisms that control E2F specificity remam to be

determined. The specific induction of Chk2 by E2F I suggests that transcriptional control

of pro-apoptotic , and possibly anti-apoptotic , genes defines the primary difference

between the activator E2F family members in the apoptosis pathway. Specificity for

E2FI-mediated transactivation of the subset of E2F responsive genes involved in

apoptosis would provide an attractive mechanism for the initiation of the apoptosis

program. However, transcriptional activation or derepression, while required for

apoptosis, does not account for all of the observed consequences of E2F I expression. The

difference between E2FI and E2F2 or E2F3a in the formation of DNA damage-like 155

be important for Mre II foci suggests that additional E2F I-specific domains may mediating this response. Specificity for E2F family members inactivation of the apoptotic pathway would therefore depend on multiple domains that are unique to E2FI and serve specific fuctions in activation of components of the pathway. r-,

156

CHAPTER VII

APPENDIX A 157

5 12

ug: pC DNA pCDNA E2F1

Figure AI. E2F! induces apoptosis.

HEL fibroblasts were transfected with the indicated amounts of either pCDNA or pCDNA-E2F1. Cells were harvested and apoptosis detected at 96 hpi. Error bars repesent standard deviation of experiment performed in triplicate. 158

is 2

siCon siChk2b siCon siChk2b

pCDNA pCDNA E2F1

Figure A2. siChk2 inhibits E2Fl-indueed apoptosis.

HEL fibroblasts were cotransfected with the marked siRNA together with either pCDNA or pCDNA-E2F1. Cells were harvested and apoptosis detected at 96 hpi. Error bars represent standard deviation of experiment performed in triplicate. 159

E2F1

E2F1 actin .n8

Figure A3. DNChkl and DNChk2 do not effect E2Fl expression.

Normal human fibroblasts were coinfected with AdDNChkI or AdDNChk at an MOI of

1000 and with AdE2FI at an MOI of 1000. Cells were harvested and lysates generated at

24 hpi. Western blot analysis was performed to determine levels of E2F I protein. Actin blots are shown as protein loading controls. - ,=-'" - ..=.

160

c: 7 2 6

g' 5

0: 2

Con E7 Con E7 Con E7 Con E7 Con E7 Con E7 siE2F3a siE2F3b siCon siE2F1a siE2F2a siE2F2b

Figure A4. Specific requirement for E2Fl in HPV-l6 E7 indueed apoptosis. infectioQ. HEL fibroblasts were transfected with the indicated siRNA 24 hours prior to with AdCon or AdE7 at an MOI of 1000. Cells were harvested and apoptosis detected at

96 hpi. Error bars represent standard deviation of experiment performed in triplicate. 161

CHAPTER VIII

REFERENCES 162

Abraham , R. T. 2001. Cell cycle checkpoint signaling through the ATM and ATR kinases. Genes Dev 15:2177-96. Adams, J. M. 2003. Ways of dying: multiple pathways to apoptosis.

Genes Dev 17:2481-95.

Adams, M. R., R. Sears, F. Nuckolls, G. Leone, and J. R. Nevins. 2000.

Complex transcriptional regulatory mechanisms control expression of the

E2F3 locus. Mol Cell Bioi 20:3633-

Adams, P. D., and W. G. Kaelin, Jr. 1995. Transcriptional control by E2F.

Semin Cancer Bioi 6:99-108.

Adnane , J. , Z. Shao, and P. D. Robbins. 1995. The retinoblastoma

susceptibility gene product represses transcription when directly bound to

the promoter. J Bioi Chem 270:8837-43.

Ahn, J., and C. Prives. 2002. Checkpoint Kinase 2 (Chk2) Monomers or Dimers Phosphorylate Cdc25C after DNA Damage Regardless of

Threonine 68 Phosphorylation. J Bioi Chem 277:48418-26.

Alcorta, D. A., Y. Xiong, D. Phelps, G. Hannon, D. Beach, and J. C.

Barrett. 1996. Involvement of the cyclin-dependent kinase inhibitor p16

(INK4a) in replicative senescence of normal human fibroblasts. Proc Natl

Acad Sci USA 93:13742-

Alland, L., R. Muhle, H. Hou, Jr., J. Potes, L. Chin, N. Schreiber-Agus,

and R. A. DePinho. 1997. Role for N-CoR and histone deacetylase in

Sin3-mediated transcriptional repression. Nature 387:49-55. 163

Anderson, L., C. Henderson , and Y. Adachi. 2001. Phosphorylation and rapid relocalization of 53BP1 to nuclear foci upon DNA damage. Mol Cell Bioi 21:1719-29.

10. Appella , E. , and C. W. Anderson. 2001. Post-translational modifications and activation of p53 by genotoxic stresses. Eur J Biochem 268:2764- 72.

11. Arellano, M. , and S. Moreno. 1997. Regulation of CDKlcyclin complexes during the cell cycle. Int J Biochem Cell Bioi 29:559- 73. 12. Arroyo, M., and P. Raychaudhuri. 1992. Retinoblastoma-repression of E2F-dependent transcription depends on the ability of the retinoblastoma protein to interact with E2F and is abrogated by the adenovirus E1A oncoprotein. Nucleic Acids Res 20:5947- 54. 13. Ashcroft, M., M. H. Kubbutat, and K. H. Vousden. 1999. Regulation of p53 function and stability by phosphorylation. Mol Cell Bioi 19:1751-

14. Ashkenazi, A. 2002. Targeting death and decoy receptors of the tumour-

necrosis factor superfamily. Nat Rev Cancer 2:420- 30.

15. Attardi , L. D. , S. W. Lowe , J. Brugarolas , and T. Jacks. 1996. Transcriptional activation by p53 , but not induction of the p21 gene , is essential for oncogene-mediated apoptosis. Embo J 15:3693- 701.

16. Bagchi , 5. , R. Weinmann , and P. Raychaudhuri. 1991. The retinoblastoma protein Copurifies with E2F- , an E1A-regulated inhibitor of the transcription factor E2F. Cell 65:1063- 72. 164

17. Bakkenist, C. J., and M. B. Kastan. 2003. DNA damage activates A through intermolecular autophosphorylation and dimer dissociation.

Nature 421 :499-506.

18. Bandara, L. R., V. M. Buck, M. Zaman ian, L. H. Johnston, and N. B. La

Thangue. 1993. Functional synergy between DP-1 and E2F-1 in the cell

cycle-regulating transcription factor DRTF1/E2F. Embo J 12:4317-24.

19. Bandara, L. R., and N. B. La Thangue. 1991. Adenovirus E 1 a prevents the retinoblastoma gene product from complexing with a cellular

transcription factor. Nature 351 :494-

20. Banin, S., L. Moyal, S. Shieh, Y. Taya, C. W. Anderson, L. Chessa, N. I. Smorodinsky, C. Prives, Y. Reiss, Y. Shiloh, and Y. Ziv. 1998.

Enhanced phosphorylation of p53 by A TM in response to DNA damage.

Science 281 :1674-

21. Barlow, C., K. D. Brown, C. X. Deng, D. A. Tagle, and A. Wynshaw-

Boris. 1997. Atm selectively regulates distinct p53-dependent cell-cycle

checkpoint and apoptotic pathways. Nat Genet 17:453-

22. Bartek, J., and J. Lukas. 2003. Chk1 and Chk2 kinases in checkpoint

control and cancer. Cancer Cell 3:421-

23. Bartek, J., and J. Lukas. 2001. Mammalian G1- and S-phase

checkpoints in response to DNA damage. Curr Opin Cell Bioi 13:738-47.

24. Bartek, J., and J. Lukas. 2001. Pathways governing G1/S transition and

their response to DNA damage. FEBS Lett 490:117-22. 165

25. Bates, S., A. C. Philips, P. A. Clark, F. Stott, G. Peters, R. L. Ludwig,

and K. H. Vousden. 1998. p14ARF links the tumour suppressors RB and

p53. Nature 395: 124-

26. Beijersbergen, R. L., R. M. Kerkhoven, L. Zhu, L. Carlee, P. M. Voorhoeve, and R. Bernards. 1994. E2F- , a new member of the E2F

gene family, has oncogenic activity and associates with p107 in vivo.

Genes Dev 8:2680-90.

27. Berezutskaya, E., B. Yu, A. Morozov, P. Raychaudhuri, and S. Bagchi.

1997. Differential regulation of the pocket domains of the retinoblastoma

family proteins by the HPV16 E7 oncoprotein. Cell Growth Differ 8:1277-

86.

28. Bergh, G., M. Ehinger, I. Olsson, S. E. Jacobsen, and U. Gullberg.

1999. Involvement of the retinoblastoma protein in monocytic and

neutrophilic lineage commitment of human bone marrow progenitor cells.

Blood 94:1971-

29. Blasina, A., B. D. Price, G. A. Turenne, and C. H. McGowan. 1999.

Caffeine inhibits the checkpoint kinase ATM. Curr Bioi 9:1135-

30. Blattner, C. , A. Sparks, and D. Lane. 1999. Transcription factor E2F-1 is

upregulated in response to DNA damage in a manner analogous to that of

p53. Mol Cell Bioi 19:3704- 13.

31. Boshart, M., L. Gissmann, H. Ikenberg, A. Kleinheinz, W. Scheurlen, and H. zur Hausen. 1984. A new type of papillomavirus DNA , its 166

presence in genital cancer biopsies and in cell lines derived from cervical

cancer. Embo J 3:1151-

Hyde, W. 32. Bouchard, C., K. Thieke, A. Maier, R. Saffrich, J. Hanley- Ansorge, S. Reed, P. Sicinski, J. Bartek, and M. Eilers. 1999. Direct

induction of cyclin 02 by Myc contributes to cell cycle progression and

sequestration of p27. Embo J 18:5321-33.

33. Boyer, S. N., D. E. Wazer, and V. Band. 1996. E7 protein of human

papilloma virus- 16 induces degradation of retinoblastoma protein through

the ubiquitin-proteasome pathway. Cancer Res 56:4620-

34. Brehm, A., E. A. Miska, D. J. McCance, J. L. Reid, A. J. Bannister, and

T. Kouzarides. 1998. Retinoblastoma protein recruits histone deacetylase

to repress transcription. Nature 391: 597 -601.

35. Brown, A. L., C. H. Lee, J. K. Schwarz, N. Mitiku, H. Piwnica-Worms, and J. H. Chung. 1999. A human Cds1-related kinase that functions

downstream of A TM protein in the cellular response to DNA damage. Proc

Natl Acad Sci USA 96:3745-50.

36. Brown, E. J., and D. Baltimore. 2000. ATR disruption leads to

chromosomal fragmentation and early embryonic lethality. Genes Dev

14:397-402.

37. Bruce, J. L., R. K. Hurford, Jr., M. Classon , J. Koh, and N. Dyson.

2000. Requirements for cell cycle arrest by p161NK4a. Mol Cell 6:737-42. 167

38. Buck, V., K. E. Allen, T. Sorensen, A. Bybee, E. M. Hijmans, P. Voorhoeve, R. Bernards, and N. B. La Thangue. 1995. Molecular and

functional characterisation of E2F- , a new member of the E2F family.

Oncogene 11:31-

39. Burd, E. M. 2003. Human papillomavirus and cervical cancer. Clin

Microbiol Rev 16:1-17.

40. Buscemi, G., C. Savio, L. Zannini, F. Micciche, D. Masnada, M.

Nakanishi, H. Tauchi, K. Komatsu, S. Mizutani, K. Khanna, P. Chen, P. Concannon, L. Chessa, and D. Delia. 2001. Chk2 activation

dependence on Nbs1 after DNA damage. Mol Cell Bioi 21 :5214-22.

41. Cai , J. , J. Yang, and D. P. Jones. 1998. Mitochondrial control of

apoptosis: the role of cytochrome c. Biochim Biophys Acta 1366: 139-49.

42. Campanero, M. R., and E. K. Flemington. 1997. Regulation of E2F

through ubiquitin-proteasome-dependent degradation: stabilization by the

pRB tumor suppressor protein. Proc Natl Acad Sci USA 94:2221-

43. Canman, C. E., D. S. Lim, K. A. Cimprich, Y. Taya, K. Tarnai, K. Sakaguchi, E. Appella, M. B. Kastan, and J. D. Silciano. 1998.

Activation of the A TM kinase by ionizing radiation and phosphorylation of

p53. Science 281: 1677 -

44. Carnac, G. , L. Fajas, A. L' Honore, C. Sardet, N. J. Lamb, and A.

Fernandez. 2000. The retinoblastoma-like protein p130 is involved in the 168

determination of reserve cells in differentiating myoblasts. Curr Bioi

10:543-

45. Carnero, A. , and G. J. Hannon. 1998. The INK4 family of CDK inhibitors.

Curr Top Microbiollmmunol 227:43-55.

46. Carnero, A., J. D. Hudson, C. M. Price, and D. H. Beach. 2000.

p161NK4A and p19ARF act in overlapping pathways in cellular

immortalization. Nat Cell Bioi 2:148-55.

47. Carney, J. P., R. S. Maser, H. Olivares, E. M. Davis, M. Le Beau, J. R.

Yates, 3rd, L. Hays, W. F. Morgan, and J. H. Petrini. 1998. The

hMre11/hRad50 protein complex and Nijmegen breakage syndrome:

linkage of double-strand break repair to the cellular DNA damage

response. Cell 93:477-86.

48. Caspari, T. 2000. How to activate p53. Curr BioI10:R315-

49. Caspari, T., and A. M. Carr. 2002. Checkpoints: how to flag up double-

strand breaks. Curr Bioi 12:R105-

50. Castilo, J. P., A. D. Yurochko, and T. F. Kowalik. 2000. Role of human

cytomegalovirus immediate-early proteins in cell growth control. J Virol

74:8028-37.

51. Celeste, A., S. Petersen, P. J. Romanienko, o. Fernandez-Capetilo, H. T. Chen, O. A. Sedelnikova, B. Reina-San-Martin, V. Coppola, E. Meffre, M. J. Difilippantonio, C. Redon, D. R. Pilch, A. Olaru, M.

