<<

CYCLIN E PROVIDES A LINK BETWEEN ,

DNA REPAIR AND

A dissertation submitted

to Kent State University in collaboration with the

Lerner Research Institute, Cleveland Clinic

in partial fulfillment of the requirements for the

degree of Doctor of Philosophy

by

Dragos Costin Plesca

May, 2008

Dissertation written by

Dragos Costin Plesca

Pharm.D., University of Medicine and Pharmacy “Carol Davila”, Romania, 2002

Ph.D., Kent State University, 2008

Approved by

______, Chair, Doctoral Dissertation Committee Alexandru Almasan, Ph.D

______, Member, Doctoral Dissertation Committee James Blank, Ph.D

______, Member, Doctoral Dissertation Committee Gail Fraizer, Ph.D

______, Member, Doctoral Dissertation Committee Olena Piontkivska, Ph.D

______, Graduate Faculty Representative Jennifer Marcinkiewicz, Ph.D

Accepted by

______, Director, School of Biomedical Sciences Robert V. Dorman, Ph.D

______, Dean, College of Arts and Sciences John R. D. Stalvey, Ph.D

ii

TABLE OF CONTENTS

List of Figures...... vi

List of Tables ...... ix

Acknowledgments ...... x

Chapter I. Introduction ...... 1

The Cell Cycle ...... 3

Cyclin E and cell cycle regulation ...... 8

Transcriptional regulation of Cyclin E ...... 14

Post-translational regulation of Cyclin E...... 20

Deregulated expression of Cyclin E in cancer...... 27

Therapeutic approaches involving cell cycle inhibitors...... 32

Apoptosis ...... 38

TRAIL, APO2L ...... 40

Adaptor proteins...... 41

Cytochrome c...... 42

Bcl-2 family members...... 42

Caspases...... 43

Other apoptosis-regulatory proteins...... 45

Apoptotic pathways ...... 46

iii DNA damage and repair mechanisms...... 51

Homologous recombination...... 55

Ku70/Ku80 heterodimer ...... 56

DNA-PKcs ...... 60

Artemis...... 62

XRCC4...... 62

Ligase IV...... 63

XLF...... 64

Non-Homologous End Joining ...... 66

Other DNA double-strand break repair pathways ...... 66

Generation of p18-Cyclin E and its role in modulating cellular

response to genotoxic stress...... 70

Chapter II. p18-Cyclin E regulates Non-Homologous End Joining by preventing

the recruitment of the XLF/XRCC4/Ligase IV heterocomplex to the

DNA repair complex ...... 73

Abstract...... 73

Introduction...... 74

Materials and Methods...... 78

Results...... 82

Discussion...... 95

iv Chapter III. Two new mechanisms for proteasome-mediated Cyclin E degradation

uncovered in hematopoietic tumor cells undergoing apoptosis ...... 102

Abstract...... 102

Introduction...... 103

Materials and Methods...... 106

Results...... 110

Discussion...... 137

Chapter IV. Summary and future directions ...... 145

Summary...... 145

Future directions ...... 148

References...... 157

Appendix. Abbreviations...... 193

v

LIST OF FIGURES

Chapter I

Fig 1.1 Cellular response to DNA damage ...... 2

Fig 1.2 Schematic representation of the cell cycle...... 4

Fig 1.3 Periodic oscillation in the expression levels of ...... 10

Fig 1.4 Schematic representation of the progression through the different cell cycle

phases...... 19

Fig 1.5 Schematic representation of the Cyclin E turnover mechanisms...... 23

Fig 1.6 Schematic representation of the cell cycle and the different therapeutic

approaches undertaken...... 37

Fig 1.7 Schematic representation of the extrinsic and intrinsic apoptotic pathways ...49

Fig 1.8 Three-dimensional structure of the Ku70/Ku80 heterodimer...... 59

Fig 1.9 Schematic representation of the XLF/XRCC4/Ligase IV heterocomplex...... 65

Fig 1.10 Schematic representation of the NHEJ pathway...... 69

Fig 1.11 Schematic representation of Cyclin E domains and cleavage sites ...... 71

vi Chapter II

Fig 2.1 Non-toxic levels of p18-Cyclin E sensitize cells to VP-16 and IR treatment..85

Fig 2.2 p18-Cyclin E inhibits in vitro DNA ligation and plasmid reactivation ...... 88

Fig 2.3 p18-Cyclin E prevents efficient repair of the DNA damage induced by IR and

CPT-11...... 91

Fig 2.4 p18-Cyclin E impairs the recruitment of XLF, XRCC4, and Ligase IV to the

DNA repair complex...... 94

Fig 2.5 Schematic representation of the NHEJ DNA repair pathway and the way in

which p18-Cyclin E affects the assembly of repair factors ...... 101

Chapter III

Fig 3.1 p18-Cyclin E is a short-lived protein that is stabilized by proteasome

inhibition...... 112

Fig 3.2 p18-Cyclin E is effectively ubiquitinated ...... 115

Fig 3.3 Fbw7 interacts with p18-Cyclin E independently of phosphorylation at the C-

terminal phosphodegron ...... 120

Fig 3.4 p18-Cyclin E is regulated by the SCFFbw7 complex and independently of Cdk2

binding ...... 125

Fig 3.5 Ku70 but not Cdk2 binding regulates p18-Cyclin E stability...... 130

Fig 3.6 p18-cyclin E sensitizes cells to Bortezomib treatment through enhanced

association with Ku70 and release of Bax from Ku70 ...... 135

Fig. 3.7 Chemical structure of the proteasome inhibitor, Bortezomib...... 141

vii Chapter IV

Fig 4.1 Dual regulation of apoptosis and DNA repair by p18-Cyclin E...... 147

Fig 4.2 Localization of p18-Cyclin E-GFP, Cyclin E-GFP, and Cyclin ER130A-GFP 151

Fig 4.3 Cyclin E sequence alignment across human, mouse, rat, rabbit, chicken, and

frog...... 153

viii

LIST OF TABLES

Table 1. Critical protein substrates of Caspase-3...... 45

ix

ACKNOWLEDGMENTS

None of the scientific results or achievements that I have had during my last five

years would have been possible without the sustained professional and moral support that

my adviser, Dr. Alex Almasan has given me. I thank him for his guidance, motivating

spirit and for nurturing my scientific creativity and independence. I feel very fortunate for

having studied in his lab as he provided me along the years with a unique Ph.D.

experience and great learning opportunities which will continue to serve me well in the future.

I would also like to thank the colleagues in my lab that throughout the years have supported and encouraged me both professionally and personally and who have shared with me the ups and downs of scientific research: Drs. Marcela Oancea, Meredith

Crosby, Erica DuPree, Damodar Gupta, Subrata Ray and John Hissong. I would like to thank in particular Dr. Suparna Mazumder for her never-ending patience, kindness and advice that she had for me at all times.

I would also like to extend my appreciation to my dissertation committee members Dr. Gail Fraizer, Dr. James Blank, Dr. Olena Piontkivska, and Dr. Bryan

Williams for their constructive comments and help at different stages during my studies.

Thank you also to Dr. Jennifer Marcinkiewicz for joining the committee as graduate

x faculty representative. Furthermore, I thank the School of Biomedical Sciences and the

Department of Cancer Biology for giving me the opportunity to pursue my Ph.D. degree.

Finally, I thank my mother and father, Gabriela and Gheorghe, and my dearest

Carolina whose unending love and emotional support have buffered the frustrations and gave me the strength to take critical decisions in the most difficult of times. They have been a constant source of energy and optimism and they have always had confidence in me whenever I seemed to lose mine. There is nothing that I have accomplished, or will achieve, that would be possible without them.

Time passes and when success or disappointment fades away, what matters the most is what we have influenced and changed with our presence and what we have been able to take with us in our hearts and minds to make us a better person. The pursuit of the

Ph.D. degree was a priceless and unique experience which has profoundly changed my perspectives and character.

xi

CHAPTER I

Introduction

The human DNA undergoes several thousand to a million damaging events each

day, generated by either external or internal metabolic processes. These damage events

can be caused by either gamma rays resulting from the decay of naturally occurring

radionuclides in the earth or by reactive oxygen species such as hydrogen peroxide or

superoxide anions (Chance et al., 1979). In response to DNA damage, a cell may arrest its progression through the cell cycle in order to repair the lesions (Kuerbitz et al., 1992).

Successful repair is essential for its survival and the cell resumes cycling. However, when the DNA damage overwhelms the DNA repair machinery’s capacity, an apoptotic signal is triggered leading to programmed cell death (Figure 1.1). This chapter describes the three cellular processes that are involved in the cellular response to DNA damage: cell cycle arrest, DNA repair, and apoptosis.

1 2

Figure 1.1 Cellular response to DNA damage. Upon infliction of DNA damage, the cell undergoes cell cycle arrest to allow the damage to be repaired. Depending on the severity of the lesion, the cell may be able to repair it, survive and resume cycling. On the other hand, when the damage is too extensive, the cell may choose to undergo apoptosis.

3

The Cell Cycle

The cell cycle is a fundamental cellular process characterized by a highly

conserved and ordered set of events, culminating in cell growth and division. It ensures

propagation of life from the unicellular to the multicellular organisms (Murray, 1993). In

order to generate a daughter cell, the parental cell has to undergo several changes among

which are, growth in mass, duplication of organelles, duplication and equal separation of

DNA in the two daughter cells (Mitchison, 1971). These processes take place in well

defined phases of the cell cycle. Generally the cell cycle can be broken down into

and division or the mitotic (M) phase. During the interphase the cell goes

through elaborate preparation for its division including synthesis of necessary proteins

and DNA replication. The division consists of the breakdown of the nuclear envelope,

segregation of the chromosomes by the mitotic spindle and the actual separation of the

daughter cells or . Regulation of the cell cycle is critical for the normal

development of organisms, and loss of control could lead to cell death or onset of cancer

(Sherr, 1996).

The cell cycle can be divided into four stages: gap 1 (G1), synthesis (S), and gap 2

(G2), which form the interphase and the (M) (Figure 1.2) (Baserga, 1985). The progression through these phases is governed by a precise series of events that allow the cell to grow and prepare for its division (Johnston, 1990).

4

Figure 1.2 Schematic representation of the cell cycle. During the the cell grows is size preparing for the DNA synthesis that takes place in the . After the sister chromatid has been synthesized, the cell passes through another gap phase, G2, ensuring that the environment is optimal for division. During the M phase, the chromatin condenses and the resulting chromosomes are evenly separated by the mitotic spindle into the two new daughter cells. The cells may choose to differentiate and thus stop cycling by entering the .

5

During the G1 phase, the cell monitors its environment and size and faces the decision of either preparing for S phase or to differentiate and exit the cell cycle. Cells that leave the cell cycle in G1 in order to differentiate are found in G0. This quiescence may be permanent or temporary as they may re-enter the cell cycle at a later time. After the G1 phase, DNA replication occurs in S phase.

The provides additional time for growth, with cells preparing for the M

phase. After the G2 phase, nuclear and cytoplasmic division occurs in the M phase. The

M phase is subdivided into , , , and . Each of these

cell cycle phases corresponds to specific events of the . During prophase, the

chromatin is condensed and individual chromosomes become visible. In metaphase, sister

chromatids are paired and aligned at the center of the cell in what is called the metaphasic

plaque. In anaphase, the chromosomes are separated by being pulled to the opposite sides

of the cell by mitotic spindle fibers which are attached at their kinetochores. The mitotic

spindle disassembles and the chromosomes decondense during telophase. The physical

division of the cytoplasm containing the organelles takes place during cytokinesis

(Nasmyth, 2001).

During both G1 and G2 phases there are checkpoints whose role is to ensure the

DNA integrity (Andreassen et al., 2003). When challenged with DNA damage, a cell may decide to arrest its cycling to repair the lesion or to undergo other events that lead to apoptosis. Another role of the checkpoints is to ensure that the molecular events specific to a certain cell cycle phase have taken place before progressing to the next, downstream process (Hartwell and Weinert, 1989). Furthermore, they allow the system to integrate 6

and be controlled by signals from the environment (e.g. presence of mitogens). For example, the G1/S phase progression is controlled by the G1 checkpoint, also called the , whose role is to ensure that the cell is big enough and that the environment is favorable for DNA synthesis. Rapidly replicating human cells go through a round of division in approximately 24 hours: G1 lasting ~10 hours, S phase 10 hours,

G2 ~3.5 hours, and M ~ 30 minutes.

The tight control of the cell cycle is achieved through the temporal expression of cyclins which bind and activate their catalytic partners, the cyclin-dependent kinases

(CDK), and other accessory proteins important for their stability and activity, the cyclin- dependent kinase inhibitors (Murray and Kirschner, 1989; Murray and Kirschner, 1991).

The first molecular insights into understanding the molecular mechanisms governing the cell cycle emerged from studies of the M-phase Promoting Factor (MPF) or the Cyclin

B1/Cdk1 complex (Masui and Markert, 1971). Cyclins have originally been identified by studies of the cyclic synthesis and degradation of proteins in the eggs of the sea urchin

Lytechinus pictus (Evans et al., 1983).

The G1 cyclins, D and E were identified by homology screening, subtractive hybridization following stimulation with colony-stimulating factor-1 (Matsushime et al.,

1991) and yeast complementation assays (Koff et al., 1991; Lew et al., 1991; Xiong et al.,

1991). family of proteins consisting of , D2, and D3 can interact with either Cdk4 or Cdk6. They regulate entry and progression through the cell cycle in response to mitogenic stimulation. Their main role is to phosphorylate and inactivate the cell-cycle inhibitory function of the (pRB). The other G1 cyclin, 7

Cyclin E, interacts with Cdk2 and stimulates the G1/S phase transition. The activity of the G1 cyclins and CDKs may be blocked by the CDK inhibitors proteins (CIPs), p27Kip1, p57Kip2, p21CIP/Waf1, and the family of proteins. Another regulatory protein, Cdc25A phosphatase, positively regulates activity of /Cdk2 and Cyclin E/Cdk2 complexes by removing the inhibitory phosphorylation on Cdk2 (Smits and Medema,

2001).

Cyclins A and B bind and regulate the kinase activity of Cdk1 (Minshull et al.,

1990) being the mammalian equivalent of MPF. Cyclin A expression starts at the beginning of the S phase and peaks during G2. As opposed to , it also interacts with Cdk2 during the S phase. starts to be synthesized in late S phase, peaks during metaphase and decreases at the end of anaphase. The temporal destruction of

Cyclin B is performed by Anaphase-Promoting Complex (APC) ubiquitin ligase (Glotzer,

1995). The APC catalyzes the addition of polyubiquitin chains to Cyclin B, targeting it to proteasomal degradation.

The cell cycle progression is also regulated at the transcriptional level. Several genes important for cell division have been found to be controlled by the family of transcription factors (Mudryj et al., 1991). For example, their transcriptional activity is important for the regulation of Cyclin E, dihydrofolate reductase (DHFR), Cdc6, and the minichromosome maintenance (MCM) proteins (Dyson, 1998). E2F’s cell cycle regulatory function resulted from studies in which E2F complexes were dissociated by

E1A (early region 1A product of adenovirus 5), which resulted in a loss of cellular proliferation control (Chellappan et al., 1991). Another cell cycle regulatory protein, the 8

retinoblastoma tumor suppressor protein (pRb), was found to interact with E2F

(Chellappan et al., 1991) and form a complex that acts as a growth suppressor and prevents progression through the cell cycle (Classon and Harlow, 2002). The pRb-

E2F/DP (dimerization protein) complex also attracts a histone deacetylase (HDAC) protein to the chromatin, further suppressing DNA synthesis. The E2F regulation in normal cells was first uncovered by studies using adenovirus E1A 12S product to dissociate E2F from pRb, leading to activation of E2F (Nevins, 1992).

Cyclin E and cell cycle regulation

The main families of regulatory proteins that play key roles in controlling the cell

cycle progression are the cyclins, the cyclin dependent kinases (CDKs) and, as their

critical downstream substrates, the products of retinoblastoma

(pRb) and . The activity of CDKs oscillates, leading to the cyclical changes in the

activation of key proteins, which regulate the main cell cycle processes: DNA replication,

mitosis, and cytokinesis. G1/S-cyclins commit cells to DNA replication, which is driven

by the S-phase cyclins, whereas mitosis is governed by M-cyclins. Progression through

the early G1 phase is regulated by signal transduction cascades activated by polypeptide

growth factors and by extracellular matrix components.

The human G1 cyclins, the D- and E-type cyclins, were identified functionally by

screening of human cDNA libraries for sequences that could complement G1 cyclin

mutations in S. cerevisiae (Koff et al., 1991; Lew et al., 1991). This indicated that Cyclin

E plays an important role in the regulation of the cell cycle progression. In fission yeast 9

(Schizosaccharomyces pombe) cells have to reach a critical size before entering mitosis, which is achieved by associating Cdk1 activation with cell growth. Premature activation of the mitotic CDK (Cdk1) leads to mitosis entry at a reduced size. A similar phenomenon takes place in mammalian cells when cyclin E is overexpressed. The cells have a shorter G1 and enter mitosis at a reduced size (Ohtsubo and Roberts, 1993;

Resnitzky and Reed, 1995). Furthermore, Cyclin E may replace all functions of Cyclin

D1 indicating that it is one of its downstream targets as well as a key regulator of the S phase entry (Geng et al., 1999). Together with the importance of Cyclin E in G1/S transition, these data indicate that Cyclin E is playing an important role in critical limiting steps which are essential for S phase entry.

The human Cyclin E has several forms and splice variants, with the predominant species encoding a 395 amino-acid protein with a molecular weight of 50-55 kDa (Koff et al., 1991; Lew et al., 1991). The E-type cyclin family consists of two members, Cyclin

E1 (Cyclin E) and E2. also has a typical cyclin box motif and shares 47% similarity with . The two proteins display a similar cycle-regulated expression pattern as well as biochemical properties. Cyclin E mRNA levels have a cyclic pattern of fluctuation, with synthesis starting during the G1 phase and reaching a maximum in late

G1 followed by a down-regulation in the S phase (Dulic et al., 1992; Koff et al., 1992;

Lew et al., 1991). The Cyclin E expression oscillates throughout the cell cycle reaching a peak at the G1/S transition and a minimum in the M phase (Dulic et al., 1992; Koff et al.,

1992) (Figure 1.3). 10

Figure1.3 Periodic oscillation in the expression levels of cyclins. Cyclin D partners

with Cdk4 and Cdk6 in early to mid-G1 phase to phosphorylate and inactivate the cell-

cycle inhibitory function of the retinoblastoma protein. Cyclin E levels peak at the G1/S

boundary where it exerts its roles in the G1/S phase transition. Cyclin A synthesis starts at the beginning of the S phase and reaches its peak at the beginning of the G2 phase while the mitotic Cyclin B levels attain a maximum during the M phase being required for the cell division.

11

Cyclin E is synthesized at the G1/S boundary under the influence of the E2F transcription factor family (Botz et al., 1996; Geng et al., 1996; Ohtani et al., 1995).

By interacting with and activating its catalytic partner Cdk2, Cyclin E is essential for regulating the G1/S transition (Ohtsubo et al., 1995; Resnitzky et al., 1994; Tsai et al.,

1993; van den Heuvel and Harlow, 1993) and the initiation of DNA replication (Jackson et al., 1995; Krude et al., 1997; Ohtsubo et al., 1995; Roberts and Sherr, 2003). Cyclin E2 was also found to interact with and activate Cdk2 forming a complex that can be inhibited by p27 (Gudas et al., 1999; Lauper et al., 1998; Zariwala et al., 1998). Both proteins were found to be redundant in development (Parisi et al., 2003). Cyclin E, unlike any other cyclin, can complement deficiencies of other cyclins in yeast (Koff et al., 1991;

Lew et al., 1991) and mouse (Geng et al., 1999) and it has unique and highly specialized functions. Cyclin E promotes events associated with genome replication (Reed, 1996;

Sauer and Lehner, 1995). These include regulation of key steps in the initiation of DNA replication through E2F regulation and thus of its critical S-phase target genes, CDC6 and loading of MCM and CDC45 to replication origins. Cyclin E/Cdk2 phosphorylates two of its own regulators: Cdc25A (Hoffmann et al., 1994) and p27Kip1 (Sheaff et al., 1997;

Vlach et al., 1997). Phosphorylation by Cyclin E/Cdk2 also regulates histone synthesis

(through p220/NPAT) (Ma et al., 2000; Zhao et al., 2000), centrosome duplication

(through nucleophosmin) (Okuda et al., 2000), chromatin modifications by p300/CBP

(Ait-Si-Ali et al., 1998) and SWI/SNF (Shanahan et al., 1999), pre-mRNA splicing

(through SAP155/SF3B1) (Seghezzi et al., 1998) and DNA replication (Ewen, 2000). 12

The other homologous protein, Cyclin E2, is believed to have similar functions

(Mazumder et al., 2004; Moroy and Geisen, 2004).

Cyclin/CDK complexes are negatively regulated by two families of CDK

inhibitors (Sherr and Roberts, 1995). The D-type cyclin/CDK complexes may be

inhibited by both Ink4 as well as Kip/Cip family of inhibitors, whereas the E-type cyclin/CDK complexes are inhibited exclusively by the latter one. p21Cip1 and p27Kip1 bind and inhibit Cyclin E/Cdk2 complexes by competing with their substrates but also by preventing the cyclin-activating kinase-mediated phosphorylation as well (Morgan, 1996;

Sherr and Roberts, 1995). Consequently, cells that lack these two proteins manifest a significant increase in the kinase activity of the Cyclin E/Cdk2 complex (Aleem et al.,

2005).

A number of biological functions have been attributed to Cyclin E, based mainly on its interaction with and phosphorylation of its substrates. Thus, Cyclin E/Cdk2 complexes have been shown to play an essential role in the initiation of DNA replication

(Jackson et al., 1995), in addition to the cell cycle transition (Ohtsubo et al., 1995;

Resnitzky et al., 1994). While Cyclin E complexes phosphorylate pRb, which is also phosphorylated by the D-type Cyclin-Cdk4/6 complexes (Dowdy et al., 1993) it is likely that other substrates exist during late G1. Unlike Cyclin D, Cyclin E remains essential in the absence of pRb, since: (i) its inducible expression in fibroblasts accelerates G1/S progression without affecting the kinetics of pRb phosphorylation (Resnitzky and Reed,

1995); (ii) unlike the D-type cyclins, Cyclin E is essential for cell-cycle progression in pRb-deficient cells (Ohtsubo et al., 1995); (iii) ectopic expression of cyclin E bypasses 13

pRb-mediated cell cycle arrest (Alevizopoulos et al., 1997; Lukas et al., 1997); and (iv)

Cyclin E is required for S phase entry in Drosophila (Duronio et al., 1996) and Xenopus

(Hua et al., 1997). These findings highlight a fundamental difference between the Cyclin

D and Cyclin E complexes, strongly suggesting that other key rate-limiting substrates

exist for Cyclin E/Cdk2 (Alevizopoulos et al., 1997; Lukas et al., 1997). Constitutive

activation of Cyclin E/Cdk2 results in uncoupling of the initiation of centrosome and

DNA duplication leading to unscheduled initiation of centrosome duplication prior to S-

phase entry (Mussman et al., 2000).