Eckhaus, R. D. Camerini-Otero, L. Tessarollo, F. Livak, K. Manova, W. 169

M. Bonner, M. C. Nussenzweig, and A. Nussenzweig. 2002. Genomic

instability in mice lacking histone H2AX. Science 296:922-

52. Chaturvedi , P. , W. K. Eng, Y. Zhu, M. R. Mattern , R. Mishra, M. R.

Hurle, X. Zhang, R. S. Annan , Q. Lu, L. F. Faucette, G. F. Scott, X. Li,

S. A. Carr, R. K. Johnson, J. D. Winkler, and B. B. Zhou. 1999.

Mammalian Chk2 is a downstream effector of the A TM-dependent DNA

damage checkpoint pathway. Oncogene 18:4047-54.

53. Chehab, N. H., A. Malikzay, M. Appel , and T. D. Halazonetis. 2000.

Chk2/hCds1 functions as a DNA damage checkpoint in G(1) by stabilizing

p53. Genes Dev 14:278-88.

54. Chellappan, S., V. B. Kraus, B. Kroger, K. Munger, P. M. Howley, W.

C. Phelps, and J. R. Nevins. 1992. Adenovirus E1A, simian virus 40

tumor antigen , and human papillomavirus E7 protein share the capacity to

disrupt the interaction between transcription factor E2F and the

retinoblastoma gene product. Proc Natl Acad Sci USA 89:4549-53.

55. Chellappan , S. P. , S. Hiebert, M. Mudryj, J. M. Horowitz, and J.

Nevins. 1991. The E2F transcription factor is a cellular target for the RB

protein. Cell 65: 1 053-61.

56. Chen F. C. Hung, L. Fromm, and P. A. Overbeek. 2000. Induction of

cell cycle entry and cell death in postmitotic lens fiber cells by

overexpression of E2F1 or E2F2. Invest Ophthalmol Vis Sci 41 :4223-31. 170

57. Chen, X., L. J. Ko, L. Jayaraman, and C. Prives. 1996. p53 levels functional domains, and DNA damage determine the extent of the

apoptotic response of tumor cells. Genes Dev 10:2438-51.

58. Chow, K. N., and D. C. Dean. 1996. Domains A and B in the Rb pocket

interact to form a transcriptional repressor motif. Mol Cell Bioi 16:4862-

59. Chow, K. N., P. Starostik, and D. C. Dean. 1996. The Rb family contains

a conserved cyclin-dependent-kinase-regulated transcriptional repressor

motif. Mol Cell Bioi 16:7173-81.

60. Clarke, A. R., E. R. Maandag, M. van Roon, N. M. van der Lugt, M. van

der Valk, M. L. Hooper, A. Berns, and H. te Riele. 1992. Requirement

for a functional Rb-1 gene in murine development. Nature 359:328-30.

61. Classon, M., B. K. Kennedy, R. Mulloy, and E. Harlow. 2000. Opposing

roles of pRB and p107 in adipocyte differentiation. Proc Natl Acad Sci U S

A 97: 1 0826-31 .

62. Cobrinik, D., M. H. Lee, G. Hannon, G. Mullgan, R. T. Bronson, Dyson, E. Harlow, D. Beach, R. A. Weinberg, and T. Jacks. 1996. Shared role of the pRB-related p130 and p107 proteins in limb

development. Genes Dev 10: 1633-44.

63. Coqueret, o. 2003. New roles for p21 and p27 cell-cycle inhibitors: a

function for each cell compartment? Trends Cell Bioi 13:65-70.

64. Costanzo, V., K. Robertson, M. Bibikova, E. Kim, D. Grieco, M. Gottesman, D. Carroll, and J. Gautier. 2001. Mre11 protein complex 171

prevents double-strand break accumulation during chromosomal DNA

replication. Mol Cell 8:137-47.

65. Cress, W. D., and J. R. Nevins. 1996. Use of the E2F transcription factor

by DNA tumor virus regulatory proteins. Curr Top Microbiol Immunol

208:63-78.

66. Amours, D., and S. P. Jackson. 2002. The Mre11 complex: at the

crossroads of dna repair and checkpoint signalling. Nat Rev Mol Cell Bioi

3:317-27.

67. Dannenberg, J. H., A. van Rossum, L. Schuijff, and H. te Riele. 2000. Ablation of the retinoblastoma gene family deregulates G(1) control causing immortalization and increased cell turnover under growth-

restricting conditions. Genes Dev 14:3051-64.

68. Datta, A., A. Nag, and P. Raychaudhuri. 2002. Differential regulation of

E2F1 , DP1 , and the E2F1/DP1 complex by ARF. Mol Cell Bioi 22:8398-

408.

69. de Bruin, A., B. Maiti, L. Jakoi, C. Timmers, R. Buerki, and G. Leone.

2003. Identification and characterization of E2F7 , a novel mammalian E2F

family member capable of blocking cellular proliferation. J Bioi Chem

278 :42041-

70. de Jager, M., J. van Noort, D. C. van Gent, C. Dekker, R. Kanaar, and

C. Wyman. 2001. Human Rad50/Mre11 is a flexible complex that can

tether DNA ends. Mol Cell 8:1129-35. 172

71. de Klein, A., M. Muijtjens, R. van Os, Y. Verhoeven, B. , A. Carr, A. R. Lehmann , and J. H. Hoeijmakers. 2000. Targeted disruption

of the cell-cycle checkpoint gene A TR leads to early embryonic lethality in

mice. Curr Bioi 10:479-82.

72. de la Luna, S., M. J. Burden, C. W. Lee, and N. B. La Thangue. 1996.

Nuclear accumulation of the E2F heterodimer regulated by subunit

composition and alternative splicing of a nuclear localization signal. J Cell

Sci 109 ( pt 10):2443-52.

73. de Stanchina, E., M. E. McCurrach, F. Zindy, S. Y. Shieh, G. Ferbeyre, A. V. Samuelson, C. Prives, M. F. Roussel, C. J. Sherr, and S. W.

Lowe. 1998. E1A signaling to p53 involves the p19(ARF) tumor

suppressor. Genes Dev 12:2434-42.

74. DeCaprio! J. A., J. W. Ludlow, J. Figge, J. Y. Shew, C. M. Huang, W.

H. Lee, E. Marsilio, E. Paucha, and D. M. Livingston. 1988. SV40 large tumor antigen forms a specific complex with the product of the

retinoblastoma susceptibility gene. Cell 54:275-83.

75. DeGregori, J., T. Kowalik, and J. R. Nevins. 1995. Cellular targets for

activation by the E2F1 transcription factor include DNA synthesis- and

G1/S-regulatory genes. Mol Cell Bioi 15:4215-24.

76. DeGregori, J., G. Leone, A. Miron, L. Jakoi, and J. R. Nevins. 1997.

Distinct roles for E2F proteins in cell growth control and apoptosis. Proc

Natl Acad Sci USA 94:7245-50. , .

173

77. DeGregori, J., G. Leone, K. Ohtani, A. Miron, and J. R. Nevins. 1995.

E2F- 1 accumulation bypasses a G1 arrest resulting from the inhibition of

G1 cyclin-dependent kinase activity. Genes Dev 9:2873-87.

78. Dell, G. , and K. Gaston. 2001. Human papillomaviruses and their role in

cervical cancer. Cell Mol Life Sci 58: 1923-42.

79. Demers, G. W., E. Espling, J. B. Harry, B. G. Etscheid, and D. A.

Galloway. 1996. Abrogation of growth arrest signals by human papillomavirus type 16 E7 is mediated by sequences required for

transformation. J Virol 70:6862-

80. Demers, G. W., S. A. Foster, C. L. Halbert, and D. A. Galloway. 1994.

Growth arrest by induction of p53 in DNA damaged keratinocytes is bypassed by human papillomavirus 16 E7. Proc Natl Acad Sci USA

91 :4382-

81. Di Stefano, L., M. R. Jensen, and K. Helin. 2003. E2F7 , a novel E2F

featuring DP-independent repression of a subset of E2F-regulated genes.

Embo J 22:6289-98.

82. Dicker, A. J., C. Papa, A. L. Dahler, M. M. Serewko, P. A. Hilditch- Maguire, I. H. Frazer, and N. A. Saunders. 2000. E2F- 1 induces

proliferation-specific genes and suppresses squamous differentiation-

specific genes in human epidermal keratinocytes. Oncogene 19:2887-94. 174

2000. Regulation of 83. Dimri, G. P., K. Itahana, M. Acosta, and J. Campisi.

a senescence checkpoint response by the E2F1 transcription factor and

p14(ARF) tumor suppressor. Mol Cell Bioi 20:273-85.

84. DiTullo, R. A., Jr., T. A. Mochan, M. Venere, J. Bartkova, M. Sehested,

J. Bartek, and T. D. Halazonetis. 2002. 53BP1 functions in an ATM- human dependent checkpoint pathway that is constitutively activated in

cancer. Nat Cell Bioi 4:998- 1002. , Y. Kawakubo, and T. 85. Dobashi, Y. , M. Shoji, S. X. Jiang, M. Kobayashi Kameya. 1998. Active cyclin A-CDK2 complex, a possible critical factor for cell proliferation in human primary lung carcinomas. Am J Pathol

153:963-72.

86. Duensing, S., and K. Munger. 2002. The human papillomavirus type 16

E6 and E7 oncoproteins independently induce numerical and structural

chromosome instability. Cancer Res 62:7075-82.

87. Dunaief, J. L., B. E. Strober, S. Guha, P. A. Khavari , K. Alin, J. Luban,

M. Begemann , G. R. Crabtree, and S. P. Goff. 1994. The retinoblastoma

protein and BRG1 form a complex and cooperate to induce cell cycle

arrest. Cell 79: 119-30. 1983. A 88. Durst, M., L. Gissmann, H. Ikenberg, and H. zur Hausen.

papillomavirus DNA from a cervical carcinoma and its prevalence in

cancer biopsy samples from different geographic regions. Proc Natl Acad

Sci USA 80:3812- 175

89. Dyson, N. 1998. The regulation of E2F by pRB-family proteins. Genes

Dev 12:2245-62.

90. Dyson, N., K. Buchkovich, P. Whyte, and E. Harlow. 1989. The cellular

107K protein that binds to adenovirus E1A also associates with the large T

antigens of SV40 and JC virus. Cell 58:249-55.

91. Easton, J., T. Wei, J. M. Lahti, and V. J. Kidd. 1998. Disruption of the

cyclin D/cyclin-dependent kinasellNK4/retinoblastoma protein regulatory

pathway in human neuroblastoma. Cancer Res 58:2624-32.

92. Eddy, B. E. , G. S. Borman , G. E. Grubbs, and R. D. Young. 1962.

Identification of the oncogenic substance in rhesus monkey kidney cell

culture as simian virus 40. Virology 17:65-75.

93. Edmonds, C. , and K. H. Vousden. 1989. A point mutational analysis of

human papillomavirus type 16 E7 protein. J Virol 63:2650-

94. Egan, C. , S. T. Bayley, and P. E. Branton. 1989. Binding of the Rb1

protein to E1A products is required for adenovirus transformation.

Oncogene 4:383-

95. Egan, C., T. N. Jelsma, J. A. Howe, S. T. Bayley, B. Ferguson, and

E. Branton. 1988. Mapping of cellular protein- binding sites on the

products of early-region 1A of human adenovirus type 5. Mol Cell Bioi

8:3955-

96. Eilers, M., S. Schirm, and J. M. Bishop. 1991. The MYC protein

activates transcription of the alpha-prothymosin gene. Embo J 10: 133-41. 176

97. el-Deiry, W. 5., J. W. Harper, P. M. O'Connor, V. E. Velculescu, C. E.

Canman, J. Jackman, J. A. Pietenpol, M. Burrell, D. E. Hil, Y. Wang, and et al. 1994. WAF1/CIP1 is induced in p53-mediated G1 arrest and

apoptosis. Cancer Res 54:1169-74.

98. el-Deiry, W. 5., T. Tokino, V. E. Velculescu, D. B. Levy, R. Parsons, J.

M. Trent, D. Lin, W. E. Mercer, K. W. Kinzler, and B. Vogelstein. 1993.

WAF1 , a potential mediator of p53 tumor suppression. Cell 75:817-25.

99. Enders, G. H. 2004. Colon Cancer Metastasis: Is E2F-1 a Driving Force?

Cancer Bioi Ther 3.

100. Esposito, V. , A. Baldi, A. De Luca, A. M. Groger, M. Loda, G. G.

Giordano, M. Caputi , F. Baldi, M. Pagano, and A. Giordano. 1997.

Prognostic role of the cyclin-dependent kinase inhibitor p27 in non-small

cell lung cancer. Cancer Res 57:3381-

101. Evan, G. I., A. H. Wylle, C. S. Gilbert, T. D. Littlewood, H. Land, M.

Brooks, C. M. Waters, L. Z. Penn, and D. C. Hancock. 1992. Induction

of apoptosis in fibroblasts by c-myc protein. Cell 69: 119-28.

102. Evans, T. , E. T. Rosenthal, J. Youngblom, D. Distel, and T. Hunt.

1983. Cyclin: a protein specified by maternal mRNA in sea urchin eggs

that is destroyed at each cleavage division. Cell 33:389-96.

103. Ewen , M. E., J. W. Ludlow, E. Marsilio, J. A. DeCaprio, R. C. Milikan,

S. H. Cheng, E. Paucha, and D. M. Livingston. 1989. An N-terminal

transformation-governing sequence of SV40 large T antigen contributes to 177

, p120. Cell the binding of both p110Rb and a second cellular protein

58:257-67.

104. Ewen, M. E., H. K. Sluss, C. J. Sherr, H. Matsushime, J. Kato, and

M. Livingston. 1993. Functional interactions of the retinoblastoma protein

with mammalian D-type cyclins. Cell 73:487-97.

M. Livingston. 1991. 105. Ewen, M. E. , Y. G. Xing, J. B. Lawrence, and D.

Molecular cloning, chromosomal mapping, and expression of the cDNA for

p107, a retinoblastoma gene product-related protein. Cell 66:1155-64. Distinct 106. Eymin, B. , S. Gazzeri, C. Brambila, and E. Brambila. 2001.

pattern of E2F1 expression in human lung tumours: E2F1 is upregulated

in small cell lung carcinoma. Oncogene 20:1678-87.

107. Eymin, B., L. Karayan, P. Seite, C. Brambila, E. Brambila, C. J.

Larsen, and S. Gazzeri. 2001. Human ARF binds E2F1 and inhibits its

transcriptional activity. Oncogene 20:1033-41.