The Cyclin E/Cdk2 complex was thought to be a critical regulator of the somatic

cell cycle. Surprisingly, ablating these genes in the mouse had very limited consequences.

Embryonic fibroblasts deficient in Cdk2 had normal proliferation and become immortal

after continuous passage in culture. Cdk2-/- mice are viable and can survive for up to two years, suggesting that Cdk2 may not be necessary for proliferation and survival of most cell types. It was found, however, to be necessary for completion of prophase I during meiotic cell division in male and female germ cells (Berthet et al., 2003; Ortega et al.,

2003). Similarly, it was demonstrated that E-type cyclins are largely dispensable for mouse development (Geng et al., 2003). Nevertheless, in the absence of Cyclin E, endoreplication of trophoblast giant cells and megakaryocytes is severely impaired.

Cyclin E-deficient cells proliferate normally under conditions of continuous cell cycling but are not able to re-enter the cell cycle from the quiescent G0 state. Molecular analyses revealed that cells lacking Cyclin E fail to normally incorporate MCM (minichromosome maintenance) proteins into DNA replication origins during G0 to S-phase progression. 14

These findings define a molecular function for E-type cyclins in cell cycle re-entry and

reveal a differential necessity for Cyclin E in normal versus oncogenic proliferation

(Geng et al., 2003).

In addition to the well characterized Cdk2-dependent roles in the cell cycle,

Cyclin E was also found to be involved in the G1/S progression in a kinase-independent

manner (Geng et al., 2007). This study showed that by interacting with CDT1 and MCM,

a kinase deficient Cyclin E mutant could restore the MCM replicative helicase loading

and cell cycling in Cyclin E-deficient mouse embryonic fibroblasts. Another kinase-

independent function of Cyclin E is at the G1/S transition by localizing at the

centrosomes in a Cdk2-independent manner (Matsumoto and Maller, 2004).

Transcriptional regulation of Cyclin E

Cyclin E is a regulator of proliferation of mammalian fibroblasts and thus, its

levels are tightly regulated (Koff et al., 1992; Lam and La Thangue, 1994; Nevins, 1992).

Cyclin E/Cdk2 activity is precisely regulated at multiple levels, including transcriptional

and post-transcriptional, by binding of Cdk inhibitors such as the members of the Cip/Kip family ( and p27) as well as through modification of the Cdk activity by inhibitory or activating phosphorylation (Hwang and Clurman, 2005). Normally, expression of Cyclin

E is tightly controlled so that its levels reach a peak at the G1/S phase transition and then decline to an eventual loss of expression during S phase (Dulic et al., 1992; Ekholm et al.,

2001; Koff et al., 1992). This expression pattern is achieved by regulation at both

transcriptional as well as post-transcriptional levels. There are a number of putative 15

binding sites for E2F in the cyclin E promoter region, suggesting an E2F-dependent

regulation (Le Cam et al., 1999). A variant E2F binding site is a cyclin E repressor

module responsible for the periodic down-regulation of the cyclin E promoter until the

growing cells have reached the late G1-phase (Le Cam et al., 1999). This site facilitates

transcriptional repression by binding to a large complex, containing E2F4, DP1, and a pocket protein, whose role is to delay the expression of Cyclin E until late G1 (Le Cam et al., 1999; Polanowska et al., 2001) (Figure 1.4). pRb forms a repressor complex with a histone deacetylase (HDAC) and the SNF2-like (BRG1 and hBRM) component of the mammalian hSW1/SNF nucleosome remodeling complex, starting from the end of S- phase until late G1 (Zhang et al., 2000). The SW1/SNF complexes have an important role in transcriptional regulation, altering the chromatin structure by relieving the transcription from the nucleosome-mediated repression, thereby, opening access to the activators of transcription. The phosphorylation of pRb by Cyclin D/Cdk4 abrogates its interaction with HDAC and transactivates Cyclin E and, thereby, overcomes the G1 arrest

(Zhang et al., 2000). Cyclin E/Cdk2 can phosphorylate pRb or the hSW1/SNF component after Cyclin E reaches a certain level, when the interaction of pRb-hSW1/SNF is disrupted.

Deregulation of any of the components of this transcriptional complex could lead

to the unscheduled expression of Cyclin E, which is very common in cancer cells. Our

laboratory and others have previously shown that constitutive levels of several E2F1

target genes are increased in Rb-deficient fibroblasts (Almasan et al., 1995b; Herrera et

al., 1996). Similarly, Cyclin E levels are increased constitutively when Rb has been 16

inactivated by the HPV16-E7 expression in human foreskin fibroblasts (Martin

et al., 1998).

The above reports suggest that the transcription factor responsible for Cyclin E

induction might be E2F. E2F1 has been reported to be upregulated in response to DNA

damage in a manner analogous to p53, with its levels being unchanged following DNA

damage of epithelial cells with mutated p53 (Blattner et al., 1999). In contrast, we found that Cyclin E was upregulated in cells with mutated p53 expressed either endogenously or following its stable transfection (Mazumder et al., 2000). Northern and run-on analyses have indicated that the Cyclin E levels induced by radiation result mostly from transcriptional regulation. Cyclin E induction is not restricted to radiation and the hematopoietic cell lines we have investigated. Cyclin E levels were also increased following treatment with chemotherapeutic agents, such as the topoisomerase II inhibitor

VP16 (Mazumder et al., 2000). In addition, we recently found that Cyclin E is also upregulated in other cell types, such as those of prostate and epithelial cell types.

Although further work is required to establish whether E2F plays a regulatory role in radiation-induced Cyclin E expression, it is quite possible that Cyclin E is regulated by different mechanism by mitogens and genotoxic stress such as radiation. Understanding how this regulation takes place may provide additional targets for therapy.

The oscillations in CDK activities are largely determined by the levels of their interacting partners, which in turn depend on their synthesis, through transcriptional regulation, and proteolytic degradation. Phosphorylation by Cyclin H/Cdk7 and

Wee1/Myt1 kinases and dephosphorylation by the KAP, Cdc25A, and PP2C phosphatase 17

families play another important role in the regulation of the cell cycle machinery (Cheng et al., 2000; Donzelli and Draetta, 2003; Lew and Kornbluth, 1996; Moroy and Geisen,

2004; Parker et al., 1995; Poon and Hunter, 1995). In normal cells, Cdc25A is negatively regulated by stress signals such as irradiation through checkpoint-mediated ubiquitination. In checkpoint-deficient cells, Cdc25A is overexpressed, leading to radioresistant DNA synthesis (Falck et al., 2001).

18

Figure 1.4 Schematic representation of the progression through the different cell

cycle phases. Cyclin D family of cyclins activate Cdk4 and Cdk6 to phosphorylate and

inactivate pRb and its related family members (p107 and p130). This phosphorylation event results in the release of the E2F family of transcription factors which can increase the levels of proteins essential for the S phase progression [Cyclin E, thymidylate synthase (TS), dihydrofolate reductase (DHFR)]. The INK4 family of inhibitors bind and inhibit the Cyclin D/Cdk4, 6 complexes while p21 and p27 cyclin-dependent kinase inhibitors (CKI) form inhibitory complexes with Cyclin E/Cdk2. Whenever the cell is challenged with genotoxic stress caused by DNA-damaging agents like ionizing radiation, a signaling pathway triggers the cell cycle arrest to allow the DNA repair to take place. The damage is sensed by several kinases including ATM (Ataxia telangiectasia mutated) and the signal is relayed either directly or indirectly by means of

Chk1 or Chk2 kinases to the p53 tumor suppressor protein. Induction of p53 leads to increased transcription of the CKIs, which trigger cell cycle arrest.

19

20

Post-translational regulation of Cyclin E

Cyclin E periodicity is not only determined at the transcriptional level, but it is also maintained by post-translational regulation mechanisms through ubiquitin-dependent proteolysis (Clurman et al., 1996; Won and Reed, 1996). Ubiquitin-mediated protein degradation is a regulatory mechanism of a wide variety of cellular and tissue processes

such as cell stress response, DNA repair, transcription, cell cycle regulation, and cellular transformation (Hershko and Ciechanover, 1998).

The programmed degradation of many proteins is performed by the 26S proteasome. This recognizes a degradation signal consisting of polyubiquitin chains attached to lysine residues of the substrate by a cascade of enzymes, generically named

E1, E2 and E3. This ubiquitination is carried out by the successive action of ubiquitin- activating (E1), ubiquitin-conjugating (E2) and ubiquitin-protein ligase (E3) enzymes

(Hershko and Ciechanover, 1998; Jentsch, 1992)}. The E1 activates the ubiquitin (an

ATP-dependent process) and transfers it to the E2 through the formation of a thioester bond between the E2 active-site cysteine and the ubiquitin carboxy terminus (Pickart and

Rose, 1985). E3s bind to both a cognate E2 and the substrate and in the case of the

RING-E3 family they catalyze the ubiquitination of the substrate lysines directly by the

E2. E3-substrate association is the primary determinant of substrate specificity and is

often regulated by phosphorylation or other post-translational modification of the

substrate’s E3 binding motif, termed degron (Ang and Wade Harper, 2005; Skowyra et

al., 1997). Finally, the 26S proteasome recognizes the polyubiquitin chain and degrades 21

the substrate into small peptides recycling the ubiquitin molecules at the same time

(Jentsch, 1992).

One major class of E3 ligases includes those that contain proteins called .

The component of these large complexes serves as a scaffold for the assembly of a substrate recognition subunit as well as an E2 ubiquitin-conjugating enzyme (Deshaies,

1999). For example, the Cul1-dependent E3 ligase, also known as the SCF complex

(Skp1-Cul1-F box), consists of an adapter protein called Skp1, a ring-finger protein

Rbx1/Roc1, and a substrate recognition protein that contains a motif called an F box

(named after a common sequence identified in Cyclin F (Bai et al., 1996)). The human F

box protein responsible for the turnover of Cyclin E is Fbw7 (also known as hCdc4,

hSEL-10, and hAgo) (Gupta-Rossi et al., 2001; Koepp et al., 2001; Moberg et al., 2001;

Strohmaier et al., 2001). Fbw7 is expressed as three splice-variant isoforms which differ from each other only by their amino terminus (Spruck et al., 2002). Each splice variant is comprised by ten common 3′ exons and a unique 5′ exon. The common exons encode two domains critical for hCdc4 function. The function of the first domain, known as the F box

(Bai et al., 1996), is to interact with Skp1, thus associating the F box protein with the

Cul1/Cdc53 scaffold. The second domain is comprised of eight repeats of the WD40 motif (van der Voorn and Ploegh, 1992). These fold adopting a structure termed β propeller, also confirmed by the crystal structure of the yeast homolog of hCdc4 (Orlicky et al., 2003). The role of the WD40 is to recognize specific substrates with which it interacts. The hallmark of substrate recognition by the SCF complex is that substrate phosphorylation is a requirement (Figure 1.5). 22

Figure 1.5 Schematic representation of the Cyclin E turnover mechanisms. Cdk2- bound Cyclin E requires at least two phosphorylation events by GSK3β (at Thr380) and by Cdk2 (at Ser384) in order to be recognized by Fbw7. The Skp1-Cul1-Rbx1-Fbw7 complex recognizes phosphorylated Cyclin E and polyubiquitinates it. Ubiquitin is provided by the E2 Ubiquitin-conjugating enzyme, Ubc. On the other hand, free Cyclin E does not require any phosphorylation and is ubiquitinated by a Cul3-based complex. The additional factors that form this complex have not been identified yet, but may involve

BTB proteins as specificity factors.

23

24

In the cell cycle, ubiquitination plays a central role in the cell-cycle transitions

and checkpoints by establishing the strict temporal control of proteins such as cyclins and

cyclin-dependent kinase inhibitors (Sherr and Roberts, 1999). Cyclin E accumulation is

caused in part by the inactivation of SCFFbw7 ubiquitin-protein ligase that mediates its ubiquitination (Koepp et al., 2001; Strohmaier et al., 2001). Fbw7 was found to be mutated in several tumors and tumor cell lines (Hubalek et al., 2004; Moberg et al., 2001;

Rajagopalan et al., 2004; Spruck et al., 2002; Strohmaier et al., 2001). The Fbw7 gene is located in the 4q32 chromosomal region, which has been reported to be deleted in as many as 30% of human tumors (Spruck et al., 2002). Its inactivation leads to chromosomal instability (Rajagopalan et al., 2004), while its loss in mice can lead to embryonic lethality (Tetzlaff et al., 2004; Tsunematsu et al., 2004).

Degradation of Cyclin E involves both Cul1 and Cul3 proteins. Cul1 knock-out

mice die early during development with a subset of cells containing high levels of Cyclin

E (Dealy et al., 1999; Wang et al., 1999). Cul3-/- mice also arrest early in development.

However, a different subset of cells show increased levels of Cyclin E. Cul3 has been shown to bind and ubiquitinate Cyclin E in a transient transfection system (Singer et al.,

1999). It was shown that Cul3 is responsible for a constitutive pathway of Cyclin E degradation, which did not require binding to Cdk2 (Clurman et al., 1996; Singer et al.,

1999) or phosphorylation. On the other hand, Cul1-dependent turnover of Cyclin E is dependent on its association with Cdk2 and on its GSK3β-mediated (Welcker et al.,

2003) and auto-phosphorylation of Cyclin E at a series of serines and threonines

(Strohmaier et al., 2001; Yeh et al., 2001). 25

The primary Cyclin E recognition site by SCFFbw7 is Thr380, which is phosphorylated in vivo by GSK3β resulting in a phosphodegron sequence (Clurman et al., 1996; Welcker et al., 2003; Won and Reed, 1996). The unbound Cyclin E is degraded by the proteasome, and binding to Cdk2 protects it from degradation. However, phosphorylation of Cyclin E on Ser384 by its Cdk2 subunit has also been shown to be important for Fbw7-mediated Cyclin E degradation (Welcker et al., 2003). Thus, Cyclin

E/Cdk2 activity reverses the stabilizing effect of complex assembly. Skp1 and

Fbw7/Cdc4 are responsible for ubiquitin-dependent proteolysis of Cyclin E. Skp2 was thought earlier to be the F-box protein responsible for Cyclin E degradation since Skp2 gene inactivation by homologous recombination results in p27 and Cyclin E accumulation (Bai et al., 1996; Winston et al., 1999). However, since p27 is the predominant substrate of Skp2, its accumulation leads indirectly to the inhibition of

Cyclin E/Cdk2 kinase activity and phosphorylation of Cyclin E on Thr380, thus preventing its recognition by the Cyclin E-specific F-box protein. Instead, Fbw7, specifically targets ubiquitin-mediated proteolysis of Cyclin E (Koepp et al., 2001;

Strohmaier et al., 2001). The interaction of Cyclin E with this F-box protein depends on

Cyclin E phosphorylation by GSK3β on Thr380 (Welcker et al., 2003) and Thr62 autophosphorylation (Won and Reed, 1996). Some tumor cell lines that have high levels of Cyclin E also have mutations in the Fbw7 gene or express low levels of its mRNA, suggesting that Fbw7 may function as a tumor suppressor (Ekholm-Reed et al., 2004;

Spruck et al., 2002; Welcker and Clurman, 2008). 26

Recent investigation of the Cyclin E turnover mechanism determined that only α

and γ Fbw7 isoforms are required for this process (van Drogen et al., 2006). Specifically,

Fbw7γ (localizing in the nucleoli) is directly involved in the ubiquitination of Cyclin E

while Fbw7α (localized in the nucleoplasm) plays a regulatory function facilitating this

event. Fbw7α serves as cofactor for the prolyl isomerization of the Cyclin E carboxy-

terminal phosphodegron performed by the peptidyl prolyl cis-trans isomerase Pin1 (van

Drogen et al., 2006). Apart from Cyclin E, the SCFFbw7 has also been shown to bind and ubiquitinate Notch1 (Wu et al., 2001), c- (Welcker et al., 2004), and c-Jun (Wei et al., 2005), which further supports the role of Fbw7 as a tumor suppressor.

The cyclin E gene is under direct control by the E2F family of transcription factors, which places cyclin E transcription under the control of the retinoblastoma protein (pRb) and its upstream regulators (Botz et al., 1996; Duronio et al., 1996; Geng et al., 1996; Ohtani et al., 1995). Inactivating Rb as part of the normal cell cycle progression

or by viral oncoproteins leads to its elevated transcription (Weinberg, 1995). The large T

antigen (LT) of the simian virus 40 (SV40) disrupts the cellular checkpoint mechanisms

that regulate cell division, DNA transcription, replication, and repair. The key regulators

that are targeted by LT are p53 and Rb. This binds directly to pRb and its family member

pocket proteins (p107 and p130) via an N-terminal LxCxE motif. In the case of p53, LT

binds the DNA binding domain of p53 blocking the p53-dependent growth arrest and

apoptosis (Levine, 1997). Another way LT interferes with the normal activity of the cell

is apparently through binding Fbw7 and mislocalization of the Fbw7γ isoform from the

nucleoli to the nucleoplasm (Welcker and Clurman, 2005). This way LT may interfere 27

with the timely Cyclin E turnover in a substrate-like manner, thus leading to increased

Cyclin E-associated kinase activity. Unlike typical Fbw7 substrates, LT is not degraded

by the SCFFbw7, but rather acts as a competitive inhibitor.

Cross-talk between the regulation of cyclin E transcription and that of Cyclin E protein stability results in fine tuning of Cyclin E levels, emphasizing its crucial role in cell cycle regulation and predicting deleterious effects of a constitutively high level of expression of Cyclin E, as seen in many cancer cells.

Deregulated expression of Cyclin E in cancer

Normal cell proliferation is under strict regulation, governed by checkpoints

located at distinct positions in the cell cycle. Cell cycle progression is controlled by two

major checkpoints, one at the G1/S boundary, when cells commit to DNA replication,

with the other one at the G2/M boundary during the commitment of cells to mitotic

division (O'Connor et al., 2000). The G1/S transition is regulated by the Restriction point

(R point), which is a point of no return, such that, if the cell passes that point, it no longer

needs mitogens and is committed to completion of DNA replication and the cell cycle

(Pardee, 1989). Any type of deregulation of the G1/S transition, along with the

disappearance of the R-point, is a hallmark of cancer, leading to uncontrolled cell

proliferation. The periodic appearance of Cyclin E coincides precisely with the timing of

the R point. In contrast to normal cells, mitotic cyclins appear prior to G1 cyclins in

tumor cells (Keyomarsi and Pardee, 1993). Another view is that passage through the R

point is a prerequisite for Cyclin E accumulation (Ekholm et al., 2001). This study found 28

that the post-mitotic G1 cells that had not yet reached R were negative for Cyclin E

accumulation, while cells that had passed R accumulated Cyclin E at variable times (1 to

8 hours) after passage through R and 2 to 5 hours before entry into the S phase.

As these cyclins and CDKs are important components of cell cycle control and cell proliferation, alterations or mutational changes in the expression of the corresponding

genes leads to oncogenesis (Evan and Vousden, 2001; Hunter and Pines, 1994; Pardee,

1989; Sherr, 1996). Cyclin E has been shown to be deregulated and overexpressed in

several solid tumors, including breast, colon, and prostate carcinomas (Akli and

Keyomarsi, 2003; Erlandsson et al., 2003; Keyomarsi et al., 1995; Keyomarsi et al.,

1994; Keyomarsi et al., 2002; Moroy and Geisen, 2004; Sherr, 1996). High levels of

cyclin E expression have been associated with the progression of different other tumors,

such as leukemias and lymphomas. Loss of Cyclin E regulation has been associated with

aggressive disease and poor patient outcome (Donnellan and Chetty, 1999; Hwang and

Clurman, 2005; Sandhu and Slingerland, 2000). This was shown to take place either by

amplification of the gene or by its overexpression.

Overexpression of Cyclin E results in accelerated G1/S phase progression

(Ohtsubo and Roberts, 1993; Ohtsubo et al., 1995; Resnitzky et al., 1994). Transgenic

mouse models with constitutive overexpression of Cyclin E develop malignant diseases,

indicating that Cyclin E is a dominant oncoprotein (Karsunky et al., 1999; Moroy and

Geisen, 2004). For example, overexpression of Cyclin E in the mouse mammary

epithelium is associated with hyperplasia, which can ultimately lead to tumorigenesis

(Bortner and Rosenberg, 1997). Furthermore, cell lines derived from tumors 29

overexpressing Cyclin E rarely show amplification of the gene, suggesting that the deregulation is at the post-transcriptional level (Porter et al., 1997), specifically in the turnover process. Cul3-dependent degradation of Cyclin E is important for the maintenance of quiescence of hepatocytes (McEvoy et al., 2007). Moreover, Cyclin E- deficient mice show increased resistance to oncogenic transformation by c-Myc, H-Ras, and dominant-negative p53 (Geng et al., 2003). Deregulation of Cyclin E was also shown to promote chromosome instability, detected as both chromosome losses and gains

(Spruck et al., 1999). This may result in tumorigenesis by facilitating amplification of or loss of heterozygosity at tumor suppressor loci.

Although the protein is both synthesized and degraded in the cytoplasm, it exerts its functions in the nucleus where it translocates. In the nucleus, the nascent Cyclin E associates with its Cdk2 partner, activating its serine/threonine kinase activity shortly before entry into S phase (D'Urso et al., 1990; Koff et al., 1992). Accumulation of Cyclin

E in the cytoplasm either reflects increased synthesis, decreased degradation, or failure of

transport to the nucleus. In some types of cancer, the cyclin E gene (CCNE1) was found

to be frequently amplified (Schraml et al., 2003). In others, however, the post-

translational regulatory mechanism is impaired as mutations have been identified in the

cellular machinery responsible for Cyclin E proteolysis (Calhoun et al., 2003; Ekholm-

Reed et al., 2004; McVey et al., 1989; Rajagopalan et al., 2004; Spruck et al., 2002;

Strohmaier et al., 2001). Loss or low p27 expression as well as overexpression of Cyclin

E or Cdk2 are significantly associated with malignancy in ovarian cancers (Porter et al.,

1997). 30

Levels of Cyclin E and its low molecular weight derivatives in these tumor tissues

correlate strongly with survival in these patients (Keyomarsi et al., 2002). There can be

significant changes of Cyclin E or Cdk2 levels during carcinogenesis. During

progression from the primary to the lymph node-metastatic foci, the levels of Cyclin E

protein remain the same, while Cdk2 levels increase significantly (Li et al., 2001a).

However, during a similar transition from the primary to the liver-metastatic foci, Cyclin

E levels are apparently reduced, and those of Cdk2 diminish almost completely.