108. Ezhevsky, S. A., A. Ho, M. Becker-Hapak, P. K. Davis, and S.

Dowdy. 2001. Differential regulation of retinoblastoma tumor suppressor

protein by G(1) cyclin-dependent kinase complexes in vivo. Mol Cell Bioi

21 :4773-84.

109. Fattaey, A., K. Helin, and E. Harlow. 1993. Transcriptional inhibition by

the retinoblastoma protein. Philos Trans R Soc Lond B Bioi Sci 340:333-

H. , A. 110. Feber, A., J. Clark, G. Goodwin, A. R. Dodson, P. Fletcher, S. Edwards, P. Flohr, A. Falconer, T. Roe, G. Kovacs, 178

C. S. Dennis, C. Fisher, R. Wooster, R. Huddart, C. S. Foster, and Cooper. 2004. Amplification and overexpression of E2F3 in human

bladder cancer. Oncogene 23:1627-30. Ward, P. J. 111. Fernandez-Capetilo, 0., H. T. Chen, A. Celeste, I. Otero, N. Romanienko, J. C. Morales, K. Naka, Z. Xia, R. D. Camerini-

Motoyama, P. B. Carpenter, W. M. Bonner, J. Chen, and A. Nussenzweig. 2002. DNA damage-induced G2-M checkpoint activation

by histone H2AX and 53BP1. Nat Cell Bioi 4:993- Bellan, and D. 112. , R. , L. Magnaghi-Jaulin, P. Robin, A. Harel- Trouche. 1998. The three members of the pocket proteins family share histone the ability to repress E2F activity through recruitment of a

deacetylase. Proc Natl Acad Sci USA 95: 1 0493- 113. Field, S. J., F. Y. Tsai, F. Kuo, A. M. Zubiaga, W. G. Kaelin, Jr., D. Livingston, S. H. Orkin, and M. E. Greenberg. 1996. E2F-1 functions in 61. mice to promote apoptosis and suppress proliferation. Cell 85:549- 1993. E2F- 114. Flemington, E. K., S. H. Speck, and W. G. Kaelin, Jr. mediated transactivation is inhibited by complex formation with the

retinoblastoma susceptibility gene product. Proc Natl Acad Sci U

90:6914-

115. Freedman, D. A., and A. J. Levine. 1998. Nuclear export is required for

degradation of endogenous p53 by MDM2 and human papillomavirus E6.

Mol Cell Bioi 18:7288-93. 179

116. Friend, S. H. , R. Bernards, S. Rogelj, R. A. Weinberg, J. M. Rapaport D. M. Albert, and T. P. Dryja. 1986. A human DNA segment with

properties of the gene that predisposes to retinoblastoma and

osteosarcoma. Nature 323:643-

117. Fuchs, S. Y. , V. Adler, T. Buschmann, X. Wu, and Z. RonaL 1998.

Mdm2 association with p53 targets its ubiquitination. Oncogene 17:2543-

118. Fung, Y. K., A. L. Murphree, A. T'Ang, J. Qian , S. H. Hinrichs, and W.

F. Benedict. 1987. Structural evidence for the authenticity of the human

retinoblastoma gene. Science 236:1657-61.

119. Galaktionov , K., X. Chen, and D. Beach. 1996. Cdc25 cell-cycle

phosphatase as a target of c-myc. Nature 382:511-

120. Ginsberg, D. , G. Vairo, T. Chittenden, Z. X. Xiao, G. Xu, K. L. Wydner

J. A. DeCaprio, J. B. Lawrence, and D. M. Livingston. 1994. E2F- , a

new member of the E2F transcription factor family, interacts with p107.

Genes Dev 8:2665-79.

121. Girard, F., U. Strausfeld, A. Fernandez, and N. J. Lamb. 1991. Cyclin A

is required for the onset of DNA replication in mammalian fibroblasts. Cell

67:1169-79.

122. Girardi, A. J., M. R. Hileman, and R. E. Zwickey. 1962. Search for virus

in human malignancies. 2. In vivo studies. Proc Soc Exp Bioi Med 111 :84-

93. .-,

180

123. Girardi, A. J., V. B. Slotnick, and M. R. Hileman. 1962. Search for virus in human malignancies. I. In vitro studies. Proc Soc Exp Bioi Med

110:776-85. 124. Girardi, A. J., B. H. Sweet, V. B. Slotnick, and M. R. Hileman. 1962.

Development of tumors in hamsters inoculated in the neonatal period with

vacuolating virus, SV-40. Proc Soc Exp Bioi Med 109:649-60.

125. Girling, R., J. F. Partridge, L. R. Bandara, N. Burden, N. F. Totty, J. J. Hsuan, and N. B. La Thangue. 1993. A new component of the

transcription factor DRTF1/E2F. Nature 365:468.

126. Glotzer, M. , A. W. Murray, and M. W. Kirschner. 1991. Cyclin is

degraded by the ubiquitin pathway. Nature 349: 132-

127. Goldberg, M., M. Stucki, J. Falck , D. D'Amours, D. Rahman,

Pappin, J. Bartek, and S. P. Jackson. 2003. MDC1 is required for the

intra- phase DNA damage checkpoint. Nature 421 :952-

128. Gonzalez, S. L., M. Stremlau, X. He, J. R. Basile, and K. Munger. 2001.

Degradation of the retinoblastoma tumor suppressor by the human

papillomavirus type 16 E7 oncoprotein is important for functional

inactivation and is separable from proteasomal degradation of E7. J Virol

75:7583-91.

129. Green , D. R., and T. A. Ferguson. 2001. The role of Fas ligand

immune privilege. Nat Rev Mol Cell Bioi 2:917-24. 181

130. Grunstein, M. 1997. Histone acetylation in chromatin structure and transcription. Nature 389:349-52.

131. Guida, P., and L. Zhu. 1999. DP1 phosphorylation in multimeric

complexes: weaker interaction with cyclin A through the E2F1 cyclin A

binding domain leads to more efficient phosphorylation than stronger

interaction through the p107 cyclin A binding domain. Biochem Biophys

Res Commun 258:596-604.

132. Guo, Z., A. Kumagai , S. X. Wang, and W. G. Dunphy. 2000.

Requirement for Atr in phosphorylation of Chk1 and cell cycle regulation in

response to DNA replication blocks and UV-damaged DNA in Xenopus

egg extracts. Genes Dev 14:2745-56.

133. Hall, M. , and G. Peters. 1996. Genetic alterations ofcyclins , cyclin- dependent kinases, and Cdk inhibitors in human cancer. Adv Cancer Res

68:67 - 108.

134. Hall-Jackson, C. A., D. A. Cross, N. Morrice, and C. Smythe. 1999.

A TR is a caffeine-sensitive , DNA-activated protein kinase with a substrate

specificity distinct from DNA-PK. Oncogene 18:6707-13.

135. Hannon, G. J., D. Demetrick, and D. Beach. 1993. Isolation of the Rb- related p130 through its interaction with CDK2 and cyclins. Genes Dev

7:2378-91.

136. Harbour, J. W., and D. C. Dean. 2000. Chromatin remodeling and Rb

activity. Curr Opin Cell Bioi 12:685- 182

137. Harbour, J. W., and D. C. Dean. 2000. The Rb/E2F pathway: expanding

roles and emerging paradigms. Genes Dev 14:2393-409.

138. Harlow, E., P. Whyte, B. R. Franza, Jr., and C. Schley. 1986.

Association of adenovirus early-region 1A proteins with cellular

polypeptides. Mol Cell Bioi 6:1579-89. 139. Harper, J. W., S. J. Elledge, K. Keyomarsi, B. Dynlacht, L. H. Tsai, P. Zhang, S. Dobrowolski, C. Bai , L. Connell-Crowley, E. Swindell, and

et al. 1995. Inhibition of cyclin-dependent kinases by p21. Mol Bioi Cell

6:387 -400.

140. Hassig, C. A., T. C. Fleischer, A. N. BHlin, S. L. Schreiber, and D. E. Ayer. 1997. Histone deacetylase activity is required for full transcriptional

repression by mSin3A. Cell 89:341-

141. Hassig, C. A., and S. L. Schreiber. 1997. Nuclear histone acetylases and

deacetylases and transcriptional regulation: HATs off to HDACs. Curr

Opin Chem Bioi 1 :300-

142. Haupt, Y., R. Maya, A. Kazaz, and M. Oren. 1997. Mdm2 promotes the

rapid degradation of p53. Nature 387:296-

143. He, T. C., S. Zhou, L. T. da Costa, J. Yu, K. W. Kinzler, and Vogelstein. 1998. A simplified system for generating recombinant

adenoviruses. Proc Natl Acad Sci USA 95:2509- 14. , -

183

144. Heald, R., M. McLoughlin, and F. McKeon. 1993. Human wee1 cytoplasmically maintains mitotic timing by protecting the nucleus from

activated Cdc2 kinase. Cell 74:463-74. C. D. 145. Heinzel, T., R. M. Lavinsky, T. M. Mullen, M. Soderstrom, Davie, E. Laherty, J. Torchia, W. M. Yang, G. Brard, S. D. Ngo, J. R.

Seto, R. N. Eisenman, D. W. Rose, C. K. Glass, and M. G. Rosenfeld. 1997. A complex containing N-CoR , mSin3 and histone deacetylase

mediates transcriptional repression. Nature 387:43- Newport, and K. A. 146. Hekmat-Nejad, M., Z. You, M. C. Yee, J. W.

Cimprich. 2000. Xenopus ATR is a replication-dependent chromatin- Bioi binding protein required for the DNA replication checkpoint. Curr

10: 1565-73. 147. Helin, K., E. Harlow, and A. Fattaey. 1993. Inhibition of E2F- protein. Mol Cell transactivation by direct binding of the retinoblastoma

Bioi 13:6501- Fattaey. 148. Helin, K., J. A. Lees, M. Vidal, N. Dyson, E. Harlow, and A. 1992. A cDNA encoding a pRB-binding protein with properties of the

transcription factor E2F. Cell 70:337-50.

149. Helin, K., C. L. Wu, A. R. Fattaey, J. A. Lees, B. D. Dynlacht, C. Ngwu,

and E. Harlow. 1993. Heterodimerization of the transcription factors E2F- 61. 1 and DP-1 leads to cooperative trans-activation. Genes Dev 7:1850- ,::"

184

family. Curr 150, Hengst, L., and S. I. Reed. 1998. Inhibitors of the Cip/Kip Top Microbiollmmunol 227:25-41. Morgan, and P. J. 151. Herzog, K. H., M. J. Chong, M. Kapsetaki, J. I. induced cell McKinnon. 1998. Requirement for Atm in ionizing radiation- 91. death in the developing central nervous system. Science 280:1089- , M. Oren, G. 152. Hiebert, S. W., G. Packham, D. K. Strom, R. Haffner Zambetti, and J. L. Cleveland. 1995. E2F- 1 :DP- 1 induces p53 and 74. overrides survival factors to trigger apoptosis. Mol Cell Bioi 15:6864- t Veer, 153. Hijmans, E. M., P. M. Voorhoeve, R. L. Beijersbergen, L. J. van '

and R. Bernards. 1995. E2F- , a new E2F family member that interacts

with p130 in vivo. Mol Cell Bioi 15:3082- R. A. 154. Hinds, P. W., S. Mittnacht, V. Dulic, A. Arnold, S. I. Reed, and Weinberg. 1992. Regulation of retinoblastoma protein functions by 1006. ectopic expression of human cyclins. Cell 70:993- Ruland, H. 155. Hirao, A., Y. Y. Kong, S. Matsuoka, A. Wakeham, J. damage- Yoshida, D. Liu, S. J. Elledge, and T. W. Mak. 2000. DNA induced activation of p53 by the checkpoint kinase Chk2. Science 287:1824-7. of G(1) cell- 156. Ho, A. , and S. F. DO,\Y, 2002. Regulation cycle progression

by oncogenes and tumor suppressor genes. Curr Opin Genet Dev 12:47-

52. 185

157. Ho, G. H. , J. E. Calvano, M. Bisogna, and K. J. Van Zee. 2001.

Expression of E2F- 1 and E2F-4 is reduced in primary and metastatic

breast carcinomas. Breast Cancer Res Treat 69: 115-22.

158. Hofferer, M., C. Wirbelauer, B. Humar, and W. Krek. 1999. Increased levels of E2F- dependent DNA binding activity after UV- or gamma-

irradiation. Nucleic Acids Res 27:491-

159. Hollstein, M. , K. Rice, M. S. Greenblatt, T. Soussi, R. Fuchs, T. Sorlie,

E. Hovig, B. Smith-Sorensen, R. Montesano, and C. C. Harris. 1994.

Database of p53 gene somatic mutations in human tumors and cell lines.

Nucleic Acids Res 22:3551-

160. Honda, R. , H. Tanaka, and H. Yasuda. 1997. Oncoprotein MDM2 is a

ubiquitin ligase E3 for tumor suppressor p53. FEBS Lett 420:25- 161. Hopfner, K. P., L. Craig, G. Moncalian, R. A. Zinkel, T. Usui, B. A.

Owen, A. Karcher, B. Henderson , J. L. Bodmer, C. T. McMurray, J. P.

Carney, J. H. Petrini, and J. A. Tainer. 2002. The Rad50 zinc-hook is a

structure joining Mre11 complexes in DNA recombination and repair.

Nature 418:562-

162. Horsthemke, B. 1992. Genetics and cytogenetics of retinoblastoma.

Cancer Genet Cytogenet 63:1-

163. Howe, J. A. , and S. T. Bayley. 1992. Effects of Ad5 E1A mutant viruses

on the cell cycle in relation to the binding of cellular proteins including the

retinoblastoma protein and cyclin A. Virology 186: 15-24. 186

164. Howes, K. A., N. Ransom, D. S. Papermaster, J. G. Lasudry, D. M.

Albert, and J. J. Windle. 1994. Apoptosis or retinoblastoma: alternative

fates of photoreceptors expressing the HPV- 16 E7 gene in the presence

or absence of p53. Genes Dev 8:1300-10.

165. Huang, Y. , T. Ishiko, S. Nakada, T. Utsugisawa, T. Kato, and Z.

Yuan. 1997. Role for E2F in DNA damage-induced entry of cells into S

phase. Cancer Res 57:3640-

166. Humbert, P. , C. Rogers, S. Ganiatsas, R. L. Landsberg, J.

Trimarchi, S. Dandapani, C. Brugnara, S. Erdman , M. Schrenzel, R. T.

Bronson, and J. A. Lees. 2000. E2F4 is essential for normal erythrocyte

maturation and neonatal viability. Mol Cell 6:281-91.

167. Humbert, P. , R. Verona, J. M. Trimarchi, C. Rogers, S. Dandapani,

and J. A. Lees. 2000. E2f3 is critical for normal cellular proliferation.

Genes Dev 14:690-703.

168. Hupp, T. R., D. W. Meek, C. A. Midgley, and D. P. Lane. 1992.

Regulation of the specific DNA binding function of p53. Cell 71 :875-86.

169. Iavarone, A., and J. Massague. 1999. E2F and histone deacetylase

mediate transforming growth factor beta repression of cdc25A during

keratinocyte cell cycle arrest. Mol Cell Bioi 19:916-22.

170. lida, H. , M. Towatari, M. Tanimoto, Y. Morishita, Y. Kodera, and

Saito. 1997. Overexpression of cyclin E in acute myelogenous leukemia.

Blood 90:3707-13. 187

171. Ikeda, M. A., L. Jakoi, and J. R. Nevins. 1996. A unique role for the Rb growth and protein in controlling E2F accumulation during cell

differentiation. Proc Natl Acad Sci USA 93:3215-20.

172. Inoue, K., M. F. Roussel , and C. J. Sherr. 1999. Induction of ARF tumor

suppressor gene expression and cell cycle arrest by transcription factor

DMP1. Proc Natl Acad Sci USA 96:3993-

173. Ishida, S. , E. Huang, H. Zuzan, R. Spang, G. Leone, M. West, and J. R.

Nevins. 2001. Role for E2F in control of both DNA replication and mitotic

functions as revealed from DNA microarray analysis. Mol Cell Bioi

21 :4684-99.

174. Ivey-Hoyle, M., R. Conroy, H. E. Huber, P. J. Goodhart, A. Oliff, and D.

C. Heimbrook. 1993. Cloning and characterization of E2F- , a novel

protein with the biochemical properties of transcription factor E2F. Mol Cell

Bioi 13:7802- 12. 1994. 175. Iwabuchi , K., P. L. Bartel, B. Li, R. Marraccino, and S. Fields.

Two cellular proteins that bind to wild-type but not mutant p53. Proc Natl

Acad Sci USA 91 :6098- 1 02. 176. Iwabuchi, K., B. Li, H. F. Massa, B. J. Trask, T. Date, and S. Fields. 1998. Stimulation of p53-mediated transcriptional activation by the p53-

. JJ , 53BP1 and 53BP2. J Bioi Chem 273:26061- t ' binding proteins 177. Iwamoto, M., D. Banerjee, L. G. Menon, A. Jurkiewicz, P. H. Rao, N. E.

Kemeny, Y. Fong, S. C. Jhanwar, R. Gorlick, and J. R. Bertino. 2004. 188

Overexpression of E2F- 1 in Lung and Liver Metastases of Human Colon

Cancer is Associated With Gene Amplification. Cancer Bioi Ther 3.

178. Jack, M. T., R. A. Woo, A. Hirao, A. Cheung, T. W. Mak, and P. W. Lee.

2002. Chk2 is dispensable for p53-mediated G1 arrest but is required for a

latent p53-mediated apoptotic response. Proc Natl Acad Sci U

99:9825-

179. Jacks, T., A. Fazeli, E. M. , R. T. Bronson, M. A. Goodell, and R. A. Weinberg. 1992. Effects of an Rb mutation in the mouse. Nature

359:295-300.

180. Jelsma, T. N. , J. A. Howe, J. S. Mymryk, C. M. Evelegh, N. F. Cunniff,

and S. T. Bayley. 1989. Sequences in E1A proteins of human adenovirus

5 required for cell transformation , repression of a transcriptional enhancer

and induction of proliferating cell nuclear antigen. Virology 171 :120-30.

181. Jimenez, G. S., M. Nister, J. M. Stommel, M. Beeche, E. A. Barcarse,

x. Q. Zhang, S. O'Gorman, and G. M. Wahl. 2000. A transactivation-

deficient mouse model provides insights into Trp53 regulation and

function. Nat Genet 26:37-43.

182. Johnson, D. G. , W. D. Cress, L. Jakoi, and J. R. Nevins. 1994. Oncogenic capacity of the E2F1 gene. Proc Natl Acad Sci U

91:12823- 189

183. Johnson, D. G., J. K. Schwarz, W. D. Cress, and J. R. Nevins. 1993.

Expression of transcription factor E2F1 induces quiescent cells to enter S

phase. Nature 365:349-52.

184. Jones, S. N., A. T. Sands, A. R. Hancock, H. Vogel, L. A. Donehower,

S. P. Linke, G. M. Wahl, and A. Bradley. 1996. The tumorigenic potential

and cell growth characteristics of p53- deficient cells are equivalent in the

presence or absence of Mdm2. Proc Natl Acad Sci USA 93:14106- 11.

185. Jongmans, W. , M. Vuillaume , K. Chrzanowska, D. ,

Sperling, and J. Hall. 1997. Nijmegen breakage syndrome cells fail to

induce the p53-mediated DNA damage response following exposure to

ionizing radiation. Mol Cell Bioi 17:5016-22.

186. Kaelin, W. G., Jr. , W. Krek, W. R. Sellers, J. A. DeCaprio, F. Ajchenbaum, C. S. Fuchs, T. Chittenden, Y. Li, P. J. Farnham , M. A.

Blanar, and et al. 1992. Expression cloning of a cDNA encoding a

retinoblastoma-binding protein with E2F-like properties. Cell 70:351-64.

187. Kalma, Y., L. Marash, Y. Lamed, and D. Ginsberg. 2001. Expression

analysis using DNA microarrays demonstrates that E2F- 1 up- regulates

expression of DNA replication genes including replication protein A2.

Oncogene 20: 1379-87.

188. Kamb, A. 1998. Cyclin-dependent kinase inhibitors and human cancer.

Curr Top Microbiollmmunol 227:139-48. 190

189. Kamijo, T., F. Zindy, M. F. Roussel, D. E. Quelle, J. R. Downing, R. A.

Ashmun, G. Grosveld, and C. J. Sherr. 1997. Tumor suppression at the

mouse INK4a locus mediated by the alternative reading frame product

p19ARF. Cell 91 :649-59.

190. Kannan, K., N. Kaminski, G. Rechavi, J. Jakob-Hirsch, N. Amariglio,

and D. Givo!. 2001. DNA microarray analysis of genes involved in p53

mediated apoptosis: activation of Apaf-1. Oncogene 20:3449-55.

191. Kastan, M. B., and D. S. Lim. 2000. The many substrates and functions

of ATM. Nat Rev Mol Cell Bioi 1:179-86.

192. Kastan, M. B. , O. Onyekwere, D. Sidransky, B. Vogelstein, and R.

Craig. 1991. Participation of p53 protein in the cellular response to DNA

damage. Cancer Res 51 :6304-11.

193. Kato, J., H. Matsushime, S. W. Hiebert, M. E. Ewen, and C. J. Sherr.

1993. Direct binding of cyclin D to the retinoblastoma gene product (pRb)

and pRb phosphorylation by the cyclin D-dependent kinase CDK4. Genes

Dev 7:331-42.

194. Kennedy, B. K., D. A. Barbie, M. Classon, N. Dyson, and E. Harlow.

2000. Nuclear organization of DNA replication in primary mammalian cells.

Genes Dev 14:2855-68.

195. Kerr, J. F. 2002. History of the events leading to the formulation of the

apoptosis concept. Toxicology 181-182:471- 191

196. Kerr, J. F., A. H. Wylle, and A. R. Currie. 1972. Apoptosis: a basic

biological phenomenon with wide-ranging implications in tissue kinetics. Br

J Cancer 26:239-57.

197. Keyomarsi , K., D. Conte, Jr., W. Toyofuku, and M. P. Fox. 1995.

Deregulation of cyclin E in breast cancer. Oncogene 11 :941-50.

198. Kim, J. H., M. J. Kang, C. U. Park, H. J. Kwak, Y. Hwang, and G. Y.

Koh. 1999. Amplified CDK2 and cdc2 activities in primary colorectal

carcinoma. Cancer 85:546-53.

199. Kim, s. T., D. s. Urn, C. E. Canman, and M. B. Kastan. 1999. Substrate

specificities and identification of putative substrates of A TM kinase family

members. J Bioi Chern 274:37538-43. 200. King, R. W., P. K. Jackson, and M. W. Kirschner. 1994. Mitosis in

transition. Cell 79:563-71. 201. Kingston, R. E., and G. J. Narlikar. 1999. A TP-dependent remodeling

and acetylation as regulators of chromatin fluidity. Genes Dev 13:2339-52.

202. Klein, G., A. Powers, and C. Croce. 2002. Association of SV40 with

human tumors. Oncogene 21:1141-

203. Knudson, A. G., Jr. 1971. Mutation and cancer: statistical study of

retinoblastoma. Proc Natl Acad Sci USA 68:820-

204. Ko, L. J., and C. Prives. 1996. p53: puzzle and paradigm. Genes Dev

10:1054-72. 192

205. Kobayashi, J. , H. Tauchi , S. Sakamoto, A. Nakamura, K. Morishima,

S. Matsuura, T. Kobayashi, K. Tarnai, K. Tanimoto, and K. Komatsu.

2002. NBS1 localizes to gamma-H2AX foci through interaction with the

FHA/BRCT domain. Curr Bioi 12:1846-51.

206. Koff, A., F. Cross, A. Fisher, J. Schumacher, K. Leguellec, M.

Philippe, and J. M. Roberts. 1991. Human cyclin E, a new cyclin that

interacts with two members of the CDC2 gene family. Cell 66:1217-28.

207. Kornberg, R. D., and Y. Lorch. 1999. Chromatin-modifying and

remodeling complexes. Curr Opin Genet Dev 9:148-51.

208. Kovesdi , I., R. Reichel, and J. R. Nevins. 1986. Identification of a

cellular transcription factor involved in E1A trans-activation. Cell 45:219-

28.

209. Kovesdi , I., R. Reichel, and J. R. Nevins. 1987. Role of an adenovirus

E2 promoter binding factor in E1A-mediated coordinate gene control. Proc

Natl Acad Sci USA 84:2180-

210. Kowalik, T. F., J. DeGregori, G. Leone, L. Jakoi, and J. R. Nevins.

1998. E2F1-specific induction of apoptosis and p53 accumulation , which is

blocked by Mdm2. Cell Growth Differ 9:113-

211. Kowalik, T. F. , J. DeGregori, J. K. Schwarz, and J. R. Nevins. 1995.

E2F1 overexpression in quiescent fibroblasts leads to induction of cellular

DNA synthesis and apoptosis. J Virol 69:2491-500. 193

212. Krek, W. , M. E. Ewen, S. Shirodkar, Z. Arany, W. G. Kaelin, Jr. , and D.

M. Livingston. 1994. Negative regulation of the growth- promoting

transcription factor E2F- 1 by a stably bound cyclin A-dependent protein

kinase. Cell 78:161-72.

213. Krek, W., D. M. Livingston, and S. Shirodkar. 1993. Binding to DNA and the retinoblastoma gene product promoted by complex formation of

different E2F family members. Science 262: 1557 -60.

214. Kroemer, G., and J. C. Reed. 2000. Mitochondrial control of cell death.

Nat Med 6:513-

215. LaBaer, J., M. D. Garrett, L. F. Stevenson, J. M. Slingerland, C.

Sandhu, H. S. Chou, A. Fattaey, and E. Harlow. 1997. New functional

activities for the p21 family of CDK inhibitors. Genes Dev 11:847-62. 216. Lai, A., J. M. Lee, W. M. Yang, J. A. DeCaprio, W. G. Kaelin, Jr.,

Seto, and P. E. Branton. 1999. RBP1 recruits both histone deacetylase-

dependent and -independent repression activities to retinoblastoma family

proteins. Mol Cell Bioi 19:6632-41. 217. Lakin, N. D., B. C. Hann, and S. P. Jackson. 1999. The ataxia-

telangiectasia related protein A TR mediates DNA-dependent

phosphorylation of p53. Oncogene 18:3989-95.

218. Leach , F. S., S. J. Elledge, C. J. Sherr, J. K. Wilson, S. Markowitz, K.

W. Kinzler, and B. Vogelstein. 1993. Amplification of cyclin genes in

colorectal carcinomas. Cancer Res 53: 1986- 194

219. Lee, E. Y., C. Y. Chang, N. Hu, Y. C. Wang, C. C. Lai, K. Herrup, W. H.

Lee, and A. Bradley. 1992. Mice deficient for Rb are nonviable and show

defects in neurogenesis and haematopoiesis. Nature 359:288-94.

220. Lee, J. 0., A. A. Russo, and N. P. Pavletich. 1998. Structure of the

retinoblastoma tumour-suppressor pocket domain bound to a peptide from

HPV E7. Nature 391 :859-65.

221. Lee, M. H., I. Reynisdottir, and J. Massague. 1995. Cloning of p57KIP2

a cyclin-dependent kinase inhibitor with unique domain structure and

tissue distribution. Genes Dev 9:639-49. 222. Lee, M. H., B. O. Wiliams, G. Mullgan, S. Mukai, R. T. Bronson, Dyson, E. Harlow, and T. Jacks. 1996. Targeted disruption of p107:

functional overlap between p107 and Rb. Genes Dev 10:1621-32.

223. Lee, M. H. , and H. Y. Yang. 2001. Negative regulators of cyclin-

dependent kinases and their roles in cancers. Cell Mol Life Sci 58:1907-

22.

224. Lee, W. H., R. Bookstein, F. Hong, L. J. Young, J. Y. Shew, and E. Y. Lee. 1987. Human retinoblastoma susceptibility gene: cloning,

identification , and sequence. Science 235:1394-

225. Lees, J. A., K. J. Buchkovich, D. R. Marshak, C. W. Anderson, and E. Harlow. 1991. The retinoblastoma protein is phosphorylated on multiple

sites by human cdc2. Embo J 10:4279-90. 195

226. Lees, J. A., M. Saito, M. Vidal, M. Valentine, T. Look, E. Harlow, N. Dyson, and K. Helin. 1993. The retinoblastoma protein binds to a family

of E2F transcription factors. Mol Cell Bioi 13:7813-25. , A. 227. Leone, G., F. Nuckolls, S. Ishida, M. Adams, R. Sears, L. Jakoi Miron, and J. R. Nevins. 2000. Identification of a novel E2F3 product

suggests a mechanism for determining specificity of repression by Rb

proteins. Mol Cell Bioi 20:3626-32.