Additionally, the decrease of Cyclin E is significantly associated with large tumor size

and lymph node metastasis in primary carcinomas, with large tumor size and hepatic

metastasis being strongly related to low Cdk2 levels. Induced Cyclin E protein is related

to increased Cdk2, which is further associated with Ki-67 staining. Thus, Cdk2

overexpression could facilitate lymph node metastasis and both Cyclin E and Cdk2

overexpression may trigger the progression of early cancer.

Cyclin E overexpression is apparent in 10-25% of breast cancers. It is also a

strong predictor of endocrine therapy failure (Span et al., 2003), and an accurate predictor of poor clinical outcome (Keyomarsi et al., 2002; Loden et al., 2002). Studies have found

Cyclin E to be processed into more active low molecular weight (LMW) forms (Porter et

al., 2001) that are resistant to p21 and p27 inhibition and therefore associated with higher

cyclin-dependent kinase activity (Akli et al., 2004). It was shown that the LMW isoforms

are more potent than the full length Cyclin E in enhancing angiogenesis and metastasis

(Bales et al., 2005). Different studies have found Cyclin E to be cleaved by elastase

(Porter et al., 2001) as well as calpain (Wang et al., 2003). Apparently Cyclin E is able to 31

transactivate calpain 2 transcription (Libertini et al., 2005) and thus regulates its own proteolysis as well of that of other calpain substrates such as focal adhesion kinase

(Cooray et al., 1996), α-spectrin (Stabach et al., 1997), talin (Franco et al., 2004), paxilin

(Yamaguchi et al., 1994), E-cadherin (Rios-Doria et al., 2003), and β-catenin (Rios-Doria et al., 2004).

In many human cancers Cyclin E is expressed at levels substantially higher than those in normal tissues and shows frequent amplification of the genomic locus at which the cyclin E gene is located (19q12-q13). In some tumors, cyclin E gene amplification and protein accumulation are late events, whereas in other neoplasms, the increase of

Cyclin E is observed during the early stages of malignancy. Increased protein expression does not necessarily reflect a consequence of mutations in the cyclin E gene. The question of whether Cyclin E is merely a link in the chain of events that leads to cell proliferation or it is the driving force for cell replication is difficult to ascertain, as it may be tumor dependent. Elevated transcript levels of cyclins E are found in breast, colorectal, lung, and ovary/uterus tumor samples as compared to levels in normal tissues. Its overexpression in breast carcinoma could pave the way for their genomic instability

(Carroll et al., 1999; Lingle et al., 1998). Expression levels of Cyclin E are most likely to be elevated in breast tumors that lacked the estrogen receptor as compared to breast tumors with the receptor and normal breast tissue (Keyomarsi et al., 1994; Nielsen et al.,

1996).

The status of cyclin E expression is also important for other cancer types, such as sarcomas, NSCLC, leukemias, and lymphomas for prognosis and staging of tumors 32

(Akama et al., 1995; Dong et al., 2000; Erlanson and Landberg, 2001). The finding that

cyclin E is expressed at high levels in several types of leukemias, chronic lymphocytic leukemia, Hodgkin’s and Non-Hodgkin’s lymphoma (Erlanson and Landberg, 2001;

Erlanson et al., 1998), point to a role of Cyclin E in the development of these neoplastic diseases. Cyclin E and HPV expression seem to also correlate very well, thus making

Cyclin E a good biomarker for infection and early stage of preneoplastic lesions (Martin et al., 1998). Cervical, prostate, breast (Erlandsson et al., 2003), and renal cell carcinoma

(65%) (Hedberg et al., 2002) as well as leiomyosarcomas (Noguchi et al., 2000) exhibit

high expression of Cyclin E. Cyclin E may be also used as an independent marker in the

prognosis of metastasis-free survival of lymph node-negative breast cancer (Kuhling et

al., 2003) as well as in hepatocellular carcinomas (Ohashi et al., 2001) and testicular

germ cell tumors (Sugimoto et al., 2002).

Therapeutic approaches involving cell cycle inhibitors

The deregulation of cell cycle checkpoints and the molecules associated with

them may transform a normal cell into a cancer cell. This most often occurs by interfering

with the basic cell cycle regulatory machinery to activate cell cycle entry that leads to

deregulation of the cell cycle in dividing somatic cells. Finding anticancer drug treatment

strategies tailored to modulate specific cell cycle components in particular tumors is of

great interest. Candidate targets for such strategies include critical cell cycle molecules

involved in the G1 to S phase or G2 to M phase transitions (Boonstra, 2003; Gali-

Muhtasib and Bakkar, 2002; Lee and Yang, 2003). S phase sensitization through G1/S 33

cyclin or E2F deregulation could be an effective therapeutic approach. Most tumor cells

are characterized by aberrations in the cell cycle control primarily incurred through

inactivation of pRb by the phosphorylation-induced hyperactive CDKs or inactivating

viral proteins (Senderowicz, 2003). Clearly, Rb deficiency allows cells to progress into the S phase inappropriately, conditions under which they are vulnerable to chemo and radiotherapy (Almasan et al., 1995a). Rb deficiency leads to increased expression of E2F target genes that include cyclin E. Increasing levels of cyclins using proteasome inhibitors and thus sensitizing target cells has been also used effectively to target S phase

cyclin complexes as an effective therapeutic approach to induce apoptosis (Bunn, 2004).

Uncontrolled CDK activity is often the cause of human cancer. The function of

CDKs is tightly regulated by cell cycle inhibitors such as p21Cip1/Waf1 and p27Kip1.

Following DNA damage or anti-mitogenic signals, p21 and p27 associate with

Cyclin/CDK complexes to inhibit their catalytic activity and induce cell cycle arrest.

Loss of function of these genes is often observed in different types of human cancers

(Facchinetti et al., 2004; Porter et al., 1997). Molecular targeting of the p21 and p27 inhibitors has been reported in epithelial cells. Thus, cleavage of p21Cip1/Waf1 and p27Kip1 releases Cyclin A/Cdk2 from inhibition raising its expression levels and induces apoptosis (Jin et al., 2000; Levkau et al., 1998; Poon and Hunter, 1998).

Deregulation of the cell cycle components is one of the hallmarks of neoplastic cells (Sherr and McCormick, 2002). However, the most common chemotherapeutic agents used to prevent the growth of cancer cells are also toxic to normal cells. In order to identify a novel cell cycle target that would be selective against cancer cells, 34

determination of the expression pattern as well as the mechanism of the target in normal

versus tumor cells will have to be pursued so that therapeutic application of that target could be achieved. Genetic abnormalities of CDKs (e.g.1, 2, 7), cyclins (e.g. A and E) and CKIs (p21 and p27) have been reported in human cancers. Enforced perturbations of

CDKs and their interacting partners can enhance neoplastic transformation and, thereby, making them extremely attractive targets for cancer therapy (Figure 1.6).

Structural studies of cyclins and CDKs have revealed that CDKs provide suitable drug targets given their more rigid structure and identifiable domains, such as that responsible for ATP-binding. In contrast, cyclins have a highly flexible structure that is not amenable for target development (Davies et al., 2002a; Davies et al., 2002b).

Inhibitors targeting CDKs have become of great interest in cancer therapy with more than

50 compounds being evaluated for antitumor activity, some of which are already in preclinical trials (Dai and Grant, 2003; Senderowicz, 2003). These inhibitors either target the CDKs themselves, through their ATP-binding site, or their upstream regulators. The first group includes flavopiridol and roscovitine, the second UCN-01, perifostine, and lovastatin (Senderowicz, 2003). Indolinones, UCN-01, and paullones, are currently tested in clinical trials (Dai and Grant, 2003; Senderowicz, 2003). While demonstrating clinical activity, neither acts specifically against Cdk2. Other more specific Cdk2 inhibitors are currently in preclinical development (Lane et al., 2001; Monaco et al., 2004; Yu et al.,

2003a). These include purine based CDK inhibitors, such as olomoucine, purvalanols, and roscovitine, which display greater selectivity for Cdk1, 2, and probably 5. Novel derivatives, such as R-roscovitine (CYC202) show great promise (Whittaker et al., 2004). 35

The efficacy of these inhibitors may not be the result of their CDK-inhibitory activity alone but also of their effect on transcription of CDK complex components, such as

Cyclin D1 as well as apoptotic regulators (Dai and Grant, 2003).

36

Figure 1.6 Schematic representation of the cell cycle and the different therapeutic

approaches undertaken. Several pharmacologic inhibitors have been generated to target the cyclins/CDKs at different phases of the cell cycle and several of them are in preclinical trials. Most of the inhibitors prevent the activity of more than one Cyclin/CDK complex. The effect of Cyclin E can be blocked by the expression of a dominant negative

(dn) Cdk2 which may constitute another therapeutic approach to target Cyclin E.

37

38

A novel class of therapeutics comprises inhibitors of the proteasome (bortezomib) which have been shown for example to cause the accumulation of Cyclin A and B, G2/M arrest and induce apoptosis in lung cancer cell lines (Bunn, 2004). Another new emerging compound with antitumoral activity is a fusion protein made up of a Cyclin A/Cdk2 binding peptide and an F-box protein, which targets the complex to ubiquitination and proteasomal degradation. This leads to a decrease of Cyclin A and Cdk2 levels followed by apoptosis both in vivo and in vitro with no significant toxicity to normal cells.

Apparently, the down-regulation of Cyclin A is essential for the cytotoxic activity.

However, decreasing both Cyclin A and Cdk2 expression causes the highest cytotoxic effect (Chen et al., 2004). Previous studies have shown that such Cyclin A/Cdk2-binding peptides induced apoptosis by further deregulating E2F expression (Chen et al., 1999).

Thus, an S-phase checkpoint seems to be activated and this involves E2F whose DNA binding function is in turn negatively regulated by Cyclin A (Krek et al., 1995; Pagano et al., 1992; Pruschy et al., 1999). A different type of Cdk2-Cyclin A antagonists (which do not block the ATP site) bearing the RXL peptidic motif were found to selectively induce apoptosis in cells in which pRb and Cyclin D were deregulated (Mendoza et al., 2003).

Apoptosis

Apoptosis is a universal genetic program of cell death in higher eukaryotes that is

an essential component of cellular development and differentiation, as well as to the 39

responses to various types of stress (Danial and Korsmeyer, 2004). Programmed cell death was shown to be critical for morphogenesis (Zuzarte-Luis and Hurle, 2005), differentiation (Hutchins and Barger, 1998), maintenance of homeostasis (Vaux and

Strasser, 1996), as well as regulation of the immune system (Rathmell and Thompson,

2002). Loss of control of apoptosis may lead to a series of pathological effects such as cancer, neurodegenerative and autoimmune disorders, viral infections, and ischemic diseases (Fadeel and Orrenius, 2005).

Apoptosis is defined by the typical morphology of the dying cell (Wyllie et al.,

1980), specifically by the chromatin condensation which is often accompanied by nucleus fragmentation. The cell membrane blebs and appears budding, resulting in membrane- enclosed structures called apoptotic bodies (Arends and Wyllie, 1991). These apoptotic bodies are then rapidly recognized and cleared by phagocytic cells without mounting an inflammatory response. On the other hand, necrosis results in rupture of the plasma membrane, followed by the spilling of the inner cellular contents, resulting in a strong inflammatory response (Leist and Jaattela, 2001).

Many apoptotic biochemical markers have been identified and are currently used to monitor it in experimental settings: DNA fragmentation by TUNEL assay (terminal deoxynucleotidyl transferase-mediated dUTP-digoxigenin nick end labelling) or subdiploid DNA content assays, activation of caspases by western blotting or immunofluorescence, cell surface externalization of phosphatidylserine by binding of

Annexin V (Hengartner, 2000). 40

Apoptosis can be triggered by a variety of factors: DNA damage induced by

ultraviolet light, ionizing radiation or chemotherapeutic drugs, elevated concentration of

oxidants within the cell or cell surface stimuli such as death activators, and growth factor

withdrawal such as serum starvation. Genotoxic agents, such as ionizing radiation, induce

apoptosis in many cell types, most potently in those of hematopoietic origin. Apoptosis is

frequently associated with proliferating cells, implying the existence of molecules in late

G1 and S whose activities facilitate execution of the apoptotic process.

Apoptosis is mediated through two major pathways, the death receptor pathway

and the mitochondrial pathway (Hengartner, 2000). Many proteins regulate or actively take part in the execution of the apoptotic process, but the main four types of molecules that are involved in this process are the caspases, the members of the Bcl-2 family, members of the tumor necrosis factor family and its receptors, and adaptor proteins.

Apoptosis is orchestrated by the function of a set of proteases termed caspases (cysteine aspartate proteases) which are critical regulators of the execution phase of apoptosis, triggered by many factors, including genotoxic agents (e.g. γ-irradiation or treatment with anti-cancer agents) (Chen et al., 2000b; Gong and Almasan, 2000; Gong et al., 1999).

These enzymes cleave specific substrates causing the characteristic cellular morphology.

TRAIL, APO2L

TRAIL (TNF-Related Apoptosis-Inducing Ligand) is a member of the Tumor

Necrosis Factor (TNF) family of death ligands. It is a transmembrane protein which can 41

be cleaved to generate a soluble ligand. There are two types of TRAIL receptors: TRAIL-

R1 and TRAIL-R2 which are agonistic receptors and TRAIL-R3 and TRAIL-R4 which

compete with the first two for binding of the ligands. Binding of TRAIL to its agonistic

receptors results in receptor trimerization, recruitment of adaptor molecules (FADD), and

Caspase-8 to their cytosolic tails to form the Death-Inducing Signaling Complex (DISC).

Adaptor proteins

The adaptor proteins are molecules that relay the apoptotic signal from the

transmembrane TRAIL receptors to the cell death regulators and effectors, such as

caspases. One of these is the Fas-Associated Death Domain (FADD or MORT-1). Upon

engagement of the death receptors by their specific ligands or agonistic antibodies,

FADD is recruited to the cytosolic tail of these receptors by homotypic interactions with

their Death Domains (DD) (Boldin et al., 1995; Kischkel et al., 1995; Micheau and

Tschopp, 2003). In turn, FADD, recruits pro-caspase-8 by interacting with its Death

Effector Domain (DED). The complex comprised of the TRAIL-R, FADD and pro-

caspase-8 is known as the DISC.

Apaf-1 is another adaptor protein which acts as a scaffold for the formation of the

apoptosome complex. Its caspase-recruitment (CARD) domain acts as a docking station

for pro-caspase-9. With the contribution of cytochrome c and dATP, Apaf-1 oligomerizes

to form the apoptosome complex (Cain et al., 2002; Rodriguez and Lazebnik, 1999; Zou

et al., 1997). This facilitates the activation of Caspase-9 which is crucial for the

subsequent activation of the executioner Caspase-3. 42

Cytochrome c

Once the cells are committed to cell death, apoptogenic factors, the best known of which is cytochrome c, are released from mitochondria to initiate the caspase cascade

(Chen et al., 2000b). Cytochrome c acts as a cofactor to stimulate the association of

Apaf-1 with Caspase-9 (Li et al., 1997), which then initiates activation of the caspase cascade.

Bcl-2 family members

The Bcl-2 family members are the main regulators of the mitochondrial death pathway, The first discovered member of this family, Bcl-2, was identified in human B cell lymphoma while studying the chromosomal translocation t(14;18) (Tsujimoto et al.,

1984). Many Bcl-2 family members have been uncovered to date and their main role is to regulate the release of cytochrome c from the mitochondria (Kaufmann and Hengartner,

2001). Based on their structure, they may either manifest anti-apoptotic properties such as

Bcl-2, Bcl-xL, Mcl-1, Bok or promote apoptosis such as Bax, Bcl-xS, Bak, Bad, Bim,

Puma, Noxa. These proteins have the ability to homo- or hetero-dimerize and the balance in the ratio between the pro- versus anti-apoptotic determines the susceptibility of the cell to the apoptotic signal (Oltvai et al., 1993).

The cell death antagonist proteins are characterized by the presence of all Bcl-2 homology domains (BH1-BH4). As most of these have a hydrophobic transmembrane domain, they can localize to different organelles, in particular the mitochondria (Hsu et al., 1997; Janiak et al., 1994; Wilson-Annan et al., 2003). These proteins block cell death 43

by preventing the disruption of the mitochondrial membrane. On the other hand, the

death promoting proteins have all the BH domains except for the BH4 domain. Following

the apoptotic signal, Bax which is found in the cytosol, translocates to the outer mitochondrial membrane (Griffiths et al., 1999; Hsu and Youle, 1998; Wolter et al.,

1997) where it oligomerizes to form a pore-like structure, promoting the release of

cytochrome c (Antonsson et al., 2000; Wei et al., 2000). This translocation can be

promoted by another type of Bcl-2 members, those that have only the BH3 domain, such

as tBid [resulted from cleavage of Bid by caspase-8 (Desagher et al., 1999)], Bim and

Bmf [microtubule-associated proteins (Puthalakath et al., 1999; Puthalakath et al., 2001)],

Noxa and Puma [induced by p53 (Nakano and Vousden, 2001; Oda et al., 2000)]. These

hetero-dimerize with the pro-apoptotic, Bax and Bak to induce their activation. This can

be prevented by Bcl-2 or Bcl-xL by sequestering the BH-3 only proteins or by directly

interfering with channel formation, thus inhibiting the release of mitochondrial

apoptogenic factors, such as cytochrome c, AIF, Smac/Diablo and HtrA/Omi.

Caspases

Caspases are the most important proteins in the apoptotic pathway. They are

members of a cysteine protease family that can hydrolyze peptidic bonds after an aspartic

acid (Asp) residue. Fourteen members of this family have been identified to date with

eleven of them being present in humans (Earnshaw et al., 1999). Caspases are involved in

apoptosis signaling (caspases 2, 3, 6, 7, 8, 9, and 10) as well as cytokine processing

(Earnshaw et al., 1999; Strasser et al., 2000). They are synthesized as inactive zymogens 44

containing a prodomain, a large, and a small unit. They are activated by proteolytic cleavage between the large and the small subunits followed by cleavage between the large subunit and the prodomain. Upon maturation, caspases form a tetramer of two small and two large subunits. Caspases are able to activate each other resulting in an amplification of activity through a protease cascade (Thornberry and Lazebnik, 1998).

The caspases involved in apoptosis fall within two categories: the initiators

(caspase 2, 8, 9, and 10) and the effectors (caspase 3, 6, and 7). The initiator caspases have long prodomains containing protein-protein interaction domains such as DED or

CARD while the effector caspases have short domains. Caspase-8 and -10 are the initiator caspases in the death receptor pathway (Denault and Salvesen, 2002). Binding of

Fas or TRAIL to their receptors results in receptor aggregation and recruitment of FADD and Caspase-8 through their DED domains to form the DISC (Muzio et al., 1996; Peter and Krammer, 2003). Once recruited, Caspase-8 is activated and initiates apoptosis by direct cleavage of effector caspases (Alnemri et al., 1996). Caspase-8 may also be cleaved and activated by Caspase-6. On the other hand, Caspase-9 is activated following its recruitment to Apaf-1 through its CARD domain. The effector caspases -3, -6, and -7 exist in the cytosol as inactive pro-caspase dimers and are activated through cleavage by the initiator caspases to generate the fully active enzymes (Boatright et al., 2003). They target various cytosolic or nuclear substrates whose degradation leads to the characteristic apoptotic cell morphology. Among these, there are proteins crucial for the maintenance of cellular cytoskeleton, DNA repair, transcription and translation, signal transduction, and cell cycle control (Hengartner, 2000). 45

Type / pathway Caspase-3 substrates

Kinases PKCδ, MEKK-1, FAK, PAK-2, PITSLRE

Transcription / SREBPs, hnRNPs C1/C2, U1-70 kD, smRNP Translation

DNA repair / DNA-PKcs, PITSLRE, pRb, D4-GDP-DI, NuMa, Mdm-2, PARP Cell Cycle

Structural lamin A,B, G-actin, fodrin, Gas2, presenilins, gelsolin, keratin1β

Table 1. Critical protein substrates of Caspase-3

Other apoptosis-regulatory proteins

The inhibitor of apoptosis proteins (IAPs) play an important regulatory role in the

apoptotic signaling pathway by tightly controlling the activation of caspases. c-IAP1, c-

IAP2, and XIAP interact and directly inhibit Caspase-3, -7, and -9 through their BIR

domains (Salvesen and Duckett, 2002; Takahashi et al., 1998).

The activation of caspase-8 in the DISC may be inhibited by proteins belonging to the FLICE-inhibitory protein (FLIP) family. There are several viral FLIPs named v-

FLIPs while the human homolog is termed c-FLIP. This may be present in two splice variants, the short c-FLIPs and the long c-FLIPL. These proteins are characterized by the presence of two DED domains. They act by preventing the activation of Caspase-8 and -

10 (Donepudi et al., 2003; Krueger et al., 2001). 46

Another protein with an important role in the execution of apoptosis is the

Caspase-Activated DNAase (CAD). Its function is to degrade the chromosomal DNA.

CAD is kept inactive bound to ICAD (inhibitor of CAD) (Enari et al., 1998; Nagata et al., 2003). When ICAD is degraded by caspase-3, CAD translocates to the nucleus and cleaves the DNA in small fragments which is a hallmark of apoptosis.

Apoptosis Inducing Factor (AIF) is a mitochondrial protein that bears both mitochondrial and nuclear localization sequences. During apoptosis, AIF is released from the mitochondria and translocates to the nucleus where it is responsible for caspase- independent induction of condensation of chromatin and DNA fragmentation (Cande et al., 2002; Susin et al., 1999).

Apoptotic pathways

Apoptosis may be initiated and executed through two well described pathways: the death receptor pathway (the extrinsic pathway) or the mitochondrial pathway (the intrinsic pathway). The death receptor pathway is initiated when ligands belonging to the

TNF family such as FasL (Apo1L), TNF-α, and TRAIL (Apo2L) engage their receptors

(Fas or CD95, DR1, DR2) on the plasma membrane. This interaction leads to the recruitment to their cytosolic tails of the adaptor protein FADD and pro-caspase-8 or -10 to form the DISC. The formation of this complex leads to the activation of Caspase-8

(Kischkel et al., 1995; Kischkel et al., 2000; Sprick et al., 2000). In turn, activated

Caspase-8 or -10 cleaves and activates the effector Caspases -3, -6, and -7 (Chen et al.,

2001) resulting in cleavage of cellular substrates (Figure 1.7). 47

Alternatively, Caspase-8 may cleave the BH3-only protein Bid generated tBid

(truncated Bid) which activates Bax, thus initiating the mitochondrial apoptotic pathway

(Wei et al., 2000). This pathway may also be activated by virus infection or DNA damage. In this pathway, the pro-apoptotic protein, Bax, translocates from the cytosol to the outer mitochondrial membrane (Nomura et al., 1999). Here, it oligomerizes following a change in conformation, and forms pores in the membrane through which cytochrome c is being released. Together with it, many other apoptosis regulatory factors such as

Smac/DIABLO (Du et al., 2000; Srinivasula et al., 2001; Verhagen et al., 2000),

HtrA/Omi (Hegde et al., 2002; Martins et al., 2002), ICAD, AIF (Joza et al., 2001), and endonuclease G (Li et al., 2001b) are being released into the cytoplasm. Cytochrome c and pro-caspase-9 are being complexes by Apaf-1 in presence of dATP to form a large complex named the apoptosome. This leads to the activation of caspase-9 (Strasser et al.,

2000), which in turn cleaves and activates the effector caspase-3.

48

Figure 1.7 Schematic representation of the extrinsic and intrinsic apoptotic

pathways. The extrinsic apoptotic pathway consists of Apo1 and Apo2 ligands interacting with their receptors on the cell membrane leading to the formation of DISC comprised of the cyotosolic part of the receptor, FADD, and pro-caspase-8. This causes the activation of caspase-8 which cleaves activating caspase-3. The intrinsic apoptotic pathway consists of cleavage of Bid by caspase-8 generating tBid which activates Bax.

Activated Bax translocates to the mitochondria where forms pores through which cytochrome c is being released into the cytosol. Cytochrome c complexes with Apaf-1 and pro-caspase-9 forming the apoptosome. This leads to activation of caspase-9 which, in turn, cleaves and activates caspase-3. Caspase-3 cleaves many cellular protein

substrates including ICAD thus releasing the nuclease CAD. In the nucleus, p53 up-

regulates the transcription of Bax, Puma and Noxa. Together with cytochrome c, other

pro-apoptotic factors are released from the mitochondria, such as, AIF and Smac/Diablo.

49

50

Cells that are able to undergo apoptosis through the DR pathway are termed type I

cells, whereas cells in which caspase-8 cleaves Bid, which induces apoptosis through the

mitochondrial pathway are called type II cells (Scaffidi et al., 1998).

Apoptosis represents the most severe cellular response to genotoxic stress, which is mounted when the cellular damage, primarily to the DNA, is beyond the capacity of the DNA repair proteins. Experimental evidence suggests that the cytotoxic effects of radiation and many forms of chemotherapy are mediated through a final common pathway that involves the activation of apoptosis. Radiation leads to cell death mainly through apoptosis in numerous normal tissues. Lymphoid cells, immature hematopoietic cells, and epithelial cells of the small intestine are among the most radiosensitive in the

body. Many cancer therapies are designed to target the apoptotic pathways within a cell, and the failure of cytotoxic therapies in many cancer cells have been attributed to apoptosis impairment. Two key early cellular events associated with radiation-injured apoptosis are the exposure of the phosphatidylserine on the plasma membrane and activation of caspases. Once cells are committed to cell death, apoptogenic factors, the best known of which is cytochrome c, are released form mitochondria to initiate a caspase cascade. Caspase activation is a critical regulator of the execution phase of apoptosis induced by many factors, including ionizing radiation. Upon activation through proteolytic cleavage, caspase may further cleave other caspases or cellular target proteins that include Bcl-2, Cyclin E, and poly (ADP-ribose) polymerase-1 (PARP-1). While other forms of cell death have been recently described (e.g. autophagy, necrosis with 51

different variations), it is believed that the apoptotic response is the predominant

mechanism of cell death.

DNA damage and repair mechanisms

Cellular DNA damage can be responsible for mutagenesis and the development of

cancer (van Gent et al., 2001). The human DNA may be subjected to several thousand to a million damaging events per day, generated by either external or internal metabolic processes. Changes in the genome can lead to errors in transcription and subsequently translation of the proteins necessary for cellular function. Certain DNA mutations can also be carried over into daughter cells if they are not repaired prior to mitosis.

When cells lose their ability to effectively repair damaged DNA, they can have

three possible fates. One possibility is to undergo senescence and become dormant. It has been reported that senescence could occur in cancer cells in vivo as well as in vitro, by stopping mitosis and preventing the cell from evolving further (Braig et al., 2005; Chen et al., 2005b; Collado et al., 2005; Michaloglou et al., 2005). Another outcome is cell death.

Sufficient DNA damage may trigger an apoptotic signaling cascade, forcing the cell into programmed cell death (O'Driscoll and Jeggo, 2006). Finally, the cells may undergo malignant transformation, become immortal, and divide uncontrollably (Mills et al.,

2003). To be able to cope with the different types of DNA damage that may occur, cells have developed several repair mechanisms such as mismatch (MMR), base excision 52

(BER), and nucleotide excision repair (NER). Double-strand breaks are resolved by the non-homologous end joining (NHEJ) (Lieber et al., 2003) or homologous recombination

(HR) (Haber, 1995) pathways. Cells may choose to proceed into apoptosis or senescence if overwhelming damage has taken place instead of expending energy to repair the DNA damage. The rate at which a cell is able to perform DNA repair depends on the cell type and its age.

For many years, it was believed that the main cause of DNA mutations leading to cancer was DNA damage induced by exogenous sources. However, more recently it was suggested that malignancy could also result from DNA damage caused by endogenous sources (Jackson and Loeb, 2001). There are two types of DNA damaging agents, physical and chemical. Physical mutagens are mainly sources of radiation, including UV

(200-300 nm) and ionizing radiation (X-rays). UV radiation is responsible for the production of covalent bonds that crosslink adjacent pyrimidine (cytosine and thymine) bases in DNA. On the other hand, IR causes DNA damage by generating free radicals within the cell that create reactive oxygen species (ROS). These, if not neutralized, may cause single-strand and double-strand breaks in the double helix. Chemical agents may alter the DNA structure by covalently attaching alkyl groups to DNA bases. Others are found as chemical inert precursors, which have to be converted metabolically to highly reactive carcinogens. These can further react with the DNA forming DNA adducts. For example, benzopyrene has to be activated to benzopyrenediol epoxide by the cellular oxidases to be able to form covalent DNA adducts. 53

DNA damage may also result from different other endogenous biochemical processes (De Bont and van Larebeke, 2004). Depurination (loss of adenine or guanine from the deoxyribosyl phosphate chain) is the most common event (Lindahl and Nyberg,

1972). Depyrimidation (loss of thymine or cytosine) also takes place but it is a lot less common than depurination. Another type of damage is the deamination of adenine, guanine, and cytosine rings to form the unnatural bases hypoxanthine, xanthine, and uracil. If the DNA repair enzymes do not recognize and remove these bases, an uncorrected uracil may be misread as a thymine during DNA replication and result in a C to T point mutation.

Another type of DNA modification, which could lead to misreading of DNA is

DNA methylation. The molecule responsible for transferring the methyl group onto DNA is S-adenosylmethionine (SAM). SAM is an intracellular metabolic intermediate that contains a highly reactive methyl group. The addition of a methyl group results in the formation of a DNA CpG sequence as this process occurs at the 5-position of the cytosine ring of a cytidine base that is 5’ to a guanosine base. The 5-methyl-cytosine product is very prone to spontaneous deamination, which represents a significant source of mutation error. Loss of the amine group results in a thymine base. Thus, the whole process leads to the creation of a C to T point mutation.

Reactive oxygen species (ROS) resulting from cellular metabolic processes can modify both purine and pyrimidine bases by oxidation. The most common change is guanine oxidized to 8-oxo-7,8-dihydroguanine, which results in the nucleotide 8-oxo- deoxyguanosine (8-oxo-dG). However, 8-oxo-dG instead of pairing with deoxycytidine, 54

can base-pair with deoxyadenosine. Enzymes belonging to the mismatch repair pathway

are responsible for detection and correction of this error. However, if the error is not

repaired, the DNA subsequently replicated will contain a C to A point mutation. ROS

may also cause depurination, depyrimidation and single-strand or double-strand breaks in

the DNA.

The most deleterious DNA damage consists of the single strand (SSB) and double

strand breaks (DSB) in the DNA (Khanna and Jackson, 2001). SSBs may result from

damage to the deoxyribose moiety of the DNA chain. SSBs may also be caused by the

base excision repair pathway after removal of the deoxyribose phosphate by AP-1

endonuclease. DSBs occur most often during the S phase, when the DNA may be more

susceptible to breakage as it is unwinding to serve as a template for replication (Brem and

Hall, 2005). Another source of DSBs is the V(D)J recombination process (Raghavan et

al., 2001). The extraordinary variability of the immunoglobulins and the T-cell receptors

is achieved through recombination of the V, D, and J gene segments to create mature

exons encoding the final protein. The process of recombination is mediated by the RAG1,

RAG2 (recombination activating genes 1 and 2) endonucleases and HMG1 (high

mobility group 1). RAG1 and 2 bind to specific signal sequences flanking the coding

regions and create a double-strand cleavage (Fugmann et al., 2000; Gellert, 2002). The

coding sequences and the two signal ends will be joined using the NHEJ pathways

(Lieber, 1999; Lieber et al., 2003).

The danger posed by the presence of DNA DSBs caused by DNA-damaging agents, such as ionizing radiation or chemotherapeutic drugs arises from their ability to 55

cause genomic instability through loss of heterozygosity and chromosomal translocations

(van Gent et al., 2001). These will eventually lead to cancer or apoptosis. As few as one or two unrepaired breaks would be sufficient to trigger the death of a cell. Throughout the

mammalian cell cycle, it is believed that DSBs are being repaired by HR during late S

and G2 phases while NHEJ is responsible for the repair during the rest of the cell cycle

(Takata et al., 1998). While repair by HR is an error-free process (West, 2003), NHEJ can

be very imprecise and thus lead to potential gene alterations. During the repair performed by NHEJ, the broken DNA ends get resected or nucleotides are filled in to generate regions of microhomology. In general, DNA ends that do not share significant homology will be repaired and joined by the NHEJ following resection to generate sites of microhomology (Roth and Wilson, 1986). However, when the NHEJ machinery is not entirely functional due to genetic mutation or absence of one the proteins involved in it, other pathways or enzymes may be employed for the repair of the lesion (Ferguson and

Alt, 2001).

Homologous recombination

The most common type of HR employed in repair of DNA DSBs is gene

conversion. This process takes advantage of a donor sequence with homology to both

sides of the DSB (the sister chromatid) which serves as template for the fill in process

(Elliott et al., 1998; Taghian and Nickoloff, 1997). The information is faithfully

incorporated resulting in an error-free repair process which is very important for the

maintenance of genomic integrity (Haber, 1995). The DSBs repaired by this mechanism 56

are usually caused in the S phase during attempts of replicating the DNA across a single-

strand break or an unrepaired lesion which results in the collapse of the replication fork.

Even though there is evidence of preference of HR or NHEJ repair for certain

types of DNA DSBs [V(D)J recombination is performed by NHEJ while DNA breaks

caused during by Spo1 require HR (Keeney, 2001)] and despite a cell cycle

phase preference, the two pathways are still coupled for DSB repair (Richardson et al.,

1998). This indicates that the choice regarding which pathway will be used for a particular DNA repair pathway, is influenced by many other factors. Studies in Ku70-/- cells have shown increased HR activity which suggests that upon binding of Ku70 to the ends of a DSB it prevents the access of HR repair machinery (Pierce et al., 2001). It appears that NHEJ and HR may compete for the repair of the same DNA DSB.

Ku70/Ku80 heterodimer

The Ku70/Ku80 heterodimer is a critical component in the NHEJ pathway (Gullo et al., 2006; Lees-Miller and Meek, 2003). Other important roles of Ku70 are in V(D)J recombination of the antigen receptor genes (Gellert, 2002), telomere maintenance, and in the transcriptional activation or repression of several genes (Shi et al., 2007).

Furthermore, both Ku70 and Ku80 are autoantigens that have been associated with connective tissues diseases such as systemic lupus erythematosus and scleroderma. Ku70 was found to be regulated through several mechanisms: heterodimerization with Ku80, intracellular localization (Koike and Koike, 2005), post-translational modifications such 57

as phosphorylation, acetylation, ubiquitination (Gama et al., 2006), and sumoylation

(Yurchenko et al., 2008).

In the nucleus, Ku70 is found in a very tight heterocomplex with Ku80. Ku70 is

very abundant and manifests high affinity for free DNA ends (Lieber et al., 2003). The

three-dimensional structure of the Ku70/Ku80 heterodimer has the shape of a doughnut

with a bulky bottom and a narrow bridge which can encircle and bind the DNA in a

sequence-independent manner (Walker et al., 2001). Even if they share minimal sequence

homology, the Ku70 and Ku80 subunits adopt a very similar three-dimensional structure, each of them contributing to the base, pillars, and bridge (Figure 1.8). One of the main roles in the NHEJ is to recruit DNA-PKcs. However, to accommodate the large 460-kDa

DNA-PKcs, Ku70/Ku80 must slide inward on the DNA in an ATP-dependent manner

(Turchi et al., 2000; Yoo and Dynan, 1999). The recruited DNA-PKcs then phosphorylates several substrates including Artemis (Poinsignon et al., 2004), the

Mre11/Rad50/Nbs1 complex (Uziel et al., 2003), and XRCC4/Ligase IV (Wang et al.,

2001b) which ultimately performs the repair of the DNA lesion.

Studies in Ku70-/- and Ku80-/- have shown that the absence of Ku leads to

dramatic radiation hypersensitivity, deficiency in DNA DSB repair, V(D)J

recombination, and impaired lymphocyte development. Apart form the radiosensitivity

imparted by the defects in the NHEJ process, these also lead to severe combined

immunodeficiency due to impaired V(D)J recombination (O'Driscoll et al., 2004; Rooney

et al., 2004; Schwarz et al., 2003). Even though most of its functions are nuclear, Ku70 58

also plays a cytoprotective role in the cytosol by interacting with and sequestering the pro-apoptotic protein Bax (Gomez et al., 2007; Yoshida et al., 2004).

59

Figure 1.8 Three-dimensional structure of the Ku70/Ku80 heterodimer. Ku70 (pink) and Ku80 (blue) interact very tightly through a very large interface forming a heterocomplex. At the carboxy terminus of Ku70 lies the Bax binding site (yellow).

(PDB structure 1JEQ(Walker et al., 2001) visualized with Cn3D)

60

DNA-PKcs

DNA-PKcs is a 460 kDa protein and a member of the phosphoinositol-3-OH

kinase (PI3K)-related kinase (PIKK) family which contains other proteins involved in

DNA repair such as ATM and ATR (Abraham, 2004). It is recruited by the Ku70/Ku80

heterodimer to the site of the DNA DSB. Together with Ku70 and Ku80 it promotes

synapsis of the broken DNA ends. The sheer size of DNA-PKcs warrants the belief that it

acts as a molecular scaffold for the recruitment for other NHEJ proteins with which it has

been shown to interact: XRCC4/Ligase IV (Calsou et al., 2003; Hsu et al., 2002; Leber et al., 1998), Artemis (Ma et al., 2005b; Ma et al., 2002), PNK (Koch et al., 2004), and the

X family polymerases (Ma et al., 2004) which are employed for making the free ends compatible for ligation. However, DNA-PKcs was also shown to be able to bind DNA directly, independently of Ku, which led to stimulation of its kinase activity (Yaneva et al., 1997). When activated, DNA-PKcs can phosphorylate itself as well as a series of substrates involved in DNA repair, such as Artemis and H1 histone, which is released from nucleosomes upon phosphorylation (Kysela et al., 2005). Studies have shown that

DNA-PKcs kinase activity is required for the Artemis endonuclease activity (Goodarzi et al., 2006). Furthermore, it is required for the conversion of 5’-OH ends to 5’-phosphate by polynucleotide kinase (Chappell et al., 2002). When assembled on double-stranded

DNA in vitro, the DNA-PK holoenzyme phosphorylates transcription factors and other proteins, including Sp1, Oct-1 (Schild-Poulter et al., 2007), c-fos, c-jun, p53, and the 34- kDa subunit of replication protein A (Anderson, 1993; Barnes et al., 1998). 61

DNA-PKcs was also found to regulate cell cycle arrest through down-regulation

of H2B and U2 RNA levels by phosphorylating the Oct-1 transcription factor (Schild-

Poulter et al., 2003). DNA-PKcs is also involved in the signaling pathway following

DNA damage by regulating p53-induced apoptosis (Wang et al., 2000; Woo et al., 2002).

Furthermore, it has been implicated in the regulation of NF-kB (Basu et al., 1998) and

H2AX (Burma et al., 2001) following IR. Moreover, DNA-PKcs was shown to undergo

proteolytic cleavage in cells undergoing apoptosis as well as during infection with the

poliovirus (Casciola-Rosen et al., 1995; Graham et al., 2004). This appeared to be

independent of caspase activity but directly dependent on picornaviral 2A protease.

Upon binding to DNA, DNA-PKcs undergoes autophosphorylation, which results

in loss of its kinase activity and disassembly from the DNA repair complex (Chan and

Lees-Miller, 1996). When DNA-PKcs cannot undergo autophosphorylation due to

mutation of specific amino-acid residues residing between position 2609 and 2647, cells

manifest marked radiosensitivity and V(D)J coding joints show little end processing. This is confirmed by several findings which demonstrate that efficient ligation by

XRCC4/Ligase IV requires previous DNA-PKcs autophosphorylation (Block et al., 2004;

Reddy et al., 2004; Weterings et al., 2003). Several reports made a link between the

NHEJ pathway and cellular redox stress. It was shown that cellular oxidative stress would

result in decreased DNA-PKcs activity (Boldogh et al., 2003). This impacts the

autophosphorylation capability of DNA-PKcs (Bacsi et al., 2005), which is required for

its activation and role in NHEJ (Chen et al., 2005a; Reddy et al., 2004). Interestingly,

cellular deficiency in DNA-PKcs autophosphorylation leads to an even higher 62

radiosensitivity than the absence of DNA-PKcs altogether. Apparently, this is due to the

fact that DNA-PKcs restricts the access of HR proteins to the DNA damage site (Convery et al., 2005). This suggests that the kinase disassembly from the DSB site would be as important for the completion of DNA repair as for initiating the repair process (Chan and

Lees-Miller, 1996; Douglas et al., 2001; Merkle et al., 2002). Furthermore, consistent with the belief that NHEJ is active mainly in the G0 and G1 phases of the cell cycle, while HR predominates in S and G2, several studies have shown that DNA-PKcs autophosphorylation occurs primarily in G0 and G1, thus limiting the involvement of HR when the sister chromatid is not present (Chen et al., 2005a).

Artemis

DNA DSBs caused by ionizing radiation gene have “dirty”, unligatable ends.

These aberrant structures, such as the 3’-phosphate or 3’-phosphoglycolate groups must be removed prior to the ligation event mediated by Ligase IV. This resection is performed by Artemis, a protein with endonuclease activity (Ma et al., 2005b). Its activity was shown to be stimulated by phosphorylation by DNA-PKcs (Goodarzi et al., 2006).

XRCC4

XRCC4 is another important protein in the NHEJ pathway. This protein was found in complex with Ligase IV (Grawunder et al., 1998; Modesti et al., 1999) as well as forming homodimers (Modesti et al., 1999). XRCC4 is constitutively phosphorylated.

However, following treatment with ionizing radiation is further phosphorylated by DNA- 63

PKcs (Matsumoto et al., 2000; Yu et al., 2003b). A recent study showed that XRCC4 is also subject of sumoylation, a process that regulates its nuclear localization (Yurchenko et al., 2006). Furthermore, XRCC4 was shown to be mono-ubiquitinated (Foster et al.,

2006), but the exact role of this process has not yet been defined. Even though not completely understood, it appears that it plays a regulatory role on the repair complex by stabilizing Ligase IV (Bryans et al., 1999) as well as by stimulating its activity

(Grawunder et al., 1997).

Ligase IV

Ligase IV is the essential enzyme that performs the last step in the NHEJ pathway. The ligation takes place in three steps involving an adenylate intermediary

(Tomkinson et al., 2006). Ligase IV and XRCC4 knock-out mice are embryonic lethal and this has been associated with massive neuronal cell death (Frank et al., 2000; Gao et al., 1998; Pan et al., 1994). Ligase IV function is specifically required for the NHEJ pathway of DSB repair and is completely dependent on Ku. In mice deficient in Ligase

IV or XRCC4, cells harboring Ku-bound DSBs eventually die, because these lesions cannot be repaired by HR. In contrast, in the absence of Ku, HR can partially substitute for the lost NHEJ function, resulting in the less severe phenotype observed in the Ku deficient mice (Adachi et al., 2001). Of the DNA repair complex proteins, Ku70, Ku80,

XRCC4, and Ligase IV are conserved throughout evolution in all eukaryotic species known, whereas DNA-PKcs and Artemis have not been found in lower eukaryotes such as yeast (Critchlow and Jackson, 1998). 64

XLF

A new factor with an important role in the NHEJ has recently been identified

(Ahnesorg et al., 2006; Buck et al., 2006). It was named Cernunnos or XLF (XRCC4-like factor) as it showed very high structural similarity with XRCC4, despite a low sequence homology. Several mutants of XLF have been shown to cause immunodeficiency with microcephaly (Buck et al., 2006). XLF was found to form homodimers (Ahnesorg et al.,

2006) which interact with XRCC4 through their globular head domains (Deshpande and

Wilson, 2007). Many reports showed a complex comprised of XLF, XRCC4 and Ligase

IV, but apart from the role of Ligase IV in the final step of repair, much needs to be done to ascertain the biochemical functions of these proteins. It appears that XLF stimulates the ability of XRCC4/Ligase IV to join incompatible ends (Gu et al., 2007; Tsai et al.,

2007). Its newly discovered crystal structure (Li et al., 2008) confirms that XLF forms homodimers through a coiled-coil region in a similar manner to that of XRRC4. This confirms the similarity in general structure with XRCC4 but points out important structural differences indicating distinct functions in the DNA repair process. It further casts doubt upon the interaction between XLF and Ligase IV and suggests that this might take place indirectly through XRCC4, hypothesizing that the stoichiometry of the

XRCC4/XLF/Ligase IV complex is 2:2:1 (Figure 1.9).

65

Figure 1.9 Schematic representation of the XLF/XRCC4/Ligase IV heterocomplex

66

Non-Homologous End Joining

As soon as DSBs are generated, the free DNA ends are bound by Ku70/Ku80

heterodimers (Lieber et al., 2003) (Figure 1.10). This interaction is facilitated by the

doughnut-like structure of the complex (Figure 1.8). The DNA is encircled by the

Ku70/Ku80 complex ensuring a tight binding. This structure functions as a scaffold for

the subsequent recruitment of other DNA repair proteins (Ma et al., 2005a; Ma et al.,

2004). DNA-PKcs is being recruited by the Ku heterocomplex and is activated upon

binding of the DNA end (West et al., 1998). This 460-kDa serine/threonine kinase

phosphorylates and recruits other important proteins for the repair of the lesion. One of

its substrates is the endonuclease Artemis. Another protein recruited at the DSB site is a polymerase, specifically µ or λ (Ma et al., 2004; Mahajan et al., 2002) via its BRCT domains. This may perform some of the fill-in synthesis (Wilson and Lieber, 1999). The ligation of the free DNA ends is performed by Ligase IV, which is recruited as part of the

XLF/XRCC4/Ligase IV heterocomplex (Chen et al., 2000a; Nick McElhinny et al.,

2000). XLF/XRCC4/Ligase IV was shown to be recruited directly by the Ku70/Ku80 heterodimer. However, this interaction is facilitated in the presence of DNA-PKcs

(Costantini et al., 2007). The exact physiological role of XLF in this complex still needs to be elucidated.