228. Leone, G., R. Sears, E. Huang, R. Rempel, F. Nuckolls, C. H. Park, P. Giangrande, L. Wu, H. I. Saavedra, S. J. Field, M. A. Thompson, Yang, Y. Fujiwara, M. E. Greenberg, S. Orkin, C. Smith, and J.

Nevins. 2001. Myc requires distinct E2F activities to induce S phase and

apoptosis. Mol Cell 8:105-13.

229. Levine, A. J. 1997. p53 , the cellular gatekeeper for growth and division.

Cell 88:323-31.

230. Lew, D. J. , V. Dulic, and S. I. Reed. 1991. Isolation of three novel human 206. cyclins by rescue of G1 cyclin (Cln) function in yeast. Cell 66:1197- 231. Li, Y., C. Graham, S. Lacy, A. M. Duncan, and P. Whyte. 1993. The

adenovirus E1A-associated 130-kD protein is encoded by a member of

the retinoblastoma gene family and physically interacts with cyclins A and

E. Genes Dev 7:2366-77. 196

232. Li, Y., J. E. Siansky, D. J. Myers, N. R. Drinkwater, W. G. Kaelin, and P. J. Farnham. 1994. Cloning, chromosomal location , and

characterization of mouse E2F1. Mol Cell Bioi 14:1861- 233. Liao, M. J., C. Yin, C. Barlow, A. Wynshaw-Boris, and T. van Dyke. 1999. Atm is dispensable for p53 apoptosis and tumor suppression

triggered by cell cycle dysfunction. Mol Cell Bioi 19:3095- 1 02.

234. Lim, D. 5., S. T. Kim, B. Xu, R. S. Maser, J. Lin, J. H. Petrini, and M. B.

Kastan. 2000. ATM phosphorylates p95/nbs1 in an S-phase checkpoint

pathway. Nature 404:613-

235. Lin, W. C., F. T. Lin, and J. R. Nevins. 2001. Selective induction of E2F1

in response to DNA damage , mediated by A TM- dependent

phosphorylation. Genes Dev 15:1833-44.

236. Lindstrom, M. , and K. G. Wiman. 2003. Myc and E2F1 induce p53

through p14ARF- independent mechanisms in human fibroblasts.

Oncogene 22:4993-5005.

237. Liu, F., J. J. Stanton, Z. Wu, and H. Piwnica-Worms. 1997. The human Myt1 kinase preferentially phosphorylates Cdc2 on threonine 14 and

localizes to the endoplasmic reticulum and Golgi complex. Mol Cell Bioi

17:571-83. , K. Tamai, G. 238. Liu, Q., S. Guntuku, X. S. Cui, S. Matsuoka, D. Cortez Luo, S. Carattini-Rivera, F. DeMayo, A. Bradley, L. A. Donehower, and

S. J. Elledge. 2000. Chk1 is an essential kinase that is regulated by Atr 197

Dev and required for the G(2)/M DNA damage checkpoint. Genes

14:1448-59.

239. lockshin, R. A. , and C. M. Wiliams. 1965. Programmed Cell Death-- Pernyi Cytology of Degeneration in the Intersegmental Muscles of the

Silkmoth. J Insect Physiol11: 123-33. Helin. 240. lomazzi , M., M. C. Moroni, M. R. Jensen, E. Frittoli, and K.

2002. Suppression of the p53- or pRB-mediated G1 checkpoint is required

for E2F-induced S-phase entry. Nat Genet 31:190-

241. lombard, D. B., and L. Guarente. 2000. Nijmegen breakage syndrome

disease protein and MRE11 at PML nuclear bodies and meiotic telomeres.

Cancer Res 60:2331- Chen. 2003. MDC 1 is 242. lou, Z., K. Minter-Dykhouse, X. Wu, and J. response coupled to activated CHK2 in mammalian DNA damage

pathways. Nature 421 :957 -61. 243. luo, R. X., A. A. Postigo, and D. C. Dean. 1998. Rb interacts with

histone deacetylase to repress transcription. Cell 92:463-73.

244. Ma, Y., R. Croxton, R. L. Moorer, Jr., and W. D. Cress. 2002.

Identification of novel E2F1-regulated genes by microarray. Arch Biochem

Biophys 399:212-24.

245. Maclachlan, T. K., and W. 5. EI-Deiry. 2002. Apoptotic threshold is

lowered by p53 transactivation of caspase-6. Proc Natl Acad Sci USA

99:9492- 198

246. Macleod, K. F., Y. Hu, and T. Jacks. 1996. Loss of Rb activates both

p53-dependent and independent cell death pathways in the developing

mouse nervous system. Embo J 15:6178-88.

247. Magnaghi-Jaulin, L, R. Groisman, I. Naguibneva, P. Robin , S. Lorain,

J. P. Le Vilain, F. Troalen , D. Trouche, and A. Harel-Bellan. 1998.

Retinoblastoma protein represses transcription by recruiting a histone

deacetylase. Nature 391 :601-

248. Majno, G. , and I. Joris. 1995. Apoptosis , oncosis , and necrosis. An

overview of cell death. Am J PathoI146:3-15. 249. Malhotra, P., C. F. Manohar, S. Swaminathan, R. Toyama, R. Dhar, R. Reichel, and B. Thimmapaya. 1993. E2F site activates transcription in

fission yeast Schizosaccharomyces pombe and binds to a 30- kDa

transcription factor. J Bioi Chern 268:20392-401.

250. Maltzman , W., and L Czyzyk. 1984. UV irradiation stimulates levels of

p53 cellular tumor antigen in nontransformed mouse cells. Mol Cell Bioi

4: 1689-94.

251. Martell, F. , T. Hamilton, D. P. Silver, N. E. Sharpless, N. Bardeesy, M.

Rokas, R. A. DePinho, D. M. Livingston, and S. R. Grossman. 2001.

p19ARF targets certain E2F species for degradation. Proc Natl Acad Sci U

SA 98:4455-60. 199

252. Marti, A. , C. Wirbelauer, M. Scheffner, and W. Krek. 1999. Interaction between ubiquitin-protein ligase SCFSKP2 and E2F- 1 underlies the

regulation of E2F- 1 degradation. Nat Cell Bioi 1 :14-

253. Martinez-Balbas, M. A. , U. M. Bauer, S. J. Nielsen, A. Brehm, and T. Kouzarides. 2000. Regulation of E2F1 activity by acetylation. Embo J

19:662-71.

254. Maser, R. 5., O. K. Mirzoeva, J. Wells, H. Olivares, B. R. Wiliams, A. Zinkel, P. J. Farnham, and J. H. Petrini. 2001. Mre11 complex and

DNA replication: linkage to E2F and sites of DNA synthesis. Mol Cell Bioi

21 :6006-16.

255. Maser, R. 5., K. J. Monsen, B. E. Nelms, and J. H. Petrini. 1997.

hMre11 and hRad50 nuclear foci are induced during the normal cellular

response to DNA double-strand breaks. Mol Cell Bioi 17:6087-96.

256. Mason, S. L., O. Loughran, and N. B. La Thangue. 2002. p14(ARF)

regulates E2F activity. Oncogene 21 :4220-30.

257. Matsuda, K., K. Yoshida, Y. Taya, K. Nakamura, Y. Nakamura, and H.

Arakawa. 2002. p53AIP1 regulates the mitochondrial apoptotic pathway.

Cancer Res 62:2883-

258. Matsuoka, S., M. Huang, and S. J. Elledge. 1998. Linkage of A TM to cell

cycle regulation by the Chk2 protein kinase. Science 282:1893- 200

259. Matsuoka, S., G. Rotman, A. Ogawa, Y. Shiloh, K. Tarnai, and S. J.

Elledge. 2000. Ataxia telangiectasia-mutated phosphorylates Chk2 in vivo

and in vitro. Proc Natl Acad Sci USA 97:10389-94. 260. Mayol, X., X. Grana, A. Baldi, N. Sang, Q. Hu, and A. Giordano. 1993.

Cloning of a new member of the retinoblastoma gene family (pRb2) which

binds to the E1A transforming domain. Oncogene 8:2561- 261. McCaffrey, J., L. Yamasaki, N. J. Dyson, E. Harlow, and A. E. Griep. 1999. Disruption of retinoblastoma protein family function by human

papillomavirus type 16 E7 oncoprotein inhibits lens development in part

through E2F- 1. Mol Cell Bioi 19:6458-68.

262. McDonald, E. R., 3rd, and W. S. EI-Deiry. 2000. Cell cycle control as a

basis for cancer drug development (Review). Int J OncoI16:871-86.

263. McMasters, K. M., R. Montes de Oca Luna, J. R. Pena, and G. Lozano. 1996. mdm2 deletion does not alter growth characteristics of p53-deficient

embryo fibroblasts. Oncogene 13:1731-

264. Melchionna, R. , X. B. Chen, A. Blasina, and C. H. McGowan. 2000.

Threonine 68 is required for radiation- induced phosphorylation and

activation of Cds1. Nat Cell Bioi 2:762-

265. Melilo, R. M., K. Helin, D. R. Lowy, and J. T. Schiler. 1994. Positive

and negative regulation of cell proliferation by E2F- 1: influence of protein

level and human papillomavirus oncoproteins. Mol Cell Bioi 14:8241- 201

266. Meng, R. D., P. Phillps, and W. S. EI-Deiry. 1999. p53-independent

increase in E2F- 1 expression enhances the cytotoxic effects of etoposide

and of adriamycin. Int J OncoI14:5-14.

267. Mirzoeva, O. K. , and J. H. Petrini. 2001. DNA damage-dependent

nuclear dynamics of the Mre11 complex. Mol Cell Bioi 21 :281- 268. Mirzoeva, O. K., and J. H. Petrini. 2003. DNA replication-dependent

nuclear dynamics of the Mre11 complex. Mol Cancer Res 1 :207- 18.

269. Miyashita, T., S. Krajewski, M. Krajewska, H. G. Wang, H. K. Lin, D. A. Liebermann, B. Hoffman, and J. C. Reed. 1994. Tumor suppressor p53

is a regulator of bcl-2 and bax gene expression in vitro and in vivo.

Oncogene 9: 1799-805.

270. Moberg, K., M. A. Starz, and J. A. Lees. 1996. E2F-4 switches from

p130 to p107 and pRB in response to cell cycle reentry. Mol Cell Bioi

16:1436-49.

271. Mochan , T. A. , M. Venere, R. A. DiTullo, Jr., and T. D. Halazonetis.

2003. 53BP1 and NFBD1/MDC1-Nbs1 function in parallel interacting

pathways activating ataxia-telangiectasia mutated (A TM) in response to

DNA damage. Cancer Res 63:8586-91.

272. Morgan, D. 0. 1995. Principles of CDK regulation. Nature 374:131-

273. Morgenbesser, S. D., B. O. Wiliams, T. Jacks, and R. A. DePinho.

1994. p53-dependent apoptosis produced by Rb-deficiency in the

developing mouse lens. Nature 371 :72- , !'

202

274. Mori, A., H. Higashi, Y. Hoshikawa, M. Imamura, M. Asaka, and M. Hatakeyama. 1999. Granulocytic differentiation of myeloid progenitor cells

by p130 , the retinoblastoma tumor suppressor homologue. Oncogene

18:6209-21.

275. Morkel, M., J. Wenkel , A. J. Bannister, T. Kouzarides, and C. Hagemeier. 1997. An E2F-like repressor of transcription. Nature 390:567-

276. Moroni , M. C., E. S. Hickman, E. L. Denchi, G. Caprara, E. Coll,

Cecconi, H. Muller, and K. Helin. 2001. Apaf- 1 is a transcriptional target

for E2F and p53. Nat Cell Bioi 3:552-

277. Morris, J. D. , T. Crook, L. R. Bandara, R. Davies, N. B. LaThangue, and K. H. Vousden. 1993. Human papillomavirus type 16 E7 regulates

E2F and contributes to mitogenic signalling. Oncogene 8:893-

278. Motokura, T. , T. Bloom , H. G. Kim, H. Juppner, J. V. Ruderman, H. M. Kronenberg, and A. Arnold. 1991. A novel cyclin encoded by a bcl1-

linked candidate oncogene. Nature 350:512-

279. Muller, H., A. P. Bracken, R. Vernell, M. C. Moroni, F. Christians, E. Grassili, E. Prosperini, E. Vigo, J. D. Oliner, and K. Helin. 2001. E2Fs

regulate the expression of genes involved in differentiation , development

proliferation , and apoptosis. Genes Dev 15:267-85.

280. Muller, M. , S. Wilder, D. Bannasch , D. Israeli , K. Lehlbach, M. Li-

Weber, S. L. Friedman, P. R. Galle, W. Stremmel, M. Oren, and P. 203

Krammer. 1998. p53 activates the CD95 (APO- 1/Fas) gene in response

to DNA damage by anticancer drugs. J Exp Med 188:2033-45.

281. Murray, A. W. 2004. Recycling the cell cycle: cyclins revisited. Cell

116:221-34.

282. Murray, A. W. , M. J. Solomon, and M. W. Kirschner. 1989. The role of

cyclin synthesis and degradation in the control of maturation promoting

factor activity. Nature 339:280-

283. Nahle, Z., J. Polakoff, R. V. Davuluri , M. E. McCurrach , M. D.

Jacobson, M. Narita, M. Q. Zhang, Y. Lazebnik, D. Bar-Sagi, and S. W.

Lowe. 2002. Direct coupling of the cell cycle and cell death machinery by

E2F. Nat Cell Bioi 4:859-64.

284. Nakano, K. , and K. H. Vousden. 2001. PUMA, a novel proapoptotic

gene, is induced by p53. Mol Cell 7:683-94.

285. Nelms, B. E. , R. S. Maser, J. F. MacKay, M. G. Lagally, and J.

Petrini. 1998. In situ visualization of DNA double-strand break repair in

human fibroblasts. Science 280:590-

286. Nevins, J. R. 1992. E2F: a link between the Rb tumor suppressor protein

and viral oncoproteins. Science 258:424-

287. Nevins, J. R. 1998. Toward an understanding of the functional complexity

of the E2F and retinoblastoma families. Cell Growth Differ 9:585-93.

288. Nevins, J. R., J. DeGregori, L. Jakoi, and G. Leone. 1997. Functional

analysis of E2F transcription factor. Methods EnzymoI283:205-19. 204

289. Nghiem, P. , P. K. Park, Y. Kim, C. Vaziri, and S. L. Schreiber. 2001.

ATR inhibition selectively sensitizes G1 checkpoint-deficient cells to lethal

premature chromatin condensation. Proc Natl Acad Sci USA 98:9092- 290. Nielsen, S. J., R. Schneider, U. M. Bauer, A. J. Bannister, A. Morrison,

D. O' Carroll, R. Firestein, M. Cleary, T. Jenuwein, R. E. , and T. Kouzarides. 2001. Rb targets histone H3 methylation and HP1 to

promoters. Nature 412:561- 291. Nilsson, I., and I. Hoffmann. 2000. Cell cycle regulation by the Cdc25

phosphatase family. Prog Cell Cycle Res 4:107- 14. 292. Nip, J., D. K. Strom, C. M. Eischen, J. L. Cleveland, G. P. Zambetti,

and S. W. Hiebert. 2001. E2F- 1 induces the stabilization of p53 but

blocks p53-mediated transactivation. Oncogene 20:910-20. 293. Novitch, B. G., D. B. Spicer, P. S. Kim, W. L. Cheung, and A. B.

Lassar. 1999. pRb is required for MEF2-dependent gene expression as

well as cell-cycle arrest during skeletal muscle differentiation. Curr Bioi

9:449-59.

294. O'Connor, D. J., and X. Lu. 2000. Stress signals induce transcriptionally

inactive E2F- 1 independently of p53 and Rb. Oncogene 19:2369-76.

295. Oda, E. , R. Ohki, H. Murasawa, J. Nemoto, T. Shibue, T. Yamashita, T.

Tokino, T. Taniguchi , and N. Tanaka. 2000. Noxa, a BH3-only member

of the BcI-2 family and candidate mediator of p53-induced apoptosis.

Science 288: 1 053- 205

296, Oda, K. , H. Arakawa, T. Tanaka, K. Matsuda, C. Tanikawa, T. Mori, H.

Nishimori, K. Tamai, T. Tokino, Y. Nakamura, and Y. Taya. 2000.

p53AIP1 , a potential mediator of p53-dependent apoptosis , and its

regulation by Ser-46-phosphorylated p53. Cell 102:849-62.

297. Ohtani, K., and J. R. Nevins. 1994. Functional properties of a Drosophila

homolog of the E2F1 gene. Mol Cell Bioi 14:1603-12.

298. Ohtsubo, M., A. M. Theodoras, J. Schumacher, J. M. Roberts, and

Pagano. 1995. Human cyclin E, a nuclear protein essential for the G1-to-

S phase transition. Mol Cell Bioi 15:2612-24.

299. Ormondroyd, E., 5. de la Luna, and N. B. La Thangue. 1995. A new

member of the DP family, DP- , with distinct protein products suggests a

regulatory role for alternative splicing in the cell cycle transcription factor

DRTF1/E2F. Oncogene 11:1437-46.

300. Osborn, A. J., 5. J. Elledge, and L. Zou. 2002. Checking on the fork: the

DNA-replication stress-response pathway. Trends Cell Bioi 12:509- 16.

301. Owen-Schaub, L. B., L. 5. Angelo, R. Radinsky, C. F. Ware, T. G. Gesner, and D. P. Bartos. 1995. Soluble Fas/APO- 1 in tumor cells: a

potential regulator of apoptosis? Cancer Lett 94: 1-

302. Pagano, M., M. Durst, S. Joswig, G. Draetta, and P. Jansen-Durr.

1992. Binding of the human E2F transcription factor to the retinoblastoma

protein but not to cyclin A is abolished in HPV- 16-immortalized cells.

Oncogene 7:1681- 206

303. Palmero, I., C. Pantoja, and M. Serrano. 1998. p19ARF links the tumour

suppressor p53 to Ras. Nature 395: 125-

304. Pan, H., and A. E. Griep. 1994. Altered cell cycle regulation in the lens of

HPV-16 E6 or E7 transgenic mice: implications for tumor suppressor gene

function in development. Genes Dev 8: 1285-99.

305. Pan, H. , C. Yin, N. J. Dyson, E. Harlow, L. Yamasaki, and T. Van Dyke.

1998. Key roles for E2F1 in signaling p53-dependent apoptosis and in cell

division within developing tumors. Mol Cell 2:283-92.

306. Pan , W., A. Datta, G. R. Adami, P. Raychaudhuri, and S. Bagchi. 2003.

P19ARF inhibits the functions of the HPV16 E7 oncoprotein. Oncogene

22:5496-503.

307. Pardee, A. B. 1989. G1 events and regulation of cell proliferation. Science 246:603- 308. Paull, T. T., and M. Gellert. 1998. The 3' to 5' exonuclease activity of Mre

11 facilitates repair of DNA double-strand breaks. Mol Cell 1 :969-79. 309. Paull, T. T., E. P. Rogakou, V. Yamazaki, C. U. Kirchgessner, M. Gellert, and W. M. Bonner. 2000. A critical role for histone H2AX in

recruitment of repair factors to nuclear foci after DNA damage. Curr Bioi

10:886-95.

310. Peeper, D. S. , P. Keblusek, K. Helin, M. Toebes, A. J. van der Eb, and

A. Zantema. 1995. Phosphorylation of a specific cdk site in E2F- 1 affects

-: I 207

its electrophoretic mobility and promotes pRB-binding in vitro. Oncogene

10:39-48.

311. Peng, A., and P. L. Chen. 2003. NFBD1 , like 53BP1 , is an early and

redundant transducer mediating Chk2 phosphorylation in response to

DNA damage. J Bioi Chem 278:8873-

312. Peng, C. Y. , P. R. Graves, R. S. Thoma, Z. Wu, A. S. Shaw, and H. Piwnica-Worms. 1997. Mitotic and G2 checkpoint control: regulation of

14- 3 protein binding by phosphorylation of Cdc25C on serine-216.

Science 277:1501-

313. Phelps, W. C. , K. Munger, C. L. Yee, J. A. Barnes, and P. M. Howley.

1992. Structure-function analysis of the human papillomavirus type 16 E7

oncoprotein. J Virol 66:2418-27.

314. Phillps, A. C. , M. K. Ernst, S. Bates, N. R. Rice, and K. H. Vousden.

1999. E2F- 1 potentiates cell death by blocking antiapoptotic signaling

pathways. Mol Cell 4:771-81. 315. Philpott, A., and S. H. Friend. 1994. E2F and its developmental

regulation in Xenopus laevis. Mol Cell Bioi 14:5000-

316. Pierce, A. M. , I. B. Gimenez-Conti, R. Schneider-Broussard, L. A.

Martinez, C. J. Conti, and D. G. Johnson. 1998. Increased E2F1 activity

induces skin tumors in mice heterozygous and nullizygous for p53. Proc

Natl Acad Sci USA 95:8858-63. 208

317. Pierce, A. M. , R. Schneider-Broussard, I. B. Gimenez-Conti, J. L. Russell, C. J. Conti, and D. G. Johnson. 1999. E2F1 has both

oncogenic and tumor-suppressive properties in a transgenic model. Mol

Cell Bioi 19:6408- 14.

318. Pines, J. 1995. Cyclins and cyclin-dependent kinases: theme and

variations. Adv Cancer Res 66:181-212.

319. Pines, J. 1991. Cyclins: wheels within wheels. Cell Growth Differ 2:305-

10.

320. Polyak, K., M. H. Lee, H. Erdjument-Bromage, A. Koff, J. M. Roberts,

P. Tempst, and J. Massague. 1994. Cloning of p27Kip1 , a cyclin-

dependent kinase inhibitor and a potential mediator of extracellular

antimitogenic signals. Cell 78:59-66.

321. Pomerantz, J. , N. Schreiber-Agus, N. J. Liegeois, A. Silverman , L. Alland, L. Chin, J. Potes, K. Chen, I. Orlow, H. W. Lee, C. Cordon- Cardo, and R. A. DePinho. 1998. The Ink4a tumor suppressor gene

product, p19Arf, interacts with MDM2 and neutralizes MDM2' s inhibition of

p53. Cell 92:713-23.

322. Porse, B. T. , T. A. Pedersen , X. Xu, B. Lindberg, U. M. Wewer, L. Friis-

Hansen, and C. Nerlov. 2001. E2F repression by C/EBPalpha is required

for adipogenesis and granulopoiesis in vivo. Cell 107:247-58.

323. Prives, C. , and P. A. Hall. 1999. The p53 pathway. J Pathol187: 112-26. 209

D. Adams. 1994. 324. Qin, X. D. M. Livingston, W. G. Kaelin, Jr., and P.

Deregulated transcription factor E2F- 1 expression leads to S-phase entry

and p53-mediated apoptosis. Proc Natl Acad Sci USA 91: 1 0918-22.

325. QueUe, D. E., R. A. Ashmun, G. J. Hannon, P. A. Rehberger, D. Trono,

K. H. Richter, C. Walker, D. Beach, C. J. Sherr, and M. Serrano. 1995.

Cloning and characterization of murine p161NK4a and p151NK4b genes.

Oncogene 11 :635-45.

326. Ramirez-Parra, E., Q. Xie, M. B. Boniotti, and C. Gutierrez. 1999. The

cloning of plant E2F , a retinoblastoma-binding protein , reveals unique and conserved features with animal G(1)/S regulators. Nucleic Acids Res

27:3527-33.

327. Rappold, I., K. Iwabuchi , T. Date, and J. Chen. 2001. Tumor suppressor

p53 binding protein 1 (53BP1) is involved in DNA damage-signaling

pathways. J Cell Bioi 153:613-20. 328. Raychaudhuri, P., S. Bagchi, S. H. Devoto, V. B. Kraus, E. Moran, and J. R. Nevins. 1991. Domains of the adenovirus E1A protein required for

oncogenic activity are also required for dissociation of E2F transcription

factor complexes. Genes Dev 5: 1200-11.

329. Rayman, J. B., Y. Takahashi, V. B. Indjeian, J. H. Dannenberg, S.

Catchpole, R. J. Watson, H. te Riele, and B. D. Dynlacht. 2002. E2F

mediates cell cycle-dependent transcriptional repression in vivo by 210

recruitment of an HDAC1/mSin3B corepressor complex. Genes Dev

16:933-47.

330. Reed, S. I. 2003. Ratchets and clocks: the cell cycle , ubiquitylation and

protein turnover. Nat Rev Mol Cell Bioi 4:855-64.

331. Rempel, R. E., M. T. Saenz-Robles, R. Storms, S. Morham , S. Ishida,

A. Engel , L. Jakoi, M. F. Melhem, J. M. Pipas, C. Smith, and J.

Nevins. 2000. Loss of E2F4 activity leads to abnormal development of

multiple cellular lineages. Mol Cell 6:293-306.

332. Ren, B. , H. Cam, Y. Takahashi, T. Volkert, J. Terragni, R. A. Young,

and B. D. Dynlacht. 2002. E2F integrates cell cycle progression with DNA

repair, replication , and G(2)/M checkpoints. Genes Dev 16:245-56.

333. Rich , T., R. L. Allen, and A. H. Wylle. 2000. Defying death after DNA

damage. Nature 407:777-83.

334. Richon , V. M., R. E. Lyle, and R. E. McGehee, Jr. 1997. Regulation and

expression of retinoblastoma proteins p107 and p130 during 3T3- L 1

adipocyte differentiation. J Bioi Chem 272:10117-24.

335. Robertson , K. D., and P. A. Jones. 1998. The human ARF cell cycle

regulatory gene promoter is a CpG island which can be silenced by DNA

methylation and down-regulated by wild-type p53. Mol Cell Bioi 18:6457-

73. 211

336. Robles, A. I., N. A. Bemmels, A. B. Foraker, and C. C. Harris. 2001.

APAF- 1 is a transcriptional target of p53 in DNA damage-induced

apoptosis. Cancer Res 61 :6660-

337. Rogakou, E. P. , C. Boon , C. Redon, and W. M. Bonner. 1999.

Megabase chromatin domains involved in DNA double-strand breaks in

vivo. J Cell Bioi 146:905-16.

338. Rogakou, E. P. , D. R. Pilch, A. H. Orr, V. S. Ivanova, and W.

Bonner. 1998. DNA double-stranded breaks induce histone H2AX

phosphorylation on serine 139. J Bioi Chem 273:5858-68. 339. Rogers, K. T., P. D. Higgins, M. M. Mila, R. S. Philips, and J.

Horowitz. 1996. DP- , a heterodimeric partner of E2F: identification and

characterization of DP-2 proteins expressed in vivo. Proc Natl Acad Sci U

SA 93:7594-

340. Rogoff, H. A. , M. T. Pickering, M. E. Debatis, S. Jones, and T. F.

Kowalik. 2002. E2F1 induces phosphorylation of p53 that is coincident

with p53 accumulation and apoptosis. Mol Cell Bioi 22:5308-18.

341. Ross, J. F., X. Liu, and B. D. Dynlacht. 1999. Mechanism of

transcriptional repression of E2F by the retinoblastoma tumor suppressor

protein. Mol Cell 3:195-205.

342. Russell , J. L., J. T. Powers, R. J. Rounbehler, P. M. Rogers, C. J.

Conti, and D. G. Johnson. 2002. ARF Differentially Modulates Apoptosis

Induced by E2F1 and Myc. Mol Cell Bioi 22:1360- 212

343. Saavedra, H. I., B. Maiti, C. Timmers, R. Altura, Y. Tokuyama, K.

Fukasawa, and G. Leone. 2003. Inactivation of E2F3 results in

centrosome amplification. Cancer Cell 3:333-46.

344. Saenz-Robles, M. T. , C. S. Sullvan, and J. M. Pipas. 2001.

Transforming functions of Simian Virus 40. Oncogene 20:7899-907. 345. Sage, J., G. J. Mullgan, L. D. Attardi, A. Miler, S. Chen, B. Willams,

E. Theodorou, and T. Jacks. 2000. Targeted disruption of the three Rb- related genes leads to loss of G(1) control and immortalization. Genes

Dev 14:3037-50.