Other DNA double-strand break repair pathways

Interestingly, cells that harbor mutations in the NHEJ pathway even though they

show a marked reduction in the DNA repair ability, still rejoin the majority of their DSBs 67

albeit by a slow operating process (DiBiase et al., 2000; Nevaldine et al., 1997; Wang et al., 2001a). Additionally, this process is completely independent of HR (Wang et al.,

2001a). This acts as an alternative back-up NHEJ pathway to the classical one involving

Ku, DNA-PKcs, Artemis, XRCC4, and Ligase IV. The essential proteins taking part in this alternate pathway are PARP-1 and DNA Ligase III (Audebert et al., 2004; Wang et al., 2006), however much remains to be characterized in this pathway.

68

Figure 1.10 Schematic representation of the NHEJ pathway. DNA DSBs caused by

IR or chemotherapeutic agents are repaired preferentially by NHEJ. The first step in the repair pathway is the binding of the Ku70/Ku80 heterodimer to the free DNA ends. This is followed by the recruitment of the serine/threonine kinase DNA-PKcs. This, in turn, phosphorylates and recruits Artemis, an endonuclease, and µ or λ polymerases, whose

roles are to trim or fill in the DNA overhangs in order to make them compatible for

ligation. The final step in the pathway is the recruitment of the XLF/XRCC4/Ligase IV

heterocomplex. Ligase IV is responsible for the ligation of the DNA ends while XLF and

XRCC4 appear to have regulatory functions.

69

70

Generation of p18-Cyclin E and its role in modulating cellular response to genotoxic

stress

Another Cdk2-independent role of Cyclin E was identified in the regulation of

programmed cell death. We have previously shown that Cyclin E is regulated by genotoxic stress caused by ionizing radiation or other chemotherapeutic drugs and that

this plays a functional role in the apoptosis of hematopoietic cells (Mazumder et al.,

2000). We observed a surge in Cyclin E levels during genotoxic stress, which is followed

by caspase activation and membrane display of phosphatidylserine, all hallmarks of

apoptosis. We have further reported the cleavage of the full length 50 kDa Cyclin E to

generate an 18 kDa fragment which was identified as the carboxy terminus of the Cyclin

E (Mazumder et al., 2002). This fragment, termed p18-Cyclin E, lacks the Cdk2 binding

domain and therefore, fails to activate Cdk2 (Figure 1.11).

Overexpression of p18-Cyclin E leads to onset of apoptosis in a caspase-

dependent fashion. We identified caspase-3 as the responsible caspase for the cleavage of

Cyclin E and generation of p18-Cyclin E. Overexpression of the p50-Cyclin E as well as

of a cleavage resistant mutant of Cyclin E, which was generated by mutating the cleavage

site, did not result in the activation of apoptosis. However, overexpression of the cleavage

resistant mutant of Cyclin E not only prevented the induction of apoptosis, but also the

generation of p18-Cyclin E, thus acting in a dominant negative manner (Mazumder et al.,

2002). Taken together, these data suggest that in addition to the cell cycle regulatory

function, Cyclin E plays another important role in the mechanism of apoptosis. 71

Figure 1.11 Schematic representation of Cyclin E domains and cleavage sites. Cyclin

E was found to be cleaved by calpain resulting in low molecular weight (LMW)

hyperactive forms. On the other hand, Cyclin E was also found to be cleaved by elastase

resulting in a truncated form (81-363) whose three-dimensional structure has been solved.

Based on this structure, the interface between Cyclin E and Cdk2 was defined as amino-

acids 94-112, 182-256, and 340-344. Through analogy with the interaction between

Cyclin A and p27, it was suggested that 129-136 and 170-173 are the residues responsible for the binding and inhibition of Cyclin E by p27. The 129-215 sequence illustrated in blue represents the Cyclin box, a conserved domain among cyclins. p18-

Cyclin E is generated following a Caspase-3 mediated cleavage at the Asp275 residue and thus, lacks the Cdk2 binding domain failing to bind and activate it. Human Cyclin

E1, accession number AAM54043 (NM_057182), was used as a reference.

72

Later on, using a yeast-two-hybrid assay, we identified several potential p18-

Cyclin E-interacting proteins (Mazumder et al., 2007b). Among all candidates, Ku70 stood out through its roles in Non-Homologous End-Joining DNA repair (Lieber et al.,

2003) as well as its cytoprotective function in the cytosol (Cohen et al., 2004;

Subramanian et al., 2005). As a result of this interaction, we witnessed a release of Bax from its cytoprotective sequestration to Ku70 in the cytosol and its activation, which could be responsible for the observed onset of apoptosis following generation of p18-

Cyclin E (Mazumder et al., 2007a; Mazumder et al., 2007b).

In this dissertation I will discuss a novel, important role of p18-Cyclin E in the regulation of NHEJ DNA repair pathway as well as newly uncovered regulatory mechanisms of p18-Cyclin E turnover and their implications in clinical therapeutics.

CHAPTER II

p18-Cyclin E regulates Non-Homologous End Joining by preventing the recruitment

of the XLF/XRCC4/Ligase IV heterocomplex to the DNA repair complex

Abstract

Cyclin E/Cdk2 is a critical regulator of cell cycle progression from G1 to S phase in mammalian cells that is deregulated in many types of neoplasms. We found that genotoxic stress leads to a dramatic decrease in Cyclin E levels in hematopoietic tumor cells, coinciding with the timely appearance of its proteolytic fragment, p18-Cyclin E.

Overexpression of p18-Cyclin E in a variety of cell types induces apoptosis. We have identified Ku70, a critical component of non-homologous end joining (NHEJ) DNA repair as a new interacting partner of p18-Cyclin E. Cells stably expressing p18-Cyclin E at non-toxic levels are more sensitive to etoposide or ionizing radiation treatment. Neutral comet assays used to determine residual double-strand breaks (DSBs) indicative of an ineffective NHEJ, showed significant increase in both tail length and tail moment in the presence of p18-Cyclin E following irradiation. Moreover, a plasmid reactivation assay indicated that p18-Cyclin E reduced the colony formation ability known to be associated with NHEJ activity as compared to cell lysates containing wild-type Cyclin E. Gel electrophoretic analyses indicated an impairment of end-ligation dependent on the expression levels of p18-Cyclin E. DNA pull-down assays showed that the assembly of

73 74

Ku70/Ku80 and DNA-PKcs is not affected by the presence of p18-Cyclin E. However, the recruitment of XRCC4, Ligase IV, and the recently identified accessory factor XLF was greatly impaired. These data indicate a profound effect of p18-Cyclin E on cellular survival and on NHEJ that is dependent on its interaction with Ku70 and probably caused by interference with the recruitment of the XLF/XRCC4/Ligase IV heterocomplex to the sites of DSBs. Interaction of Ku70 with p18-Cyclin E and Bax may be unique in providing the molecular switch between cell cycle control, DNA repair, and apoptosis activation for the cellular decisions made following genotoxic stress.

Introduction

The integrity of the genome is a critical requirement for propagation of life.

Damage to cellular DNA is involved in mutagenesis and development of cancer. Among the different types of DNA damage encountered daily by a human cell, the DSBs are the most dangerous for the survival of the cell, making their repair a necessity (Hoeijmakers,

2001; Jackson, 2001; Shiloh, 2003; Valerie and Povirk, 2003). Double strand breaks are naturally produced as a consequence of ionizing radiation (IR), DNA replication over a nicked template or as a result of enzymatic cleavage such as by RAG1 and RAG2 endonucleases during V(D)J recombination. The emergence of DNA DSBs caused by IR or other chemotherapeutic drugs leads to activation of a signal transduction cascade resulting in cell cycle arrest, assembly of a DNA repair complex, and changes in gene transcription. Inability to efficiently repair these DSBs causes genomic instability as shown by the presence of chromosome fusions (Khanna and Jackson, 2001). DSBs are 75

potentially catastrophic lesions which, if not repaired, would lead to loss of genetic

information, genomic instability or cell death.

Depending on the cell cycle phase, this type of lesion may be repaired by

homologous recombination (HR) or NHEJ. The general belief is that HR is employed in

late S and G2 cell cycle phases when the sister chromatid is available and may be used as

a template while NHEJ is used during the rest of the cell cycle. For this reason, NHEJ is considered as the most commonly used repair mechanism for the repair of DSBs. HR requires extensive sequence homology so that one DNA strand could invade a homologous sequence and initiate the repair (Paques and Haber, 1999). On the other hand, NHEJ rejoins the broken DNA with little or no sequence homology but it requires processing of the ends prior to ligation, a process during which some nucleotides are

deleted or inserted (Jeggo, 1998). While HR takes place with high accuracy as one

chromatid serves as template, NHEJ is largely an imprecise DNA repair process. Mice

that are deficient in components of the NHEJ machinery show radio hypersensitivity,

immunodeficiency, and are prone to tumor formation (Ferguson and Alt, 2001; van Gent

et al., 2001).

The NHEJ machinery relies on the formation of a large DNA repair complex

encompassing the Ku70/Ku80 heterodimer, DNA-PKcs, and XLF/XRCC4/Ligase IV.

The absence of any of these proteins confers increased radiation hypersensitivity as well

as immune deficiency due to impaired V(D)J recombination (Weterings and van Gent,

2004). Ku70/Ku80 shows very high affinity for free DNA ends and binds them in order

to keep them in close proximity (DeFazio et al., 2002). Next, the Ku70/Ku80 heterodimer 76

recruits DNA-PKcs, a serine/threonine kinase which phosphorylates many substrates

including itself (Smith and Jackson, 1999). These substrates include the nuclease Artemis

(Ma et al., 2002), that is responsible for the resection of 5’ and 3’ overhangs and DNA polymerases µ and λ (Bebenek et al., 2003; Capp et al., 2006; Mahajan et al., 2002;

Zhang et al., 2001), which perform some of the fill-in synthesis. The final step of ligation is performed by Ligase IV, whose activity is regulated by two proteins with which it is found in complex: XRCC4 (Critchlow et al., 1997; Grawunder et al., 1997) and

XLF/Cernnunos (Ahnesorg et al., 2006; Buck et al., 2006). Given the important role of

NHEJ in DNA DSB repair and cell survival, inhibiting this pathway represents an attractive opportunity to increase cell sensitivity to IR or chemotherapy (He et al., 2007;

Li et al., 2003; Marangoni et al., 2000).

Cyclin-dependent kinase 2 (Cdk2) and its regulatory factor Cyclin E form the key regulatory complex of the G1 to S phase cell cycle progression. Cyclin E has been shown to be deregulated and overexpressed in several solid tumors, including breast, colon, and prostate carcinomas (Akli and Keyomarsi, 2003; Erlandsson et al., 2003; Keyomarsi et al., 1995; Keyomarsi et al., 1994; Keyomarsi et al., 2002; Moroy and Geisen, 2004;

Sherr, 1996). High levels of Cyclin E expression have been associated with the progression of different other tumors, such as leukemias and lymphomas (Erlanson and

Landberg, 2001; Erlanson et al., 1998).

We have previously shown that Cyclin E is critical for genotoxic stress-induced apoptosis of tumor cells of hematopoietic origin (Mazumder et al., 2000). An 18-kDa

Cyclin E fragment (p18-Cyclin E) is generated through caspase-mediated proteolytic 77

cleavage of Cyclin E (Mazumder et al., 2002) in hematopoietic cells undergoing

apoptosis. Recently, we have identified Ku70 as a specific p18-Cyclin E-interacting

protein which mediates p18-Cyclin E-induced apoptosis (Mazumder et al., 2007a;

Mazumder et al., 2007b). Apart from its role in the NHEJ, Ku70 also plays a

cytoprotective role by binding and sequestering Bax in the cytosol (Cohen et al., 2004;

Subramanian et al., 2005). Mapping the p18-Cyclin E interaction domain of Ku70 using a

series of deletion mutants of Ku70, revealed that their interface resides at the N-terminus

of Ku70, which is different from the Bax binding site located at the C-terminus.

Furthermore, we have shown that binding of p18-Cyclin E to Ku70 results in Bax release

leading to amplification of the apoptotic signal. However, the presence of p18-Cyclin

both in the cytosol and in the nucleus, taken together with the important role of Ku70 in

NHEJ DNA repair, led to the hypothesis that p18-Cyclin E may also be involved in

regulation of DNA repair during cell death induced by genotoxic stress.

In our study we observed that cells that stably express p18-Cyclin E at non-toxic levels are more sensitive to DNA damaging agents, such as ionizing radiation and etoposide. Furthermore, p18-Cyclin E impairs DNA repair in vitro as shown by the failure to re-ligate linear plasmid DNA. Moreover, a plasmid reactivation assay showed that the presence of p18-Cyclin E led to a dramatic reduction in colony formation. Tail length and tail moment of unrepaired DSBs determined by neutral comet assays, indicated that the DNA DSBs repair is less efficient in cells expressing p18-Cyclin E as compared to control cells. Mechanistically, DNA pull-down assays which recapitulate the

DNA repair complex assembly showed that the DNA binding of Ku70/Ku80 and the 78

subsequent DNA-PKcs association are not affected by p18-Cyclin E. Conversely, the recruitment of XLF/XRCC4/Ligase IV to the DNA repair complex is severely reduced in the presence of p18-Cyclin E.

Here we report a novel role of the C-terminal fragment of Cyclin E (p18-Cyclin

E) in regulating the NHEJ DNA repair pathway in cells undergoing genotoxic stress. This may be critical in hematopoietic tumor cells that are known to have high NHEJ activity that may be activated by the DSBs produced by apoptotic nucleases. Blocking NHEJ could prevent, for example translocations leading to further tumor progression in these cells. Thus, p18-Cyclin E upon being generated may on one hand trigger the release of

Bax from its complex with Ku70 in the cytosol and on the other hand prevent DNA repair by NHEJ in the nucleus. This way, p18-Cyclin E may play an important role in the cellular decision making process between cellular survival and apoptosis.

Materials and methods

Cell lines and treatments. HEK293T human kidney cells were maintained in Dulbecco modified Eagle medium (Gibco-BRL) containing 10% fetal bovine serum (Hyclone,

Logan, UT) with L-glutamine and 100 units/ml penicillin/streptomycin at 37°C and in an atmosphere containing 5% CO2. Cells ~75% confluent were either treated with etoposide

(VP16) or irinotecan (CPT-11) from Sigma (St. Louis, MO), or irradiated with 10 Gy

(137Cs source; fixed dose rate of 2.0 Gy/min). HA-tagged p18-Cyclin E stably expressing 79

293T cells were generated by infecting cells with a lentivirus carrying HA-p18-Cyclin E

and EGFP separated by an IRES2 sequence and under the control of the EF1α promoter.

Antibodies. The following primary antibodies were used: anti-cyclin E (HE-12), anti-

Ku70 (E-5), anti-Ku80 (B-1) from Santa Cruz Biotechnology (Santa Cruz, CA), anti-β- actin from Sigma, anti-HA (HA.11) from Covance (Berkeley, CA), anti-PARP-1 from

Cell Signaling (Beverly, MA), anti-DNA-PKcs (Ab-1, 18-2) from Lab Vision (Fremont,

CA), anti-XLF (A300-730A) from Bethyl Laboratories (Montgomery, TX), anti-XRCC4

(AHP387) and anti-Ligase IV (AHP554) from AbD Serotec (Raleigh, NC).

Immunoblot analyses. Following irradiation or treatment with VP16, cells were lysed at

4°C for 30 min in buffer A (20 mM HEPES pH 7.5, 150 mM NaCl, 1 mM EDTA, 1%

NP-40, containing 1 mM phenylmethylsulfonyl fluoride (PMSF), 1 µg/mL pepstatin A, aprotinin, and leupeptin. Proteins (10-75 µg/lane) were resolved by SDS-PAGE, followed

by transfer to 0.1 µm nitrocellulose membrane and blocking in 5% nonfat dry milk for 1 h

at room temperature. The blots were then incubated with the appropriate primary

antibody in phosphate-buffered saline with 0.1% Tween 20 (PBST) containing 5% nonfat

dry milk for 16 h at 4°C followed by horseradish peroxidase (HRP)-conjugated

secondary antibodies.

Caspase-3 activation assay. Treated cells were lysed in lysis buffer containing 1% NP-

40, 20 mM HEPES (pH 7.5), 4 mM EDTA and aprotinin (10 µg/ml), leupeptin (10 80

µg/ml), pepstatin (10 µg/ml), and PMSF (1 mM). Equal amounts of protein lysates were taken in a microtiter plate to 200 µL with reaction buffer containing 100 mM HEPES (pH

7.5), 20% v/v glycerol, 5 mM dithiothreitol (DTT), 0.5 mM EDTA and caspase-3 substrate (Ac-DEVD-pNA; Calbiochem EMD, Gibbstown, NJ) was added to 100 µM.

The samples were incubated at 37°C for 1-2 h and the enzyme-catalyzed release of p-NA was determined by measuring the absorbance spectrophotometrically at 405 nM.

Comet assay (single-cell gel electrophoresis). HEK-293T cells were transiently transfected with p18-Cyclin E-HA using Lipofectamine 2000 (Invitrogen, Carlsbad, CA) according to the manufacturer’s indications. 24 h post-transfection cells were irradiated with 10 Gy and collected after 16 h. To evaluate the degree of DNA damage, we used

CometAssay Silver (Trevigen, Gaithersburg, MD), which combines a neutral lysis in low point melting agarose with silver staining or SYBR Green for visualization of the DNA.

Lysis and electrophoresis were performed according to the manufacturer’s protocol.

Image analysis and quantification have been performed with the software package NIH

ImageJ. Tail Moment (TM) and Tail Length (TL) were used to quantify the DNA damage. TM = % of DNA in the tail x TL; where % of DNA in the tail = tail area (TA) x tail average intensity (TAI) x 100/(TA x TAI) + [head area (HA) x head area intensity

(HAI)].

DNA-agarose pull-down. Nuclear extracts of HEK293T stably expressing HA-tagged p18-Cyclin E were prepared by hypotonic swelling lysis in 20 mM Tris-HCl pH 8.0, and 81

1 mM DTT. After incubation on ice for 30 min, nuclei were collected by centrifugation at

8000xg for 5 min and extracted in 20 mM Tris-HCl, pH 8.0, 500 mM NaCl, 5 mM

o MgCl2, 10% glycerol, and 1 mM DTT for 30 min, gently rocking at 4 C. All buffers contained 1x HALT phosphatase inhibitor cocktail (Pierce, Rockford, IL), 1µg/mL aprotinin, leupeptin, pepstatin, and 1 mM phenylmethylsulfonyl fluoride. Extracts were clarified by centrifugation at 16.000xg for 10 min and diluted (1:3) with 20 mM Tris-HCl pH 8.0, 5 mM MgCl2, 10% glycerol, 1 mM DTT. A slurry (50 µL) of denatured calf

thymus DNA-agarose beads (Amersham, Piscataway, NJ), were washed three times, added to the extracts and incubated at 4oC with rotation overnight. Beads were washed 3 times with 1 mL of buffer and the proteins eluted with 500 mM KCl and separated by

SDS-PAGE.

DNA end-ligation and plasmid reactivation assay. 1 µg of EcoR1-digested pUC18

DNA (DSB) was incubated with nuclear extracts of p18-Cyclin E and Cyclin E transfected 293T cells or control (GFP-expressing) and p18-Cyclin E stably expressing

293T cells, in reaction buffer X (40 mM Tris-HCl (pH 7.5), 10 mM MgCl2, 50 µM dNTPs, 2 mM ATP, 1 mM DTT and 100 µg/ml BSA). The end ligation mixture was incubated at 37oC for 1 h. The product was separated and analyzed by electrophoresis on

0.6% agarose gels together with monomer pUC18, used as a negative control and monomer pUC18 ligated with T4 DNA ligase, used as a positive control. For the plasmid reactivation assay, EcoRI-linearized pUC18 DNA was incubated with nuclear extract of transfected 293T cells in reaction buffer X at 37oC for 1 h. DNA was purified, 82

transformed in DH5α E.coli and transformed cells were plated on LB with Amp, X-gal, and IPTG.

Results

Non-toxic levels of p18-Cyclin E sensitize cells to VP-16 and IR treatment

Our previous experiments have shown that in hematopoietic cells subjected to

genotoxic stress, Cyclin E undergoes a caspase-3-mediated cleavage to generate an 18-

kDa carboxy-terminal fragment termed p18-Cyclin E (Mazumder et al., 2002). We have

also demonstrated that the pro-apoptotic effect of this protein is taking place in the

cytosol and it is mediated by its interaction with Ku70 through release and activation of

Bax (Mazumder et al., 2007b). To study the effect p18-Cyclin E may have on NHEJ in

the nucleus in the absence of its action in the cytosol, we have generated an HA-p18-

Cyclin E stably expressing 293T cell line using a lentiviral approach (Figure 2.1A).

These cells express p18-Cyclin E at moderate non-toxic levels and are viable and

thriving. Furthermore, as Cyclin E cleavage does not take place, therefore these cells are

ideally suited to study the regulation and role of p18-Cyclin E.

To determine the effect of p18-Cyclin E on cell viability under these experimental

conditions, we treated control cells (GFP-expressing) or p18-Cyclin E stably expressing

293T cells with etoposide at increasing concentrations. Increased PARP-1 cleavage in

p18-Cyclin E-expressing cells suggested that p18-Cyclin E may be a mediator of

etoposide-induced cell death (Figure 2.1B). Moreover, E cells stably expressing p18- 83

Cyclin were more sensitive than control (GFP-expressing only) cells to irradiation as shown by a caspase-3 activation assay. Taken together, these data suggest that p18-

Cyclin E is able to sensitize cells to DNA damaging agents and we hypothesize that this

effect could result from its interaction with Ku70 in the nucleus and subsequent

interference with NHEJ repair.

84

Figure 2.1 Non-toxic levels of p18-Cyclin E sensitize cells to VP-16 and IR treatment. A) p18-Cyclin E lentivirus expression vector. B) HEK293T cells stably expressing p18-Cyclin E or EGFP were treated with increasing doses of etoposide (VP-

16) and cell death was determined by PARP-1 cleavage [full length (fl), cleaved (cl)] .