346. Sager, R. 1991. Senescence as a mode of tumor suppression. Environ

Health Perspect 93:59-62.

347. Sardet, C. , M. Vidal , D. Cobrinik, Y. Geng, C. Onufryk, A. Chen , and R.

A. Weinberg. 1995. E2F-4 and E2F- , two members of the E2F family,

are expressed in the early phases of the cell cycle. Proc Natl Acad Sci U S

A 92:2403-

348. Sarkaria, J. N., E. C. Busby, R. S. Tibbetts, P. Roos, Y. Taya, L. M. Karnitz, and R. T. Abraham. 1999. Inhibition of ATM and ATR kinase

activities by the radiosensitizing agent, caffeine. Cancer Res 59:4375-82.

349. Sax, J. K. , and W. S. EI-Deiry. 2003. p53 downstream targets and

chemosensitivity. Cell Death Differ 10:413- 213

350. Sax, J. K. , P. Fei, M. E. Murphy, E. Bernhard, S. J. Korsmeyer, and W.

S. EI-Deiry. 2002. BID regulation by p53 contributes to chemosensitivity.

Nat Cell Bioi 4:842-

351. Scheffner, M. , B. A. Werness, J. M. Huibregtse, A. J. Levine, and P.

Howley. 1990. The E6 oncoprotein encoded by human papillomavirus

types 16 and 18 promotes the degradation of p53. Cell 63:1129-36.

352. Schneider, J. W., W. Gu, L. Zhu, V. Mahdavi, and B. Nadal-Ginard.

1994. Reversal of terminal differentiation mediated by p107 in Rb-

muscle cells. Science 264:1467-71.

353. Schultz, L. B., N. H. Chehab, A. Malikzay, and T. D. Halazonetis. 2000.

p53 binding protein 1 (53BP1) is an early participant in the cellular

response to DNA double-strand breaks. J Cell Bioi 151:1381-90.

354. Schwarz, J. K., C. H. Bassing, I. Kovesdi, M. B. Datto, M. Blazing, S.

George, X. F. Wang, and J. R. Nevins. 1995. Expression of the E2F1

transcription factor overcomes type beta transforming growth factor-

mediated growth suppression. Proc Natl Acad Sci USA 92:483-

355. Schwarz, J. K., S. H. Devoto, E. J. Smith , S. P. Chellappan, L. Jakoi

and J. R. Nevins. 1993. Interactions of the p107 and Rb proteins with

E2F during the cell proliferation response. Embo J 12:1013-20.

356. Schwarz, J. K., C. M. Lovly, and H. Piwnica-Worms. 2003. Regulation

of the Chk2 protein kinase by oligomerization-mediated cis- and trans-

phosphorylation. Mol Cancer Res 1 :598-609. 214

357. Scuderi , R., K. A. Palucka, K. Pokrovskaja, M. Bjorkholm , K. G.

Wiman , and P. Pisa. 1996. Cyclin E overexpression in relapsed adult

acute lymphoblastic leukemias ofB-celilineage. Blood 87:3360-

358. Sears, R. , K. Ohtani, and J. R. Nevins. 1997. Identification of positively

and negatively acting elements regulating expression of the E2F2 gene in

response to cell growth signals. Mol Cell Bioi 17:5227-35. 359. Sekine, M., M. Ito, K. Uemukai , Y. Maeda, H. Nakagami, and A. Shinmyo. 1999. Isolation and characterization of the E2F-like gene in

plants. FEBS Lett 460:117-22.

360. Serrano, M. , G. J. Hannon, and D. Beach. 1993. A new regulatory motif

in cell-cycle control causing specific inhibition of cyclin D/CDK4. Nature

366:704-

361. Serrano, M., H. Lee, L. Chin, C. Cordon-Cardo, D. Beach, and R. A. DePinho. 1996. Role of the INK4a locus in tumor suppression and cell

mortality. Cell 85:27-37.

362. Shan, B., X. Zhu, P. L. Chen, T. Durfee, Y. Yang, D. Sharp, and W. H.

Lee. 1992. Molecular cloning of cellular genes encoding retinoblastoma-

associated proteins: identification of a gene with properties of the

transcription factor E2F. Mol Cell Bioi 12:5620-31.

363. Sherr, C. J. 1996. Cancer cell cycles. Science 274:1672-

364. Sherr, C. J. 1994. G1 phase progression: cycling on cue. Cell 79:551-

365. Sherr, C. J. 2004. Principles of tumor suppression. Cell 116:235-46. 215

366. Sherr, C. J., and F. McCormick. 2002. The RB and p53 pathways in

cancer. Cancer Cell 2: 1 03-12.

367. Sherr, C. J., and J. M. Robert. 1995. Inhibitors of mammalian G1 cyclin-

dependent kinases. Genes Dev 9:1149-63. 368. Shi, Y. 2002. Mechanisms of caspase activation and inhibition during

apoptosis. Mol Cell 9:459-70. 369. Shi, Y., M. Zou, N. R. Farid, and S. T. al-Sedairy. 1996. Evidence of gene deletion of p21 (WAF1/CIP1), a cyclin-dependent protein kinase

inhibitor, in thyroid carcinomas. Br J Cancer 74: 1336-41. 370. Shieh, S. Y., J. Ahn, K. Tarnai, Y. Taya, and C. Prives. 2000. The human homologs of checkpoint kinases Chk1 and Cds1 (Chk2)

phosphorylate p53 at multiple DNA damage-inducible sites. Genes Dev

14:289-300.

371. Shiloh, Y. 1997. Ataxia-telangiectasia and the Nijmegen breakage

syndrome: related disorders but genes apart. Annu Rev Genet 31 :635-62.

372. Shiloh, Y. 2001. ATM and ATR: networking cellular responses to DNA

damage. Curr Opin Genet Dev 11:71-

373. Shiloh, Y. 2003. ATM and related protein kinases: safeguarding genome

integrity. Nat Rev Cancer 3:155-68. 374. Silciano, J. D., C. E. Can man, Y. Taya, K. Sakaguchi, E. Appella, and

M. B. Kastan. 1997. DNA damage induces phosphorylation of the amino

terminus of p53. Genes Dev 11 :3471-81. 216

375. Singh, P., J. Coe, and W. Hong. 1995. A role for retinoblastoma protein

in potentiating transcriptional activation by the glucocorticoid receptor.

Nature 374:562-

376. Smith, J. R., and O. M. Pereira-Smith. 1996. Replicative senescence:

implications for in vivo aging and tumor suppression. Science 273:63-

377. Stamato, T. , S. Guerriero, and N. Denko. 1993. Two methods for

assaying DNA double-strand break repair in mammalian cells by

asymmetric field inversion gel electrophoresis. Radiat Res 133:60- 378. Stambolic, V., D. MacPherson, D. Sas, Y. Un , B. Snow, Y. Jang, S.

Benchimol, and T. W. Mak. 2001. Regulation of PTEN transcription by

p53. Mol Cell 8:317-25.

379. Stanelle, J., T. Stiewe, F. Rodicker, K. Kohler, C. Theseling, and B. M.

Putzer. 2003. Mechanism of E2F1-induced apoptosis in primary vascular

smooth muscle cells. Cardiovasc Res 59:512-

380. Stanelle, J., T. Stiewe, C. C. Theseling, M. Peter, and B. M. Putzer.

2002. Gene expression changes in response to E2F1 activation. Nucleic

Acids Res 30:1859-67.

381. Stein , G. H., L. F. Drullnger, A. Soulard, and V. Dulic. 1999. Differential

roles for cyclin-dependent kinase inhibitors p21 and p16 in the

mechanisms of senescence and differentiation in human fibroblasts. Mol

Cell Bioi 19:2109- 17. . -

217

382. Stevaux, 0., and N. J. Dyson. 2002. A revised picture of the E2F

transcriptional network and RB function. Curr Opin Cell Bioi 14:684-91.

383. Stevens, C. , L. Smith , and N. B. La Thangue. 2003. Chk2 activates

E2F- 1 in response to DNA damage. Nat Cell Bioi 5:401-

384. Stewart, G. S., R. S. Maser, T. Stankovic, D. A. Bressan, M. I. Kaplan

N. G. Jaspers, A. Raams, P. J. Byrd, J. H. Petrini , and A. M. Taylor.

1999. The DNA double-strand break repair gene hMRE11 is mutated in

individuals with an ataxia-telangiectasia-like disorder. Cell 99:577-87.

385. Stewart, G. S., B. Wang, C. R. Bignell, A. M. Taylor, and S. J. Elledge.

2003. MDC1 is a mediator of the mammalian DNA damage checkpoint.

Nature 421 :961-

386. Stiegler, P. , A. De Luca, L. Bagella, and A. Giordano. 1998. The

COOH-terminal region of pRb2/p130 binds to histone deacetylase

(HDAC1), enhancing transcriptional repression of the E2F-dependent

cyclin A promoter. Cancer Res 58:5049-52.

387. Stiegler, P. , and A. Giordano. 2001. The family of retinoblastoma

proteins. Crit Rev Eukaryot Gene Expr 11 :59-76.

388. Strobeck, M. W., K. E. Knudsen, A. F. Fribourg, M. F. DeCristofaro, B.

E. Weissman, A. N. Imbalzano, and E. S. Knudsen. 2000. BRG- 1 is

. required for RB-mediated cell cycle arrest. Proc Natl Acad Sci USA

97:7748-53. 218

389. Strober, B. E., J. L. Dunaief, Guha, and S. P. Goff. 1996. Functional

interactions between the hBRM/hBRG1 transcriptional activators and the

pRB family of proteins. Mol Cell Bioi 16: 1576-83.

390. Sugihara, T., S. C. Kaul, J. Kato, R. R. Reddel, H. Nomura, and

Wadhwa. 2001. Pex19p dampens the p19ARF-p53-p21WAF1 tumor

suppressor pathway. J Bioi Chem 276:18649-52.

391. Suzuki , A., and A. Hemmati-Brivanlou. 2000. Xenopus embryonic E2F

is required for the formation of ventral and posterior cell fates during early

embryogenesis. Mol Cell 5:217-29.

392. Sweet, B. H., and M. R. Hileman. 1960. The vacuolating virus, S.V. 40.

Proc Soc Exp Bioi Med 105:420-

393. Symonds, H. , L. Krall, L. Remington, M. Saenz-Robles, S. Lowe, T.

Jacks, and T. Van Dyke. 1994. p53-dependent apoptosis suppresses

tumor growth and progression in vivo. Cell 78:703- 11.

394. Taalman, R. D., N. G. Jaspers, J. M. Scheres, J. de Wit, and T.

Hustinx. 1983. Hypersensitivity to ionizing radiation , in vitro , in a new

chromosomal breakage disorder, the Nijmegen Breakage Syndrome.

Mutat Res 112:23-32.

395. Takahashi , Y. , J. B. Rayman, and B. D. Dynlacht. 2000. Analysis

promoter binding by the E2F and pRB families in vivo: distinct E2F

proteins mediate activation and repression. Genes Dev 14:804- 16. 219

396. Takai, H., K. Naka, Y. Okada, M. Watanabe, N. Harada, S. Saito, C. W. Anderson , E. Appella, M. Nakanishi, H. Suzuki, K. Nagashima,

Sawa, K. Ikeda, and N. Motoyama. 2002. Chk2-deficient mice exhibit radioresistance and defective p53-mediated transcription. Embo J

21 :5195-205.

397. Tanaka, H., I. Matsumura, S. Ezoe, Y. Satoh , T. Sakamaki,

Albanese, T. Machii, R. G. Pestell, and Y. Kanakura. 2002. E2F1 and c-

Myc potentiate apoptosis through inhibition of NF-kappaB activity that

facilitates MnSOD-mediated ROS elimination. Mol Cell 9:1017-29.

398. Tao, Y., R. F. Kassatly, W. D. Cress, and J. M. Horowitz. 1997. Subunit

composition determines E2F DNA-binding site specificity. Mol Cell Bioi

17:6994-7007.

399. Todaro, G. J., and H. Green. 1964. Enhancement by Thymidine Analogs

of Susceptibility of Cells to Transformation by Sv40. Virology 24:393-400.

400. Todaro, G. J., and H. Green. 1966. High frequency of SV40

transformation of mouse cell line 3T3. Virology 28:756-

401. Tolbert, D., X. Lu, C. Yin, M. Tantama, and T. Van Dyke. 2002.

p19(ARF) is dispensable for oncogenic stress- induced p53-mediated

apoptosis and tumor suppression in vivo. Mol Cell Bioi 22:370-

402. Trentin, J. J., Y. Yabe, and G. Taylor. 1962. The quest for human cancer

viruses. Science 137:835-41. 220

403. Trimarchi, J. M. , B. Fairchild, J. Wen, and J. A. Lees. 2001. The E2F6

transcription factor is a component of the mammalian Bmi1-containing

polycomb complex. Proc Natl Acad Sci USA 98:1519-24.

404. Trimarchi , J. M., and J. A. Lees. 2002. Sibling rivalry in the E2F family.

Nat Rev Mol Cell Bioi 3:11-20.

405. Trouche, D. , A. Cook, and T. Kouzarides. 1996. The CBP co-activator

stimulates E2F1/DP1 activity. Nucleic Acids Res 24:4139-45.

406. Trouche, D. , C. Le Chalony, C. Muchardt, M. Yaniv, and Kouzarides. 1997. RB and hbrm cooperate to repress the activation

functions of E2F 1. Proc Natl Acad Sci USA 94: 11268-73.

407. Trujilo, K. M. , S. S. Yuan, E. Y. Lee, and P. Sung. 1998. Nuclease

activities in a complex of human recombination and DNA repair factors

Rad50 , Mre11 , and p95. J Bioi Chem 273:21447-50.

408. Tsai , K. Y. , Y. Hu, K. F. Macleod, D. Crowley, L. Yamasaki , and T.

Jacks. 1998. Mutation of E2f- 1 suppresses apoptosis and inappropriate S

phase entry and extends survival of Rb-deficient mouse embryos. Mol Cell

2:293-304.

409. Tsai , K. Y., D. MacPherson, D. A. Rubinson, D. Crowley, and T. Jacks.

2002. ARF is not required for apoptosis in Rb mutant mouse embryos.

Curr Bioi 12:159-63.