Levels of PARP-1, p18-Cyclin E, and β-actin, used as a loading control were determined by immunoblotting. C) 293T cells stably expressing p18-Cyclin E or EGFP were irradiated with 10 Gy ionizing radiation and cell death was assessed by activation of caspase-3. (Data are representative of three independent experiments)

85

A

B

C

86

p18-Cyclin E inhibits in vitro DNA ligation and plasmid reactivation

To determine whether p18-Cyclin E is capable of interfering with DNA repair we tested this hypothesis in an in vitro DNA end-ligation study. In the presence of p18-

Cyclin E the plasmid DNA was observed only in its monomeric form. In contrast, in the presence of Cyclin E or T4 DNA ligase used as positive control, the plasmid DNA was efficiently re-ligated, as shown by the higher molecular weight concatemeric forms

(Figure 2.2A left and middle panel). The expression of p18-Cyclin E and Cyclin E was

confirmed by immunoblotting using anti-HA antibodies (Figure 2.2A right panel). This

suggests that p18-Cyclin E was able to inhibit the plasmid re-ligation as observed by the

absence of the concatemeric DNA forms. Furthermore, when we examined the ability of

the re-ligated plasmid to form colonies following transformation in E. coli, we observed a

dramatic reduction in the number of colonies resulted in the presence of p18-Cyclin E to

~10% as compared to ~60% for Cyclin E or T4 DNA ligase considered to be 100%

(Figure 2.2B). These data suggest that p18-Cyclin E is able to interfere and prevent the

DNA repair in an in vitro assay.

87

Figure 2.2 p18-Cyclin E inhibits in vitro DNA ligation and plasmid reactivation. A)

Nuclear cellular extracts from control (GFP-expressing), 293T stably expressing p18-

Cyclin E (left panel) or transfected with increasing DNA amounts of p18-Cyclin E and

Cyclin E (middle panel) were incubated with a DNA plasmid previously linearized with

the EcoRI restriction enzyme. The reaction was incubated at 37oC to allow the plasmid to be re-ligated. T4 DNA ligase was used as a positive control. The DNA ligation reaction was separated by gel electrophoresis (left and middle panel). Expression of the transfected constructs was determined by immunoblotting with anti-HA antibody (right panel). The second and third lanes of the left panel represent identical reactions. B) The

DNA ligation reaction mix was transformed in E. coli that were then plated on agar

containing X-Gal and IPTG. The colonies were counted and the number was expressed as

percentage of the T4 DNA ligase treated colonies, that was considered to be 100%.

88

89

p18-Cyclin E prevents efficient repair of the DNA damage induced by IR or CPT-11

To determine whether p18-Cyclin E affects DNA repair in vivo, we examined the

ability of human cells to repair the DNA damage induced by either IR or irinotecan

(CPT-11, inhibitor of topoisomerase I) in the presence or absence of p18-Cyclin E. The

treated cells were subjected to neutral comet assay (single-cell gel electrophoresis). The residual, unrepaired DSBs were determined by quantification of the tail lengths and

moments of the resulting comets (Figures 2.3A and 2.3B upper panels). We found that p18-Cyclin E impairs the repair of DNA damage induced by IR (Figure 2.3A lower panel) or CPT-11 (Figure 2.3B lower panel). Taken together, these data indicate that p18-

Cyclin E interferes with the DNA repair in both in vitro and in vivo experiments.

90

Figure 2.3 p18-Cyclin E prevents efficient repair of the DNA damage induced by IR

and CPT-11. A) 293T cells were transfected with HA-p18-Cyclin E (p18). At 24 h post-

transfection cells were irradiated (10 Gy) and collected after 16 h. Lysis and

electrophoresis were performed according to the manufacturer’s indications (upper

panel). After the slides were silver stained, the image analysis and quantification was

performed with NIH ImageJ. Tail length (lower left panel) and tail moment (lower right panel) were determined and calculated according to the following formula: Tail moment

= % of DNA in the tail x Tail length; where % of DNA in the tail = tail area (TA) x tail average intensity (TAI) x 100/(TA x TAI) + [head area (HA) x head area intensity

(HAI)]. B) 293T cells were transfected with HA-p18-Cyclin E. At 24 h post-transfection,

cells were treated with 100 ng/mL CPT-11 and collected after 16 h. Neutral comet assay

image analysis was performed as described above. DNA was stained in this case with

SYBR Green. (Data are representative of three independent experiments)

91

92

93

p18-Cyclin E impairs the recruitment of XLF/XRCC4/Ligase IV heterocomplex to

the DNA repair complex

We have shown previously that p18-Cyclin E interacts specifically with Ku70, a protein very well studied for its role in the NHEJ DNA repair pathway (Mazumder et al.,

2007b). Upon appearance of DNA DSBs, Ku70/Ku80 heterodimers bind the free DNA ends to keep them in close proximity and recruit other important DNA repair factors such as DNA-PKcs, Artemis, several polymerases, XRCC4, XLF, and Ligase IV. The interaction between p18-Cyclin E and Ku70, together with the demonstrated ability of p18-Cyclin E to impair the DNA repair, prompted the question whether p18-Cyclin E inhibits the NHEJ machinery by interfering with the Ku70-DNA binding step or with the subsequent recruitment of DNA repair factors. To determine at which step in the NHEJ pathway p18-Cyclin E acts we reproduced the NHEJ repair in nuclear extracts from cells expressing p18-Cyclin E or only GFP as control. The DNA DSBs were mimicked by denatured calf thymus DNA fragments crosslinked to agarose beads. The DNA repair

complexes were allowed to form at the ends of the DNA fragments, then eluted with high

saline buffer and separated by SDS-PAGE. Interestingly, the binding of the Ku70/Ku80

heterodimer to the DNA fragments was not affected at all. Similarly, p18-Cyclin E did

not impact the recruitment of DNA-PKcs to DSBs. In contrast, the recruitment of the

XLF/XRCC4/Ligase IV heterocomplex was dramatically reduced in presence of p18-

Cyclin E (Figure 2.4 upper panel) suggesting its interference with the last step in NHEJ,

the ligation of the DNA ends. Moreover, p18-Cyclin E was present at the sites of DSBs

(Figure 2.4 lower panel). 94

Figure 2.4 p18-Cyclin E impairs the recruitment of XLF, XRCC4, and Ligase IV to the DNA repair complex. Nuclear extracts of HEK293T cells stably expressing p18-

Cyclin E or EGFP were prepared by hypotonic swelling lysis. Extracts were incubated with denatured calf thymus DNA-agarose beads overnight. Beads were washed and the proteins eluted with a high salt buffer and separated by SDS-PAGE. The levels of the recruited proteins were determined with the respective antibodies: DNA-PKcs, Ligase IV,

Ku80, Ku70, XRCC4, and XLF (upper panel). A higher percentage gel was used to determine the presence of p18-Cyclin E, as well as that of Ku70 and Ku80 as controls

(lower panel). (Data are representative of three independent experiments)

95

Discussion

Ionizing radiation and chemotherapeutic treatment are responsible for the induction of several types of DNA damage including base damage, single strand breaks and DSBs (Collis and DeWeese, 2004; Hoeijmakers, 2001; Petrini and Stracker, 2003).

Among these, the DSBs are the most toxic ones, their presence being able to lead to either cell death or chromosomal translocation, which is a major cause for malignant transformation (van Gent et al., 2001). Therefore, their accurate repair is essential for cell survival. In eukaryotic cells, DNA DSBs can be repaired by two major pathways, homologous recombination (HR) (Haber, 1995) or non-homologous end joining (NHEJ)

(Lieber et al., 2003). HR performs the repair with very high accuracy since it employs the sister chromatid as template for the DNA synthesis. For this reason, HR takes place mainly in late S and G2 phases of the cell cycle. On the other hand, NHEJ which is the most common pathway for DSB repair in mammalian cells is very imprecise and may lead to mutations in genome. The purpose of treating tumor cells with DNA-damaging agents such as IR or chemotherapeutics is to induce enough genotoxic stress that would lead to cell death. Attempts to replicate damaged DNA would cause increased cell death, making tumor cells with high replication rates more sensitive to this type of therapy.

However, the toxicity of the DNA-damaging drugs may be reduced in cells that manifest very high levels of DNA repair activity, thus leading to inefficacy of the treatment and survival of the tumor cells. Furthermore, as some of the DNA repair pathways are inactivated in many types of cancers, the DNA repair mechanisms are an attractive target for cancer therapy. 96

Cyclins in association with their catalytic subunits, the cyclin-dependent kinases play an important regulatory role in the cell cycle progression (Murray, 2004). Cyclin

E/Cdk2 complex has an essential role at the rate-limiting step in the G1 to S phase

transition (Dulic et al., 1992; Koff et al., 1991). Loss of its regulation has been shown to lead to aberrant cellular division and malignant transformation (Ohtsubo and Roberts,

1993; Ohtsubo et al., 1995; Resnitzky et al., 1994). We have reported that in cells of hematopoietic origin, genotoxic stress induced by IR leads to caspase-3-mediated proteolytical cleavage of Cyclin E resulting in generation of its C-terminal proteolytic fragment, p18-Cyclin E (Mazumder et al., 2002). Unlike Cyclin E, p18-Cyclin E lacks the Cdk2 binding domain and therefore cannot interact with Cdk2 suggesting that its functions are independent of Cdk2 and different from those of Cyclin E. Furthermore, we have recently demonstrated that by binding to Ku70 in the cytosol, p18-Cyclin E causes the dissociation of Bax from Ku70 and its activation, thus leading to apoptosis

(Mazumder et al., 2007a; Mazumder et al., 2007b).

The nuclear abundance of Ku70 as well as the cytosolic and nuclear presence of

p18-Cyclin E warranted the investigation of its involvement in NHEJ DNA repair. This

hypothesis was also supported by the finding that stable, non-toxic levels of p18-Cyclin E

were able to sensitize cells to different DNA-damaging agents such as IR or etoposide.

The fact that no apoptosis was observed in untreated control cells suggests that p18-

Cyclin E was expressed at levels below those that would trigger release of Bax in the

cytosol. As p18-Cyclin E and Bax expression levels are quite low compared to those of

Ku70, stoichiometrically, it is less likely that a p18-Cyclin E molecule would meet and 97

interact with a Ku70/Bax complex to result in the release of Bax. This stable cell line allowed us to study the effect of p18-Cyclin E strictly on DNA repair in the nucleus. The observed sensitivity of p18-Cyclin E-expressing cells to genotoxic stress suggested that this may be a result of unrepaired DNA damage which caused the cells to undergo apoptosis.

To test this hypothesis, we next examined the effect of p18-Cyclin E on in vitro plasmid DNA ligation. Increasing amounts of p18-Cyclin E in nuclear extracts effectively inhibited the ligation reaction as shown by the presence of DNA only in linear, monomeric form. On the other hand, Cyclin E did not interfere with the end ligation and allowed the reaction to take place to a similar extent as that catalyzed by T4 DNA ligase.

This indicated that the presence of p18-Cyclin E could account for the inability of DNA repair enzymes in the nuclear extracts to perform the ligation. This conclusion was further confirmed by the extensive reduction in number of colonies resulted from transformation of the DNA in E.coli. The capability of p18-Cyclin E to prevent the repair of DNA damage induced by IR or CPT-11 was also indicated by the increased tail length and moment measured in a neutral comet assay. Taken together, these data clearly prove the involvement of p18-Cyclin E in the regulation of DNA repair and presumably of

NHEJ as suggested by its interaction with Ku70.

The NHEJ DNA repair pathway relies on the assembly of a large protein complex at the DNA DSB. This process starts with the binding of the Ku70/Ku80 heterodimer to the free DNA ends (DeFazio et al., 2002) followed by the recruitment of the large molecular weight DNA-PKcs (Smith and Jackson, 1999). This kinase, in turn, 98

phosphorylates and recruits other important repair enzymes such as Artemis and several

polymerases (Koch et al., 2004; Ma et al., 2002). The final step of the repair process, the

ligation, is performed by the XLF/XRCC4/Ligase IV heterocomplex (Critchlow et al.,

1997; Grawunder et al., 1997). The interaction of Ku70 with p18-Cyclin E would suggest that the most likely step in the pathway that p18-Cyclin E may interfere with, would be the association of Ku70/Ku80 with the DNA ends, a destabilization of the Ku70/Ku80 complex, or the recruitment of DNA-PKcs. However, a DNA pull-down assay which reconstitutes the DNA repair complex assembly did not show any difference in the synapsis formation by the Ku70, Ku80, and DNA-PKcs proteins. Interestingly, the recruitment of the XLF/XRCC4/Ligase IV heterocomplex was severely impaired, suggesting that this may be prevented by the binding of p18-Cyclin E to Ku70.

Several studies have shown that XRCC4/Ligase IV can be recruited to the DSB repair site even in the absence of DNA-PKcs (Costantini et al., 2007; Mari et al., 2006) and that it is Ku70/Ku80 that is indispensable for recruiting Ligase IV. Furthermore, it has been reported that Ligase IV interacts directly with Ku and that Ku70 and Ku80 have to be present in the heterodimeric form. But, even though DNA-PKcs may not be required for attracting Ligase IV to the DNA repair complex, it appears that this has a stabilizing effect on its recruitment (Calsou et al., 2003). This provides an explanation for the mechanism of how p18-Cyclin E regulates NHEJ. Binding of p18-Cyclin E to Ku70 could prevent the interaction between Ku and the XLF/XRCC4/Ligase IV complex resulting in inhibition of the ligation step (Figure 2.5). 99

The fact that Ku stimulates Ligase IV activity and that XRCC4 stabilizes the

NHEJ complex by interacting with DNA-PKcs (Calsou et al., 2003; Hsu et al., 2002;

Kysela et al., 2003) explains how diminished recruitment of XLF, XRCC4, and Ligase

IV caused by p18-Cyclin E could lead to impairment of the DNA ligation.

The ability of p18-Cyclin E to prevent the DNA repair in the nucleus and to trigger the release and activation of Bax in the cytosol are indicative of the important role played by p18-Cyclin E in the amplification of the apoptotic signal, thus committing the cells to apoptosis. In conclusion, the recently discovered interaction between p18-Cyclin

E and Ku70 uncovers a new role of Cyclin E in the regulation of cellular physiology which may be unique in linking cell cycle control, DNA repair, and activation of apoptosis following genotoxic stress as well as provide mechanistic insights into the choice between cell death and cell survival depending on the cell type and nature of the genotoxic challenge.

100

Figure 2.5 Schematic representation of the NHEJ DNA repair pathway and the way

in which p18-Cyclin E affects the assembly of repair factors. The free DNA ends of a

DSB are bound very rapidly and tightly by the Ku70/Ku80 heterocomplexes. These complexes slide inwards on the DNA and recruit the large serine/threonine kinase DNA-

PKcs which together with Ku70/Ku80 forms the synaptic complex keeping the DNA

ends in close proximity. DNA-PKcs phosphorylates and recruits Artemis (overhang

resection) and several polymerases (nucleotide fill-in) necessary for the processing of the

DNA ends in order to make them compatible for ligation. Finally, XLF/XRCC4/Ligase

IV is recruited to the DNA repair complex to perform the final step of the repair pathway,

the ligation of DNA ends. However, in the presence of p18-Cyclin E, the recruitment of

this heterocomplex is impaired and the repair of the DNA damage fails to take place,

resulting in cell death. 101

CHAPTER III

Two new mechanisms for proteasome-mediated Cyclin E degradation uncovered in

hematopoietic tumor cells undergoing apoptosis

Abstract

We have shown earlier that in hematopoietic tumor cells, caspase-mediated

cleavage of Cyclin E generates p18-Cyclin E. Its expression can induce apoptosis or

sensitize to apoptotic stimuli in many cell types by associating with Ku70, leading to dissociation of Bax from Ku70 and its activation. However, it has a much shorter half-life than Cyclin E. Here, we show that p18-Cyclin E is more effectively ubiquitinated and degraded by the 26S proteasome. Interestingly, recognition by the Skp1-Cul1-Fbw7

(SCF) complex and the interaction with the Fbw7 family of protein isoforms can take place independently of phosphorylation of p18-Cyclin E or its association with Cdk2.

However, the SCFFbw7 complex is implicated in this ubiquitination process only to a limited extent, with a novel pathway involving Ku70 and Hdm2 being more effective in the degradation of p18-Cyclin E. In addition, blocking its degradation using specific proteasome inhibitors, such as Bortezomib and MG132 can stabilize p18-Cyclin E as well as prolong its association with Ku70 and thereby, lead to enhanced apoptosis.

Moreover, cells expressing p18-Cyclin E are more sensitive to Bortezomib treatment. By preventing its proteasomal degradation, p18-Cyclin E may become an effective

102 103

therapeutic target for Bortezomib and apoptotic effectors in various hematopoietic

malignancies.

Introduction

Deregulation of cellular proliferation together with defects in apoptosis control are two critical processes responsible for tumor formation (Green and Evan, 2002).

Cyclins, in association with their catalytic subunits, the cyclin-dependent kinases

(CDKs), control cell cycle progression by regulating events that drive the transition between cell cycle phases (Elledge, 1996; Murray, 2004; Reed, 2003). The Cyclin

E/Cdk2 complex plays an essential and rate-limiting role in the transition between the G1 and S phases of the cell cycle (Ohtsubo et al., 1995; Resnitzky and Reed, 1995) and in the initiation of DNA replication (Jackson et al., 1995; Ohtsubo et al., 1995). Regulation of

Cyclin E takes places at both transcriptional and post-transcriptional level.

Transcriptionally, Cyclin E is a target of the E2F transcription factor family whereas post-transcriptionally, Cyclin E is targeted by the ubiquitin-proteasomal pathway to 26S proteasomal degradation (Clurman et al., 1996; Won and Reed, 1996). The required ubiquitination is achieved following recognition by and interaction with the E3 ubiquitin ligase, the F-box tumor suppressor protein, Fbw7 (also known as hCdc4). Fbw7 is a specificity factor for the Skp1-Cul1-F-box protein ubiquitin ligase complex that targets a variety of proto-oncogene products for ubiquitin-mediated degradation, including Cyclin

E (Zhang and Koepp, 2006). Cullin-3 (Cul3) on the other hand is responsible for the free 104

Cyclin E degradation (McEvoy et al., 2007; Singer et al., 1999). The role of Cyclin E expression in apoptosis, particularly independent of its interaction with Cdk2 is not clear.

The ubiquitin-proteasome pathway is essential for the degradation of the majority of intracellular proteins (Hershko and Ciechanover, 1998). A number of key regulatory proteins involved in cell proliferation, differentiation, survival, as well as cell death are regulated by proteasome-mediated proteolysis, that results in the activation or inhibition of specific cell signaling pathways (Adams, 2004). Apoptosis is regulated by the ubiquitin/proteasome system at different levels and importantly, inhibition of proteasome function may induce cell death (Herrmann et al., 1998). Recently, inhibition of the proteasomal function with specific inhibitors has evolved as a novel approach for the treatment of various cancers (Aghajanian et al., 2002; Chauhan et al., 2005; Papandreou et al., 2004). Highly specific inhibitors of the 26S proteasome, such as the reversible inhibitor MG132 have been shown to induce apoptosis in many tumor cell lines (Adams and Kauffman, 2004; Zhang et al., 1999). Bortezomib (PS-341, Velcade), a modified dipeptydil boronic acid is a potent and selective inhibitor of the 26S proteasome, a multisubunit protein complex responsible for the regulation of the turnover of cellular proteins, thus maintaining cellular homeostasis. It controls protein expression and function by degradation of ubiquitinated proteins as well as cleanses the cell of abnormal or misfolded proteins. Bortezomib inhibits the proteasome by binding reversibly to the chymotrypsin-like site in the 20S core. The proteolytic inhibition disrupts the homeostasis leading to cell death. 105

We have shown that Cyclin E is critical for genotoxic stress-induced apoptosis of

tumor cells of hematopoietic origin (Mazumder et al., 2000). An 18-kDa Cyclin E

fragment (p18-Cyclin E) is generated through caspase-mediated proteolytic cleavage of

Cyclin E (Mazumder et al., 2002). Cleavage of Cyclin E results in the generation of p18-

Cyclin E, which lacks the Cdk2-interaction domain as well as the N-terminal

phosphodegron known to be necessary for the optimal ubiquitination and degradation of

Cyclin E. However, the presence of the main phosphodegron comprised of the Ser372,

Thr380, and Ser384 residues would suggest a similar regulatory mechanism of p18-

Cyclin E as Cyclin E by the SCF complex. Here we show that, indeed, the turnover of

p18-Cyclin E can be regulated by SCFFbw7. Strikingly, the recognition of p18-Cyclin E by the SCFFbw7 can be independent of its phosphorylation at the Cdc4 phosphodegron (CPD)

(Nash et al., 2001) or binding to Cdk2. Moreover, SCF has a minor contribution to the

degradation of p18-Cyclin E suggesting a parallel mechanism responsible for its turnover

that is independent of Cul1 or Cul3 complexes or the C-terminus of Hsp70 interacting

protein (CHIP). Here we propose that a novel pathway involving Ku70, known as a

critical component of Non Homologous End-joining DNA repair, that we have recently

identified as a specific p18-Cyclin E-interacting protein, plays a significant role in the

proteasomal degradation of p18-Cyclin E. We are reporting here the interaction between

Ku70 and the E3 ubiquitin ligase Hdm2 (Human homologue of Murine Double Minute).

Hdm2 is known for negatively regulating and ubiquitinating the p53 tumor suppressor

protein (Haupt et al., 1997; Kubbutat et al., 1997). Furthermore, Hdm2 has been shown to 106

associate with Ku70 in human umbilical vein endothelial cells (HUVEC) (Gama et al.,

2008, unpublished).

While attractive, the short half-life of p18-Cyclin E limits its clinical use. Since extensive ubiquitination after its generation leads to rapid proteasomal degradation, we tested whether enhancing its stability using proteasome inhibitors may potentiate its effect. Indeed, blocking its degradation caused a massive elevation of its levels as well as its association with Ku70 leading to Bax activation and enhanced apoptosis. Cells expressing p18-Cyclin E were more sensitive to cell death induction upon treatment with

Bortezomib, as compared to the parental cells. By preventing its proteasomal degradation, p18-Cyclin E may become an effective therapeutic target for Bortezomib when it is used in the therapy of hematopoietic or solid cancers.

Materials and Methods

Cell lines and treatments. HEK293T human kidney and NCI-H1299 non-small cell lung

carcinoma cells were maintained in DMEM (Gibco-BRL) containing 10% fetal bovine

serum (Hyclone, Logan, UT) with L-glutamine, and 100 units/ml penicillin/streptomycin.

Cells ~75% confluent were irradiated with 5 Gy (137Cs source; fixed dose rate of 2.0

Gy/min) (Gong et al., 1999). Bortezomib (Velcade) was obtained from Millenium

Pharmaceuticals (Cambridge, MA), MG132 and cycloheximide from Sigma (St. Louis,

MO). For the half-life study, cells were treated with 10 µM cycloheximide and collected 107

at the indicated times after treatment. HA-p18-Cyclin E stably expressing 293T and

H1299 cells were generated by infecting cells with a lentivirus carrying HA-p18-Cyclin E as well as EGFP separated by an IRES2 sequence and under the control of an EF1α

promoter.

Western blotting and immunoprecipitation analyses. For immunoblots, cells were

lysed in a buffer consisting in 20 mM HEPES, pH 7.5, 1 mM EDTA, 150 mM NaCl, 1%

NP-40, 1 mM DTT and protease inhibitors (1 mM PMSF, 2 µg/ml aprotinin, leupeptin,

and pepstatin) and immunoblotting was performed with anti-HA (Covance; Berkeley,

CA), Cyclin E1 (HE-12), Cdk2 (D-12), p53 (DO1), Ku70 (E-5 and H-308; Santa Cruz

Biotechnology; Santa Cruz, CA), PARP-1 (1:1000; Cell Signaling; Beverly, MA), and β-

actin (Sigma) antibodies. For immunoprecipitation, the lysis buffer composition was

similar to that employed for Westerns except that instead of 1% NP-40, for the

interactions between Cdk2 and p18-Cyclin E or Cyclin E, 0.1% NP-40 was used, while

for the interactions between p18-Cyclin E and Fbw7α, β, or γ, 0.2% NP-40 was used. For the interaction between Bax and Ku70 cells were lysed in 1% CHAPS.

Immunoprecipitation was performed with Bax antibody (N-20, Santa Cruz

Biotechnologies). Cell lysates (500-1000 µg) were incubated with 0.5-2 µg antibody for 2 h at 4oC, followed by incubation with Protein A plus G agarose beads for 1 h at 4oC.

Beads were washed with lysis buffer and boiled with 2x sample buffer. The eluted

proteins were resolved by SDS-PAGE and Western blotting was performed using the

appropriate antibodies, as described above. For determining the interaction between 108

Hdm2 and Ku70 in HEK293T cells, untreated or 2h etoposide (10 µM) treated cells were lysed in 1% Triton-X buffer in PBS, containing 1x protease inhibitor cocktail (Sigma), and 1 mM PMSF. Immunoprecipitation was performed with 2 µg anti-Hdm2 antibody

(2A10; Calbiochem: San Diego, CA) for 2 h at 4°C. 2 µg normal mouse IgG were used as

a negative control. For the determination of Ku70 and Hdm2 interaction in IM-9 cells,

these were irradiated with 4 Gy and collected after 8h.

For determining the ubiquitination status, cells were treated with 25 µM MG132

prior to harvesting and lysed in the same 1% NP-40 lysis buffer as described above also

containing 1x HALT phosphatase inhibitor cocktail (Pierce, Rockford, IL), 5 µM MG132 and 5 µM N-ethyl-maleimide (Sigma). Immunoprecipitation was performed with HA or

FLAG antibody (Sigma) for tagged p18-Cyclin E, with (HE-111, Santa Cruz

Biotechnology) for Cyclin E for 3 h at 4oC followed by incubation with Protein A plus G agarose beads for 1 h. Immunoblotting was performed with either HA antibody, anti-

Ubiquitin mouse monoclonal antibody (Chemicon, Temecula, CA) or rabbit polyclonal antibody (Dako, Carpinteria, CA).

Transfections. H1299 cells were transfected with 10 µg p18-Cyclin E-HA and 3 µg

FLAG-Fbw7α, β, or γ. At 24 h post-transfection, cells were collected and lysed.

Immunoprecipitation was performed with anti-HA antibody followed by immunoblotting

with anti-FLAG antibody. For the overexpression of FLAG-tagged Fbw7α, β, γ and dominant negative Cul1, dominant negative Cul3, 3 µg of construct and Lipofectamine

2000 (Invitrogen, Carlsbad, CA) was used according to the protocol provided by the 109

manufacturer. Knock-down of Cdk2 and Fbw7 was performed using 75-100 nM of

siRNA from Dharmacon (Lafayette, CO). As a negative control we used siCONTROL

non-targeting siRNA pool #1 from Dharmacon. In both the overexpression and knock-

down experiments cells were collected after 48 h. To effectively knock-down Ku70,

293T cells were transfected with 100 nM siKu70 or siCONTROL (Dharmacon). At 24 h

post transfection, cells were collected and plated followed by a second transfection the

second day with cells being finally collected the next day (72 h after the initial

transfection). Overexpression of CHIP (kind gift form Dr. E. Ficker, Metrohealth,

Cleveland) was achieved by transfection of an expression construct followed by collection after 24h. CHIP was knocked-down using siRNA from Dharmacon. The transfected cells were collected after 24 and 48 h respectively.

Construction of expression vectors and p18-Cyclin E phosphomutants. FLAG-tagged

p18-Cyclin E was constructed by subcloning p18-Cyclin E into the HindIII/BamHI

restriction sites of the p3xFLAG-myc-CMV-24 vector (Sigma) using the forward primer

5’-CGACAAGCTTGCCATGGACTGCCTTG-3’ and the reverse primer 5’-

TAGATGGATCCTCTCACGCCATTTCCGG-3’ resulting in an expression construct

that has the 3xFLAG at the N-terminus, but no myc tag. S384A-p18-Cyclin E was created

by mutating the Ser384 to Ala in the HA-tagged p18-Cyclin E using the ExSite PCR-

based site-directed mutagenesis from Stratagene (La Jolla, CA) and the primers

S384A-b: 5’-CTGTGGCGGGGTGAGGAGC-3’ and S384A-P: 5’-phospho-

GCCGGTAAGAAGCAGAGCAGCG-3’. C-terminal HA-tagged T380A-p18-Cyclin E 110

was created by subcloning T380A-p18-Cyclin E from the T380A-Cyclin E-GFP

construct obtained from Dr. Geisen (Burnham Institute) (Geisen and Moroy, 2002) into

the C-terminal HA-containing pcDNA3.1 vector. The resulting construct was used for the

generation of S372A/T380A-p18-Cyclin E by mutating the Ser372 to Ala using the

primers S372A-b: 5’-AGCCCTATTTTGTTCAGACAACATG-3’ and S372A-P: 5’-

phospho-GCTCCTCTCCCCAGTGGGCTCCT-3’. The resulting construct was further

used for the generation of S372A/T380A/S384A-p18-Cyclin E using the primers S384A-

T380A: 5’-CTGTGGCGGCGCCAGGAGC-3’ and S384A-P. The T380A/S384A-p18-

Cyclin E was generated by mutating Ser384 to Ala in T380A-p18-Cyclin E using the primers S384A-T380A and S384A-P. All mutations were verified by DNA sequencing.

Results

p18-Cyclin E is a short-lived protein that is stabilized by proteasome inhibition

Our previous experiments have shown that Cyclin E is cleaved during genotoxic

stress induced by IR generating an 18-kDa C-terminal fragment, p18-Cyclin E

(Mazumder et al., 2007b). To examine the stability of p18-Cyclin E compared to full

length Cyclin E (Cyclin E), the half-life of these two proteins was determined following

protein translation inhibition by Cycloheximide treatment. Results of these experiments

indicate that p18-Cyclin E had a remarkably short half life of less than 0.5 h. In contrast,

the half-life of Cyclin E was longer than 8 h in HEK293T cells stably expressing p18-

Cyclin E (Fig 3.1A). These cells were generated in order to overcome the drawbacks 111

related to transient expression of p18-Cyclin E. Furthermore, these cells express p18-

Cyclin E at low, non-toxic levels and Cyclin E cleavage does not take place.

Strikingly, proteasome inhibition achieved by treatment with either MG132 or the new, clinically approved compound, Bortezomib (Velcade) led to a dramatic stabilization of p18-Cyclin E, indicating that this protein is very rapidly turned over by its proteasome- mediated degradation. Protein stabilization took place in both HEK293T and NCI-H1299 cells stably expressing p18-Cyclin E (Fig 3.1B). Interestingly, the stabilizing effect of proteasome inhibition on Cyclin E was much less robust than that on p18-Cyclin E.

112

Fig 3.1 p18-Cyclin E is a short-lived protein that is stabilized by proteasome

inhibition. A) 293T cells stably expressing HA-tagged p18-Cyclin E were treated with

10 µM Cycloheximide for the indicated times. The levels of p18-Cyclin E, Cyclin E, and

β-actin used as a loading control were determined by Western blotting using the respective primary antibodies. B) 293T and H1299 cells stably expressing HA-p18-

Cyclin E were either untreated (Control, C) or treated with 1 µM Bortezomib

(Velcade:Vel) or 10 µM MG132 (MG) and collected after 3 h. The levels of p18-Cyclin

E, Cyclin E, and β-actin were determined by Western blotting. (Data are representative of three independent experiments)

A

0.25 .5124824Time(h) - Cyclin E

- p18-Cyclin E

- β-actin

B

293T H1299 C Vel MG C Vel MG -CyclinE - p18-Cyclin E - β-actin 113

p18-Cyclin E is effectively ubiquitinated

The above experiments revealed that proteasome inhibition led to a massive

increase in the levels of p18-Cyclin E. Poly-ubiquitination of the target protein is the

most common signal for proteasome-mediated degradation. The higher molecular weight,

modified forms of p18-Cyclin E were readily visible following a long exposure upon

MG132 treatment in the presence of N-ethylmaleimide (NEM), an inhibitor of the

ubiquitin isopeptidases, indicating that p18-Cyclin E is degraded through the addition of

multiple ubiquitin or ubiquitin-like residues that target it for proteasomal degradation

(Fig 3.2A). To determine whether p18-Cyclin E is modified by addition of Ubiquitin,

Flag-tagged p18-Cyclin E was co-transfected with an HA-Ubiquitin expression construct

in 293T cells. Immunoprecipitation with anti-FLAG antibody for p18-Cyclin E followed by immunoblotting with anti-HA antibody for Ubiquitin shows the characteristic smear

(Fig 3.2B left panel) as well as the characteristic laddering (Fig 3.2B right panel) of poly-

ubiquitinated p18-Cyclin E. To compare the extent of ubiquitination between p18-Cyclin

E and Cyclin E, the latter was immunoprecipitated followed by immunoblotting with an

anti-HA antibody for the ubiquitinated protein forms (Fig 3.2B left panel). This

experiment revealed that the extent of Cyclin E ubiquitination was less than that of p18-

Cyclin E, a result which confirms the finding above that Cyclin E has a much longer half-

life than its carboxy terminal fragment, p18-Cyclin E (Fig 3.1A). Addition of poly-

ubiquitin chains was also observed when HA-p18-Cyclin E was immunoprecipitated from stably expressing 293T cells followed by immunoblotting with two different anti-

Ubiquitin antibodies (Fig 3.2C). 114

Fig 3.2 p18-Cyclin E is effectively ubiquitinated. A) 293T cells were transfected with

HA-tagged p18-Cyclin E or empty vector (C) and treated 24 h later with 2 µM or 10 µM

MG132 for an additional 16 h and 4 h, respectively. The levels of p18-Cyclin E and β- actin were determined by Western blotting. B) 293T cells were co-transfected with

FLAG-tagged p18-Cyclin and HA-tagged Ubiquitin. At 24 h post-transfection, cells were treated with 25 µM MG132 for 3 h. p18-Cyclin E and Cyclin E were immunoprecipitated from whole-cell lysates (WCL) with anti-FLAG (1st and 2nd lane) or anti-Cyclin E (3rd lane) antibodies and separated by SDS-PAGE. Ubiquitination of p18-Cyclin E and Cyclin

E (left top panel), levels of p18-Cyclin E and β-actin in WCL (right panel) were

determined by Western blotting. C) 293T cells stably expressing HA-p18-Cyclin E or

only GFP as control were treated with 25 µM MG132 for 3 h. p18-Cyclin E was immunoprecipitated using anti-HA antibody. Ubiquitination of p18-Cyclin E (left panel rabbit anti-Ubiquitin, middle panel mouse anti-Ubiquitin), levels of p18-Cyclin E (right panel HA antibody), and β-actin were determined by Western blotting.

115

A

C p18-Cyclin E --2µM10µM MG132 16h 4h

ubiquitinated forms p18-Cyclin E

- β-actin

B

Ub-HA + + + + + + + + + FLAG-p18-Cyclin E + - - + - - + - - poly-Ubiquitinated - protein poly-Ub- p18-Cyclin E

- p18-Cyclin E IgGκ - - β-actin WB: HA

IgGλ - p18-Cyclin E - IP: FLAG CycE WB: HA WB: HA

116

C

GFP p18 GFP p18 GFP p18

poly-Ub poly-Ub p18-Cyclin E p18-Cyclin E poly-Ub- p18-Cyclin E -IgGκ -IgGκ -IgGλ IP: HA -IgGλ WB: pAb Ubiquitin - β-actin IP: HA WB: mAbUbiquitin

117

Fbw7 interacts with p18-Cyclin E independently of phosphorylation at the C-

terminal phosphodegron

Cyclin E is known to be degraded through the ubiquitin-proteasomal pathway that

uses Fbw7 as a component of the Skp1-Cul1-F-box (SCF) complex (Koepp et al., 2001;

Moberg et al., 2001; Strohmaier et al., 2001). There are three splice-derived isoforms of

Fbw7 designated as α, β, and γ that localize to different cellular compartments (Spruck et al., 2002). To determine whether p18-Cyclin E is also targeted for proteasomal degradation by the SCFFbw7 complex, we examined the interaction between p18-Cyclin E and all three Fbw7 isoforms. To determine the interactions of p18-Cyclin E with the different isoforms of Fbw7, HEK293T cells were transiently transfected with HA-p18-

Cyclin E and FLAG-Fbw7α, β, γ expression constructs and immunoprecipitated with

anti-HA to pull-down p18-Cyclin E followed by Western blot performed with an anti-

FLAG antibody. p18-Cyclin E associated with all three isoforms of Fbw7, which indicates that it is a ubiquitination target of Fbw7, which regulates its degradation (Fig

3.3A).

Phosphorylation plays an important role in the interaction between the Fbw7 isoforms and Cyclin E (Koepp et al., 2001; Moberg et al., 2001; Strohmaier et al., 2001).

To examine the influence of phosphorylation upon the expression levels of p18-Cyclin E, amino-acid residues known to be important for the phosphorylation-dependent turnover

of Cyclin E (Ye et al., 2004) were mutated alone or in combination. We generated the

following phosphorylation mutants: T380A, S384A, T380A/S384A, S372A/T380A, and

S372A/T380A/S384A. Unlike the effect that is known for these point mutations on 118

Cyclin E (Ye et al., 2004), transient transfection of these mutant p18-Cyclin E expression

constructs has shown very striking differences. The Thr380 residue, which has been

reported to play a crucial role in the turnover of Cyclin E (Clurman et al., 1996; Won and

Reed, 1996), does not affect the stability of p18-Cyclin E. Moreover, the mutation of

Ser384 instead of increasing the levels of p18-Cyclin E leads to its destabilization. Only

mutation of Ser372 showed the expected increase in levels of the protein (Fig 3.3B), with

the triple mutant having less effect, most likely because of the destabilizing effect of the

Ser384 mutation. Furthermore, co-transfection of the HA-tagged triple phosphorylation

mutant S372A-T380A-S384A-p18-Cyclin E with the Fbw7α, β, and γ isoforms in 293T cells still showed their interaction (Fig 3.3C). These data indicate that p18-Cyclin E may

interact with Fbw7α, β, and γ independently of phosphorylation at S327, T380 or S384 that constitute the C-terminal phosphodegron of Cyclin E.

119

Fig 3.3 Fbw7 interacts with p18-Cyclin E independently of phosphorylation at the

C-terminal phosphodegron. A) H1299 cells were co-transfected with HA-tagged p18-

Cyclin E and FLAG-tagged Fbw7α, β, or γ. p18-Cyclin E was immunoprecipitated from whole cell lysates with anti-HA antibody and separated by SDS-PAGE. Levels of immunoprecipitated Fbw7α, β or γ were determined by Western blotting with anti-Flag antibody. B) 293T cells were transfected with empty pcDNA3.1 vector or containing HA- p18-Cyclin E and its derivative mutants: T380A, S384A, T380A/S384A,

S372A/T380A/S384A, and S372A/T380A. Their levels were determined by Western blotting using the anti-HA antibody. C) 293T cells were co-transfected with HA-

S372A/T380A/S384A-p18-Cyclin E (triple mutant - TM) and FLAG-Fbw7α, β, or γ.

S372A/T380A/S384A-p18-Cyclin E was immunoprecipitated from whole cell lysates with anti-HA antibody and separated by SDS-PAGE. Levels of immunoprecipitated

Fbw7α, β, or γ were determined by Western blotting with anti-Flag antibody. (* - cross- reacting protein) D) Schematic representation of Cyclin E phosphodegrons showing the relative positions of the nuclear localization sequence (NLS) and its two groups of phosphorylation sites at the N-terminus (S58, T62, S75, S88) and C-terminus (S372,

T380, S384) necessary for its optimal recognition by SCFFbw7 and ubiquitination.

Caspase-3 dependent cleavage at Asp275 generates an 18-kDa carboxy terminal fragment, which carries only the C-terminus phosphodegron. (Data are representative of three independent experiments)

120

A

Fbw7 C p18 Lys -Fbw7α

-IgG -IgG

-Fbw7β

-Fbw7γ

-IgG IP: HA WB: Flag B

- p18-Cyclin E - β-actin

121

C

IP TM Lys ++++TM +--+ Fbw7 -Fbw7β -IgG -Fbw7γ -IgG IP TM Lys + + + + TM - + - + Fbw7 -Fbw7α *

D

122

p18-Cyclin E is regulated by the SCFFbw7 complex and independently of Cdk2 binding

To further determine the role of Fbw7 in p18-Cyclin E regulation, p18-Cyclin E- expressing 293T cells were transfected with the α, β, and γ isoforms of Fbw7. Fbw7

overexpression enhanced the turnover of p18-Cyclin E as well as that of Cyclin E.

Conversely, the ectopic expression of dominant negative (dn) Cul1 led to a decrease in

their degradation (Fig 3.4A). The effect on p18-Cyclin E levels was limited while that on

Cyclin E was dramatic leading to a massive accumulation of the protein. The striking

difference in the increase in levels of Cyclin E as compared to p18-Cyclin E would

suggest a different mechanism employed for their regulation by the SCFCul1 complex.

Cyclin E is known to be regulated through both the Cul1 and Cul3 pathways

(Koepp et al., 2001; McEvoy et al., 2007; Singer et al., 1999). To determine the influence

of Cul1 and Cul3 upon the degradation of p18-Cyclin E, stably expressing 293T-p18-

Cyclin E cells were transfected with dnCul1 (Jin et al., 2003) or dnCul3 (Cullinan et al.,

2004) expression constructs. Even though this led to a minor stabilization of p18-Cyclin

E (Fig 3.4B), this increase was far less compared to the robust increase in levels of Cyclin

E (Fig 3.4A). This suggests that even if SCFCul1-Fbw7 plays a role in the degradation of

p18-Cyclin E, this mechanism represents only a minor component and that another

regulatory pathway plays a predominant role in its turnover. Moreover, these data

indicate that Cul3, known to be important for degradation of free Cyclin E (McEvoy et

al., 2007; Singer et al., 1999) has no role in p18-Cyclin E regulation. Finally, to clearly

establish the role of Fbw7 in the turnover of p18-Cyclin E, RNA interference (RNAi) was 123

used to diminish Fbw7 levels in 293T cells stably expressing p18-Cyclin E.

Immunoblotting with anti-HA antibody for p18-Cyclin E and anti-Cyclin E (HE-12)

antibody for Cyclin E (Fig 3.4B) at 48 h post-transfection, indicated that their levels

increased, indicating a regulatory role for Fbw7.

Cdk2 is known to be responsible for the phosphorylation of Ser384 on Cyclin E

when it is present in complex with Cdk2 (Welcker et al., 2003). To assess whether Cdk2

affects the levels of p18-Cyclin E, even in the absence of their interaction (Fig 3.5A and

3.5B), Cdk2 was knocked-down by siRNA (Fig 3.4C). However, despite a robust

depletion of Cdk2 levels, there was only a minimal increase in p18-Cyclin E levels

despite the lack of Cdk2 binding domain and its implicit interaction with Cdk2. Cyclin E

increased to some extent in the absence of Cdk2 but much less compared to when Fbw7

was knocked down. The C-terminus of Hsp70 interacting protein (CHIP) is an E3 ligase

that partners with chaperones Hsp70, Hsc70, and Hsp90 targeting the degradation of misfolded proteins. Furthermore, it has been shown to participate in the proteasome- mediated degradation of base excision repair proteins such as XRCC1 and Pol β, which have not been recruited to a DNA repair complex (Sobol, 2008). To determine whether

CHIP contributes to the turnover of p18-Cyclin E, it was overexpressed in 293T cells stably expressing HA-p18-Cyclin and at 48 h post-transfection the levels of p18-Cyclin E were determined by Western blotting. It appears that CHIP does not potentiate the degradation of p18-Cyclin E. At most, it may show a slightly opposite effect (Fig 3.4D).

Moreover, knocking-down CHIP by siRNA also showed no significant effect on the levels of p18-Cyclin E (Figure 3.4E). 124

Fig 3.4 p18-Cyclin E is regulated by the SCFFbw7 complex and independently of

Cdk2 binding. A) 293T cells stably expressing HA-tagged p18-Cyclin E were transfected with a combination of FLAG-tagged Fbw7α, β and γ or dominant negative

Cul1 (dnCul1). The levels of Fbw7α, β, γ, dnCul1, Cyclin E, p18-Cyclin E, and β-actin were determined by Western blotting. B) 293T cells stably expressing HA-p18-Cyclin E were transfected with dominant negative Cul1 (dnCul1) and dominant negative Cul3

(dnCul3) expression constructs. The levels of dnCul1, dnCul3, p18-Cyclin E, and β-actin, as a loading control, were determined by Western blotting. C) 293T cells stably

expressing HA-p18-Cyclin E were transfected with 100 nM siFbw7, siCdk2, and

siControl. The levels of Cyclin E, Cdk2, p18-Cyclin E, and β-actin were determined by

Western blotting. D) 293T cells stably expressing HA-p18-Cyclin E were transfected with CHIP or empty vector (C) and cells were collected after 48 h. The levels of CHIP, p18-Cyclin E and β-actin were determined by Western blotting. E) 293T stably

expressing HA-p18-Cyclin E were transfected with 100 nM siCHIP or siCONTROL and

cells were collected after 24 or 48h. The levels of CHIP, p18-Cyclin E, and β-actin were determined by Western blotting. (Data are representative of three independent experiments)

125

A

C -Fbw7α -Fbw7β,γ -dnCul1 - Cyclin E

- p18-Cyclin E - β-actin

B

C -dnCul1 -dnCul3 - p18-Cyclin E -dnCul1 - β-actin C

- Cyclin E -Cdk2

- p18-Cyclin E

- β-actin 126

D

C CHIP -CHIP - p18-Cyclin E - β-actin E

-CHIP -p18-CyclinE - β-actin

127

Ku70 but not Cdk2 binding regulates p18-Cyclin E stability

We have previously identified p18-Cyclin E as the carboxy terminal fragment of

Cyclin E, generated following a caspase-3-mediated cleavage (Mazumder et al., 2002).

p18-Cyclin E encompasses the 276-395 amino-acid domain of Cyclin E and thus, is

lacking the Cdk2 binding domain (Mazumder et al., 2007a). To confirm that indeed

Cyclin E binds Cdk2 while p18-Cyclin E has lost this ability, HA-p18-Cylin E was

immunoprecipitated from 293T cells stably expressing it followed by immunoblotting for

Cdk2 (Fig 3.5A). Conversely, Cdk2 was immunoprecipitated from 293T expressing HA-

Cyclin E or HA-p18-Cyclin E respectively, followed by immunoblotting with HA

antibody (Fig 3.5B). These experiments, together with our previous report (Mazumder et

al., 2002) clearly indicate that p18-Cyclin E does not interact with Cdk2 while Cyclin E,

as expected, does. Taken together these data suggest that even though p18-Cyclin E does

not interact with Cdk2, Fbw7 can recruit it to the SCF complex and promote its

ubiquitination.