410. Tsai , T. , S. Davalath, C. Rankin , J. P. Radich, D. Head, F. R.

Appelbaum, and D. H. Boldt. 1996. Tumor suppressor gene alteration in 221

adult acute lymphoblastic leukemia (ALL). Analysis of retinoblastoma (Rb)

and p53 gene expression in Iymphoblasts of patients with de novo

relapsed , or refractory ALL treated in Southwest Oncology Group studies.

Leukemia 10:1901- 10.

411. Unger, T., R. V. Sionov, E. Moallem, C. L. Yee, P. M. Howley, M. Oren,

and Y. Haupt. 1999. Mutations in serines 15 and 20 of human p53 impair

its apoptotic activity. Oncogene 18:3205-12.

412. Uziel , T. , Y. Lerenthal, L. Moyal, Y. Andegeko, L. Mittelman, and Y.

Shiloh. 2003. Requirement of the MRN complex for ATM activation by

DNA damage. Embo J 22:5612-21.

413. Vafa, 0. , M. Wade, S. Kern, M. Beeche, T. K. Pandita, G. M. Hampton,

and G. M. Wahl. 2002. c-Myc can induce DNA damage, increase reactive

oxygen species , and mitigate p53 function: a mechanism for oncogene-

induced genetic instability. Mol Cell 9:1031-44.

414. Vairo, G., D. M. Livingston, and D. Ginsberg. 1995. Functional

interaction between E2F-4 and p130: evidence for distinct mechanisms

underlying growth suppression by different retinoblastoma protein family

members. Genes Dev 9:869-81.

415. Van Cruchten, S., and W. Van Den Broeck. 2002. Morphological and

biochemical aspects of apoptosis, oncosis and necrosis. Anat Histol

Embryol 31 :214-23. 222

416. van Gent, D. C. , J. H. Hoeijmakers, and R. Kanaar. 2001. Chromosomal

stability and the DNA double-stranded break connection. Nat Rev Genet

2:196-206.

417. Vaute, 0., E. Nicolas, L. Vandel, and D. Trouche. 2002. Functional and

physical interaction between the histone methyl transferase Suv39H1 and

histone deacetylases. Nucleic Acids Res 30:475-81.

418. Vemu , S. , and R. R. Reichel. 1995. Cell cycle regulation of a novel DNA

binding complex in Saccharomyces cerevisiae with E2F-like properties. J

Bioi Chem 270:20724-

419. Vigo, E., H. Muller, E. Prosperini, G. Hateboer, P. Cartright, M. C.

Moroni , and K. Helin. 1999. CDC25A phosphatase is a target of E2F and

is required for effcient E2F- induced S phase. Mol Cell Bioi 19:6379-95.

420. Vivanco, I., and C. L. Sawyers. 2002. The phosphatidylinositol 3-Kinase

AKT pathway in human cancer. Nat Rev Cancer 2:489-501.

421. Wadhwa, R., T. Sugihara, M. K. Hasan, K. Taira, R. R. Reddel, and S.

C. Kaul. 2002. A major functional difference between the mouse and

human ARF tumor suppressor proteins. J Bioi Chem 277:36665-70.

422. Walker, D. H., and J. L. Maller. 1991. Role for cyclin A in the dependence

of mitosis on completion of DNA replication. Nature 354:314-

423. Wang, B. , S. Matsuoka, P. B. Carpenter, and S. J. Elledge. 2002.

53BP1 , a mediator of the DNA damage checkpoint. Science 298:1435- 223

424. Wang, J., K. Helin, P. Jin, and B. Nadal-Ginard. 1995. Inhibition of in

vitro myogenic differentiation by cellular transcription factor E2F1. Cell

Growth Differ 6:1299-306.

425. Wang, Z. M., H. Yang, and D. M. Livingston. 1998. Endogenous E2F-

promotes timely GO exit of resting mouse embryo fibroblasts. Proc Natl

Acad Sci USA 95:15583-

426. Ward, I. M. , K. Minn , K. G. Jorda, and J. Chen. 2003. Accumulation of

checkpoint protein 53BP1 at DNA breaks involves its binding to

phosphorylated histone H2AX. J Bioi Chem 278: 19579-82.

427. Ward, I. M., K. Minn, J. van Deursen, and J. Chen. 2003. p53 Binding

protein 53BP1 is required for DNA damage responses and tumor

suppression in mice. Mol Cell Bioi 23:2556-63.

428. Weber, J. D., J. R. Jeffers, J. E. Rehg, D. H. Randle, G. Lozano, M. F.

Roussel , C. J. Sherr, and G. P. Zambetti. 2000. p53-independent

functions of the p19(ARF) tumor suppressor. Genes Dev 14:2358-65.

429. Weinberg, R. A. 1995. The retinoblastoma protein and cell cycle control.

Cell 81 :323-30.

430. Weisenburger, D. D. , W. G. Sanger, J. O. Armitage, and D. T. Purtilo. 1987. Intermediate lymphocytic lymphoma: immunophenotypic and

cytogenetic findings. Blood 69:1617-21. 224

431. Wells, J., K. E. Boyd, C. J. Fry, S. M. Bartley, and P. J. Farnham. 2000.

Target gene specificity of E2F and pocket protein family members in living

cells. Mol Cell Bioi 20:5797-807.

432. Wolf, D. A., H. Hermeking, T. Albert, T. Herzinger, P. Kind, and

Eick. 1995. A complex between E2F and the pRb-related protein p130 is

specifically targeted by the simian virus 40 large T antigen during cell

transformation. Oncogene 10:2067-78.

433. Wolfel , T., M. Hauer, J. Schneider, M. Serrano, C. Wolfel , E.

Klehmann-Hieb, E. De Plaen , T. Hankeln , K. H. Meyer zum

Buschenfelde, and D. Beach. 1995. A p16lNK4a-insensitive CDK4

mutant targeted by cytolytic T lymphocytes in a human melanoma.

Science 269:1281-

434. Wong, A. K., F. Shanahan, Y. Chen, L. Lian, P. Ha, K. Hendricks, S.

Ghaffari, D. llev, B. Penn, A. M. Woodland, R. Smith, G. Salada, A.

Carilo, K. Laity, J. Gupte, B. Swedlund, S. V. Tavtigian, D. H. Teng,

and E. Lees. 2000. BRG1 , a component of the SWI-SNF complex , is

mutated in multiple human tumor cell lines. Cancer Res 60:6171-

435. Wu, C. L. , L. R. Zukerberg, C. Ngwu, E. Harlow, and J. A. Lees. 1995.

In vivo association of E2F and DP family proteins. Mol Cell Bioi 15:2536-

46.

436. Wu, E. W. , K. E. Clemens, D. V. Heck, and K. Munger. 1993. The

human papillomavirus E7 oncoprotein and the cellular transcription factor

!i: f!J '",

225

E2F bind to separate sites on the retinoblastoma tumor suppressor

protein. J Virol 67:2402-

437. Wu, G. , W. H. Lee, and P. L. Chen. 2000. NBS1 and TRF1 colocalize at

promyelocytic leukemia bodies during late S/G2 phases in immortalized

telomerase-negative cells. Implication of NBS1 in alternative lengthening

of telomeres. J Bioi Chem 275:30618-22.

438. Wu, L. , C. Timmers, B. Maiti, H. I. Saavedra, L. Sang, G. T. Chong, F.

Nuckolls, P. Giangrande, F. A. Wright, S. J. Field, M. E. Greenberg, S.

Orkin, J. R. Nevins, M. L. Robinson, and G. Leone. 2001. The E2F1-

transcription factors are essential for cellular proliferation. Nature 414:457-

62.

439. Wu, X. , and J. Chen. 2003. Autophosphorylation of checkpoint kinase 2

at serine 516 is required for radiation-induced apoptosis. J Bioi Chem

278:36163-

440. Wu, X. , and A. J. Levine. 1994. p53 and E2F- 1 cooperate to mediate

apoptosis. Proc Natl Acad Sci USA 91 :3602-

441. Wu, Z., J. Earle, S. Saito, C. W. Anderson , E. Appella, and Y. Xu. 2002.

Mutation of mouse p53 Ser23 and the response to DNA damage. Mol Cell

Bioi 22:2441-

442. Wylle, A. H., J. F. Kerr, and A. R. Currie. 1980. Cell death: the

significance of apoptosis. Int Rev Cytol 68:251-306. 226

443. Xie, S., H. Wu, Q. Wang, J. P. Cogswell, I. Husain, C. Conn, P.

Stambrook, M. Jhanwar-Uniyal, and W. Dai. 2001. Plk3 functionally

links DNA damage to cell cycle arrest and apoptosis at least in part via the

p53 pathway. J Bioi Chem 276:43305- 12.

444. Xu, M. , K. A. Sheppard, C. Y. Peng, A. S. Yee, and H. Piwnica-Worms.

1994. Cyclin A/CDK2 binds directly to E2F- 1 and inhibits the DNA-binding

activity of E2F-1/DP-1 by phosphorylation. Mol Cell Bio/14:8420-31.

445. Xu, X., and D. F. Stern. 2003. NFBD1/MDC1 regulates ionizing radiation-

induced focus formation by DNA checkpoint signaling and repair factors.

Faseb J 17:1842-

446. Yam , C. H., T. K. Fung, and R. Y. Poon. 2002. Cyclin A in cell cycle

control and cancer. Cell Mol Life Sci 59: 1317-26.

447. Yamamoto, H. , T. Monden , H. Miyoshi, H. Izawa, K. Ikeda, M. Tsujie, T.

Ohnishi, M. Sekimoto, N. Tomita, and M. Monden. 1998. Cdk2/cdc2

expression in colon carcinogenesis and effects of cdk2/cdc2 inhibitor in

colon cancer cells. Int J OncoI13:233-

448. Yamasaki, L. , R. Bronson, B. O. Wiliams, N. J. Dyson, E. Harlow, and

T. Jacks. 1998. Loss of E2F- 1 reduces tumorigenesis and extends the

lifespan of Rb1(+/-)mice. Nat Genet 18:360-

449. Yamasaki , L. , T. Jacks, R. Bronson, E. Goilot, E. Harlow, and N. J.

Dyson. 1996. Tumor induction and tissue atrophy in mice lacking E2F-

Cell 85:537-48. 227

450. Yamazaki, V. , R. D. Wegner, and C. U. Kirchgessner. 1998.

Characterization of cell cycle checkpoint responses after ionizing radiation

in Nijmegen breakage syndrome cells. Cancer Res 58:2316-22.

451. Yang, J. , K. Winkler, M. Yoshida, and S. Kornbluth. 1999. Maintenance

of G2 arrest in the Xenopus oocyte: a role for 14- 3-mediated inhibition of

Cdc25 nuclear import. Embo J 18:2174-83.

452. Yonish-Rouach , E., D. Resnitzky, J. Lotem , L. Sachs, A. Kimchi, and

M. Oren. 1991. Wild-type p53 induces apoptosis of myeloid leukaemic

cells that is inhibited by interleukin-6. Nature 352:345-

453. Zacksenhaus, E., Z. Jiang, D. Chung, J. D. Marth, R. A. Philips, and

B. L. Galle. 1996. pRb controls proliferation , differentiation , and death of skeletal muscle cells and other lineages during embryogenesis. Genes

Dev 10:3051-64.

454. Zalvide, J., H. Stubdal, and J. A. DeCaprio. 1998. The J domain of

simian virus 40 large T antigen is required to functionally inactivate RB

family proteins. Mol Cell Bioi 18:1408- 15.

455. Zaman ian, M., and N. B. La Thangue. 1992. Adenovirus E1a prevents

the retinoblastoma gene product from repressing the activity of a cellular

transcription factor. Embo J 11 :2603- 10.

456. Zaman ian, M. , and N. B. La Thangue. 1993. Transcriptional repression

by the Rb-related protein p107. Mol Bioi Cell 4:389-96. 228

457. Zhang, H. 5., M. Gavin, A. Dahiya, A. A. Postigo, D. Ma, R. X. Luo, J.

W. Harbour, and D. C. Dean. 2000. Exit from G1 and S phase of the cell

cycle is regulated by repressor complexes containing HDAC-Rb-

hSWl/SNF and Rb-hSWI/SNF. Cell 101 :79-89.

458. Zhang, Y., and S. P. Chellappan. 1995. Cloning and characterization of

human DP2 , a novel dimerization partner of E2F. Oncogene 10:2085-93.

459. Zhang, Y. , Y. Xiong, and W. G. Yarbrough. 1998. ARF promotes MDM2

degradation and stabilizes p53: ARF-INK4a locus deletion impairs both

the Rb and p53 tumor suppression pathways. Cell 92:725-34.

460. Zhao, 5. , Y. C. Weng, s. S. Yuan, Y. T. Lin, H. C. Hsu, s. C. Lin, E.

Gerbino, M. H. Song, M. Z. Zdzienicka, R. A. Gatti, J. W. Shay, Y. Ziv, Y. Shiloh, and E. Y. Lee. 2000. Functional link between ataxia-

telangiectasia and Nijmegen breakage syndrome gene products. Nature

405:473-

461. Zhu, L., S. van den Heuvel , K. Helin, A. Fattaey, M. Ewen , D.

Livingston, N. Dyson, and E. Harlow. 1993. Inhibition of cell proliferation

by p107 , a relative of the retinoblastoma protein. Genes Dev 7:1111-25. 462. Ziebold, U., T. Reza, A. Caron, and J. A. Lees. 2001. E2F3 contributes

both to the inappropriate proliferation and to the apoptosis arising in Rb

mutant embryos. Genes Dev 15:386-91. 463. Zindy, F., C. M. Eischen, D. H. Randle, T. Kamijo, J. L. Cleveland, C. J. Sherr, and M. F. Roussel. 1998. Myc signaling via the ARF tumor 229

suppressor regulates p53-dependent apoptosis and immortalization.

Genes Dev 12:2424-33.

464. zur Hausen, H., W. Meinhof, W. Scheiber, and G. W. Bornkamm. 1974.

Attempts to detect virus-secific DNA in human tumors. I. Nucleic acid

hybridizations with complementary RNA of human wart virus. Int J Cancer

13:650- 465. zur Hausen, J., H. Schulte-Holthausen, H. Wolf, K. Dorries, and H.

Egger. 1974. Attempts to detect virus-specific DNA in human tumors. II.

Nucleic acid hybridizations with complementary RNA of human herpes

group viruses. Int J Cancer 13:657-64.