To examine the possible role of Ku70 binding in the degradation of p18-Cyclin E,

293T cells were transfected with HA-p18-Cyclin E alone or in the presence of FLAG-

Ku70. This led to a dramatic reduction in the levels of p18-Cyclin E (Fig 3.5C). To compare this change with the effect of Fbw7, HA-p18-Cyclin E was co-transfected with

Fbw7α, β, and γ. Fbw7 overexpression led to a much more limited increase in the turnover of p18-Cyclin E as compared to that induced by Ku70. To clearly establish the role of Ku70, its levels were substantially depleted by specific siRNA oligonucleotides.

Down-regulation of Ku70 by siRNA led to an increase in p18-Cyclin E levels, a change 128

that could not be observed for Cyclin E (Fig 3.5D). Hdm2 is an ubiquitin E3 ligase

known to be transcriptionally induced by p53 following DNA damage (Haupt et al.,

1997; Kubbutat et al., 1997). Its role is to polyubiquitinate p53 targeting it to proteasomal degradation, thus limiting its effect. The newly discovered interaction between Hdm2 and

Ku70 (Gama et al., 2008, unpublished) raised the question of whether Hdm2 interacts with Ku70 in our experimental system. In IM-9 cells this association was clearly induced following irradiation, that corresponded to increased p53 levels, known to lead to increased levels of Hdm2 (Figure 3.5E). Hdm2 was also immunoprecipitated from control or etoposide (VP16)-treated HEK293T cells and the presence of Ku70 was determined by immunoblotting. Ku70 and Hdm2 interacted in both control and etoposide-treated cells (Figure 3.5F), as p53 is inactive in these cells and therefore levels of Hdm2 are not expected to be induced. Taken together, these data strongly implicate

Ku70 and Hdm2 in a degradation pathway of p18-Cyclin E, the exact role of which and its mechanism still need to be investigated.

129

Fig 3.5 Ku70 but not Cdk2 binding regulates p18-Cyclin E stability. A) 293T cells

stably expressing HA-p18-Cyclin E or GFP as control were lysed with 0.1% NP-40 lysis

buffer. p18-Cyclin E was immunoprecipitated with anti-HA antibody. Levels of

immunoprecipitated Cdk2 and of p18-Cyclin E present in whole cell lysates were

determined by Western blotting. B) Cdk2 was immunoprecipitated from 293T cells transfected with HA-Cyclin E and p18-Cyclin E. Immunoprecipitated Cyclin E and p18-

Cyclin E as well as their levels from whole cell lysates were determined by Western blotting. C) 293T cells were transfected with FLAG-Ku70, or HA-p18-Cyclin E alone, or in combination with Fbw7α, β, γ or Ku70 in presence of HA-p18-Cyclin E. The levels of

Ku70, Fbw7α, β, γ, p18-Cyclin E, and β-actin were determined after 24 h post-

transfection by Western blotting. D) 293T cells stably expressing HA-p18-Cyclin E were transfected with 100 nM siKu70 or siCONTROL. At 48 h post transfection, a second

round of transfection was performed to achieve a satisfactory down-regulation of Ku70.

The levels of Ku70, p18-Cyclin E, and β-actin used as a loading control were determined by Western blotting at 72 h after the initial transfection. E) IM-9 cells were irradiated

with 4 Gy and Hdm2 was immunoprecipitation after 8h. Levels of Ku70, Hdm2, and p53

were determined by immunoblotting. F) Hdm2 was immunoprecipitated from HEK293T

cells and the levels of associated Ku70 were determined by immunoblotting.

130

A

WCL +-HA-p18-CyclinE -Cdk2 -IgGλ -IgGλ - HA-p18-CyclinE

IP: HA B

- + HA-Cyclin E WCL + - HA-p18-Cyclin E - Cyclin E

- p18-Cyclin E

IP: Cdk2 C

---+Fbw7α,β,γ +-+-Ku70 -+++p18-Cyclin E -Fbw7α -Ku70

- p18-Cyclin E

- p18-Cyclin E (long exp.) - β-actin

131

D

-Ku70 - Cyclin E - β-actin - p18-Cyclin E - β-actin E

IP Hdm2 Lysate CIR C IR -Hdm2 - Ku70 -p53 -IgGκ

F

Lys IP Hdm2 CEIgG C E -Ku70

132

p18-cyclin E sensitizes cells to Bortezomib treatment through enhanced association

with Ku70 and release of Bax from Ku70

Of the commonly used proteasome inhibitors tested Bortezomib was more

effective than MG132 in stabilizing p18-Cyclin E (Fig 3.6A). To examine the dynamics

of proteasome inhibition and onset of apoptosis, stably expressing 293T-p18-Cyclin E

cells were treated with 100 nM Bortezomib for 0.5, 1, 2, 4, 8, 24, and 48 h. Western blot

analysis was performed for PARP-1 to determine the extent of cell death, HA for the

expression of p18-Cyclin E, HE-12 for the expression of Cyclin E and β-actin as a

loading control. Apoptosis was detectable as early as 8 h, with ~10% of PARP-1 being

cleaved by 24 h and 50% by 48 h (Fig 3.6B). The level of p18-Cyclin E was increased in

the presence of Bortezomib, due to the inhibition of its proteasomal degradation. p18-

Cyclin E levels reached a maximum as early as 2 h, while Cyclin E levels did not vary

significantly during the same time interval.

While attractive, the perspective of using p18-Cyclin E for cancer treatment is limited by its very short half-life (Fig 3.1A). Therefore, enhancing its stability may

potentiate its effect. To determine whether the levels of p18-Cyclin E can enhance the

response of cells to Bortezomib treatment, control (GFP-expressing) or p18-Cyclin E-

stably expressing cells were examined at 24 h following treatment with increasing

concentrations of Bortezomib. Increased PARP-1 cleavage in p18-Cyclin E-expressing

cells suggested that p18-Cyclin E may be a mediator of Bortezomib-induced cell death

(Fig 3.6C). 133

To determine the mechanism responsible for the observed cell sensitivity, we

examined the interaction between p18-Cyclin E and Ku70 before and after treatment with

50 nM Bortezomib or irradiation. At 16 h after treatment, HA-p18-Cyclin E was

immunoprecipitated with anti-HA antibody followed by immunoblotting for Ku70.

Treatment with Bortezomib, but not IR caused an elevation of the Cyclin E and to an

even larger extent that of p18-Cyclin E levels (Fig 3.6D). Increased p18-Cyclin E levels led to an increase in its association with Ku70 (Fig 3.6E). To determine whether this

association leads to release of Bax from Ku70, Bax is immunoprecipitated followed by immunoblotting for Ku70. As expected, elevated levels of p18-Cyclin E bound more

Ku70, resulting in release of Bax (Fig 3.6F), which correlated with induction of cell death as indicated by cleavage of PARP-1 (Fig 3.6D).

134

Fig. 3.6 p18-cyclin E sensitizes cells to Bortezomib treatment through enhanced association with Ku70 and release of Bax from Ku70. A) 293T cells stably expressing

HA-p18-Cyclin E were treated with 100 nM of Bortezomib (Velcade:Vel) or MG132 and collected at the indicated intervals. The levels of PARP-1, Cyclin E, p18-Cyclin E and β-

actin were determined by Western blotting. B) 293T cells stably expressing HA-p18-

Cyclin E were treated with 100 nM Bortezomib and collected at the indicated times. The

levels of PARP-1, p18-Cyclin E, Cyclin E and β-actin as a loading control were detected by Western blotting. C) 293T cells stably expressing GFP as control or HA-p18-Cyclin E

were treated with the indicated concentrations of Bortezomib and collected after 24 h.

The levels of PARP-1, p18-Cyclin E and β-actin were determined by Western blotting.

D) 293T cells stably expressing HA-p18-Cyclin E were treated with 5 Gy IR or 50 nM

Velcade (Vel) and collected after 16 h. The levels of PARP-1, Cyclin E, p18-Cyclin E and β-actin were determined by Western blotting. E) 293T cells stably expressing HA- p18-Cyclin E were treated with 5 Gy IR or 50 nM Bortezomib and collected after 16 h.

p18-Cyclin E was immunoprecipitated with anti-HA antibody. Levels of

immunoprecipitated Ku70 as well as present in WCL, were determined by Western blotting. F) 293T cells stably expressing HA-p18-Cyclin E were treated with 5 Gy IR or

50 nM Bortezomib and collected after 16 h. Bax was immunoprecipitated with anti-Bax

(N-20) antibody and the levels of associated Ku70 were determined by Western blotting.

135

A

Bortezomib MG132 0 8 24 48 0 8 24 48 Time (h) -PARP

- Cyclin E

- p18-Cyclin E

- β-actin B

0.512482448Time (h) -PARP116kD -PARP85kD p18-Cyclin E - (long exp.) - p18-Cyclin E (short exp.) - Cyclin E - β-actin - β-actin C

293T 293T p18 0 25 100 200 0 25 100 200 Bortezomib (nM) -PARP

- p18-CyclinE

- β-actin

136

D

C IR Bort -PARP -p85 -CyclinE

- p18 Cyclin E

- β-actin

E

C IR Bort -Ku70(shortexp) - Ku70 (long exp)

IP: HA WB: Ku70 F

C IR Bort -Ku70 -IgGκ IP: Bax WB: Ku70

137

Discussion

The expression of Cyclin E, the master regulatory protein of the cell cycle

transition from the G1 to S phase, is controlled at the mRNA level as well as post-

transcriptionally., Cyclin E is regulated post-transcriptionally by the 26S proteasome

following its ubiquitination by the SCF complex. While the Cdk2-bound Cyclin E

requires phosphorylation at a conserved phosphodegron in order to be recognized and

efficiently ubiquitinated by the Skp1-Cul1-Fbw7 complex (Zhang and Koepp, 2006), the

free Cyclin E is degraded following direct interaction with Cul3 (McEvoy et al., 2007;

Singer et al., 1999).

We have previously shown that Cyclin E is regulated by genotoxic stress caused by ionizing radiation (IR) or other chemotherapeutic drugs, and which plays a functional role in apoptosis of hematopoietic cells (Mazumder et al., 2000). Moreover, we have demonstrated that caspase-3 mediates cleavage of Cyclin E that results in generation of its C-terminal proteolytic fragment, p18-Cyclin E. Protein synthesis inhibition studies with cycloheximide in HEK293T cells stably expressing p18-Cyclin E have now determined a half-life of less than 0.5 h for p18-Cyclin E as opposed to more than 8 h for

Cyclin E. This finding indicates that after being generated, p18-Cyclin E is degraded rapidly through the proteasomal degradation pathway. Indeed, the addition of proteasome inhibitors such as MG132 and Bortezomib significantly inhibited the degradation of p18-

Cyclin E, indicating that p18-Cyclin E is a target of the ubiquitin-proteasomal degradation pathway. The higher molecular weight ubiquitinated forms of p18-Cyclin E are detectable using proteasome inhibitors, suggesting the susceptibility of these 138

ubiquitinated forms to degradation. Unlike Cyclin E, p18-Cyclin E cannot interact with

Cdk-2 as it is missing the Cdk-2 binding domain. It could, alternatively be bound and

regulated by the Cul3 degradation pathway similar to free Cyclin E (McEvoy et al., 2007;

Singer et al., 1999). However, we show here that Cul3 does not have a role in the

degradation of p18-Cyclin E.

Degradation of p18-Cyclin E is dependent on ubiquitination by the Skp1-Cul1-

Fbw7 (SCF) indicated by its interaction with the different isoforms of Fbw7, an

interaction directly responsible for the ubiquitin-dependent proteolysis of a number of

proteins including proto-oncogenes such as Cyclin E and c-Myc (Yada et al., 2004).

Strikingly, we find that the interaction between Fbw7 and p18-Cyclin E takes place even in the absence of p18-Cyclin E binding to Cdk2 and independently of its phosphorylation at three key residues at the well-characterized C-terminal phosphodegron of Cyclin E.

We found that p18-Cyclin E interacts stronger with the α and γ isoforms of Fbw7, which has also been reported for Cyclin E (van Drogen et al., 2006). It should be noted that there is a possibility that when p18-Cyclin E is generated in cells following cleavage of

Cyclin E during apoptosis, it may be phosphorylated at this C-terminal phosphodegron, whereas these phosphorylation events do not take place in cells stably expressing p18-

Cyclin E. The observed difference in turnover efficiency by Cul1 suggests a different regulatory mechanism for p18-Cyclin E than that for full length Cyclin E, which may involve a parallel pathway independent of Cul1 and Cul3.

We have recently identified a specific p18-Cyclin E-interacting protein, Ku70, known as a critical component of the Non Homologous End-joining (NHEJ) DNA repair 139

pathway (Mazumder et al., 2007b). This unexpected interaction raised the possibility of

p18-Cyclin E being targeted to degradation by a pathway involving Ku70. Indeed,

overexpression of Ku70 led to a decrease in p18-Cyclin E levels while knock-down of

Ku70 by siRNA stabilizes the protein. Furthermore, we have identified Ku70 in a

complex with the ubiquitin ligase Hdm2. Taken together, these data would suggest a

novel degradation mechanism for p18-Cyclin E involving its binding to Ku70 and Hdm2,

the exact role of which and its characterization still need to be determined. It is possible that Ku70 acts as a scaffold bringing Hdm2 and p18-Cyclin E together, thus facilitating its ubiquitination and degradation. This may suggest a two-step degradation process of

Cyclin E to achieve its accelerated turnover. These observations provide new insights into the regulation of Cyclin E as well as on proteasome-mediated degradation pathways that may affect other cellular proteins.

The ubiquitin-proteasome pathway is the major non-lysosomal proteolytic system

in the cytosol and nucleus of all eukaryotic cells. As most of the cell death-regulatory

proteins are targets of the ubiquitin-proteasomal pathway, proteasome inhibitors have

emerged as an attractive target for human cancer therapy (Aghajanian et al., 2002;

Chauhan et al., 2005). Bortezomib (PS-341) and MG132 are highly specific proteasome

inhibitors that induce apoptosis in a variety of tumor cell lines. Proteasome inhibitors have been demonstrated to overcome chemoresistance of tumor cells by increasing chemosensitivity and also by working in synergy with other agents to induce apoptotic cell death of tumor cells (Hayden et al., 2007; Russo et al., 2007). However, their specific targets and the intimate mechanism responsible for inducing cell death are unclear. 140

Bortezomib was the first small molecule proteasome inhibitor (Fig 3.7) that has

shown antitumoral activity in a number of cell types and is currently undergoing clinical trials for hematopoietic as well as solid tumors (Bross et al., 2004; Hideshima et al.,

2003; Papandreou et al., 2004; Richardson et al., 2003). It has already been approved by

the FDA for the treatment of multiple myeloma and mantle cell lymphoma in patients

who have received at least one prior therapy and who indicated disease progression.

Bortezomib alone or in combination with other chemotherapeutic agents represents a new attractive therapeutic approach and is currently investigated in clinical trials for various malignancies, such as multiple myeloma, chronic lymphocytic leukemia (CLL) (Pahler et al., 2003), acute lymphocytic leukemia, follicular lymphoma, non-Hodgkin’s lymphoma, myelodysplastic syndrome, non-small cell lung carcinoma, and head and neck cancer.

MG132 is a reversible specific and potent inhibitor of the 26S proteasome that has been

shown to induce cell death in many malignancies (Drexler, 1997).

141

Fig 3.7 Chemical structure of the proteasome inhibitor, bortezomib

HO OH B

N NH

H N O N

O

142

We have demonstrated recently that by binding to Ku70, p18-Cyclin E causes the

dissociation of Bax from Ku70 and its activation, thus leading to apoptosis (Mazumder et

al., 2007b). Therefore, p18-Cyclin E may be an attractive target for cancer therapy.

However, the scope of its clinical use is limited due to its labile nature and therefore

enhancing its stability may potentiate its effect. In this study we show that as a

consequence of proteasome inhibition with Bortezomib, p18-Cyclin E is stabilized and its

association with Ku70 increased leading to enhanced apoptosis. Previously, we have

reported that p18-Cyclin E-induced cell death is due to the interaction of Ku70 with p18-

Cyclin E, which influences the interaction of Bax with Ku70 and thereby, activation of

Bax. In this context, the association of p18-Cyclin E with Ku70 is also enhanced by

Bortezomib treatment, most probably due to the increased levels of p18-Cyclin E and

counteracting the destabilizing effect of Ku70 on p18-Cyclin E. Importantly, this

interaction leads to release of Bax followed by its activation, suggesting that increased

cell death is directly related to the binding of p18-Cyclin E to Ku70. We have determined

that cells expressing p18-Cyclin E are more sensitive to cell death induction when treated

with Bortezomib, as compared to the parental cells, which is most probably due to

inhibition of proteasomal degradation of p18-Cyclin E. Moreover, the combination of

Bortezomib with a DNA-damaging agent such as VP-16 is much more effective in

stabilizing p18-Cyclin E as well as sensitization of the cells (data not shown).

We have reported earlier that following treatment with DNA-damaging agents such as irradiation and VP16, p18-Cyclin E is generated through proteolytic cleavage of

Cyclin E (Mazumder et al., 2002; Mazumder et al., 2007b). Interestingly, Bortezomib 143

treatment also generates endogenous p18-Cyclin E in hematopoietic IM-9 cells at a

significantly higher level compared to treatment with irradiation alone (data not shown).

Furthermore, Bortezomib treatment sensitized cells much more compared to the radiation treatment. Most likely, the increased cell death in case of Bortezomib treated cells is due to stabilization leading to its increased p18-Cyclin E levels. Although p18-Cyclin E is not generated in all cell types, once expressed, it can induce apoptosis in a wide variety of cellular systems that we have tested. These findings support the development of p18-

Cyclin E as a novel therapeutical approach for the treatment of cancer.

Although the exact mechanism of how proteasome inhibition causes apoptosis is

not fully understood, the fact that malignant cells are much more sensitive to the death-

promoting aspects of proteasome inhibition than normal cells (Hayden et al., 2007)

makes the ubiquitin/26S proteasome system a target for cancer therapy. Bortezomib is the

first proteasome inhibitor that has become available for clinical use. The excellent results

achieved in patients with multiple myeloma suggest the use of Bortezomib in solid

cancers, especially in combination with chemotherapy or radiotherapy. Several phase I/II

studies have used Bortezomib as a mono therapy in advanced chemotherapy refractory

solid cancers but, knowledge of in-vivo effects of combined modality approaches using

Bortezomib is limited. In this study, we demonstrate that treatment of cancer cells with

the highly potent and selective proteasome inhibitors Bortezomib and MG132 triggers

activation of caspase and apoptosis by significant induction and stabilization of p18-

Cyclin E. Furthermore, by preventing its proteasomal degradation, p18-Cyclin E may 144

become an important Bortezomib target while an adjuvant for Bortezomib treatment of solid tumors and hematopoietic malignancies.

CHAPTER IV

Summary and future directions

Summary

Conventional therapies including ionizing radiation, alkylating agents, anti- metabolites, and different targeted inhibitors have tried to take advantage of the particular biology of the tumor cells. However, many patients develop resistance over time or show no favorable response at all. Therefore, a clear understanding of the biochemical processes specific to cancer cells or which lead to malignant transformation in order to exploit them for therapeutical purposes is of utmost importance.

Our studies have uncovered two new important roles of Cyclin E in the cellular response to genotoxic stress caused by DNA-damaging agents. We have initially reported that in hematopoietic cells undergoing apoptosis induced by DNA-damaging agents,

Cyclin E generates an 18-kDa carboxy-terminal fragment, p18-Cyclin E (Mazumder et al., 2002). Then we have shown that p18-Cyclin E plays an important role in regulating the apoptotic pathway in the cytosol by binding Ku70 and releasing Bax (Mazumder et al., 2007b). Apart from its role in modulating the apoptotic response, p18-Cyclin E impairs the non-homologous end joining (NHEJ) DNA double-strand break repair pathway (Chapter II). Therefore, we have shown that Cyclin E, traditionally believed to be involved only in cell cycle regulation, has two other non-canonical roles, in regulating

145 146

apoptosis and DNA repair (Figure 4.1). Furthermore, we have characterized the

biochemical processes responsible for the turnover of p18-Cyclin E and how these can be

taken advantage of to sensitize tumor cells to therapy (Chapter III). These novel findings

extend the role of Cyclin E beyond the cell cycle control to include regulation of

apoptosis as well as that of NHEJ DNA repair.

As therapeutic resistance continues to be an important obstacle in cancer

treatment, one must consider the diverse pathways that tumor cells may utilize in

acquiring such survival capacity. Future therapeutical approach strategies may rely on

using p18-Cyclin E or a peptidic derivative to sensitize tumor cells to conventional

ionizing radiation or drug therapies.

Understanding the regulation of these three cellular processes, cell cycle, DNA

repair, and apoptosis in cancer cells would facilitate the development of targeted therapies which would result in tumor cell killing while protecting the normal cells.

147

Figure 4.1 Dual regulation of apoptosis and DNA repair by p18-Cyclin E. Genotoxic stress caused by DNA-damaging agents leads to a caspase-3-mediated cleavage of Cyclin

E generating its C-terminal fragment, p18-Cyclin E. This fragment binds Ku70 in the cytosol, probably inducing a change in its conformation which results in release and activation of Bax from its inhibitory association with Ku70. Bax translocates to the mitochondria, thus amplifying the apoptotic signal. On the other hand, p18-Cyclin E interacts with Ku70 in the nucleus impairing the NHEJ DNA repair. Both effects of p18-

Cyclin E lead to an amplification of the cell death signal committing the cell to apoptotic death.

148

Future directions

p18-Cyclin E affects DNA damage signaling and slows down the kinetic of DNA

repair

Formation of foci of different nuclear proteins is a widely used method for

assessing DNA double-strand breaks (DSB) formation and its repair. Among these are

53BP1, Nbs1, Rad51, and BRCA1 (Paull et al., 2000), with γ-H2AX being the most frequently used. Histone H2AX is phosphorylated in response to DNA damage on Ser139

(Rogakou et al., 1998) forming a nuclear focus. γ-H2AX foci serve as flags on the

chromatin for further DNA repair. This molecule links global chromatin changes and

accessory proteins with DNA repair (Redon et al., 2002). Quantification of γ-H2AX foci

has become a standard in the detection and quantification of the extent of DNA damage

following IR (Wang et al., 2005).

A very conclusive evidence of the influence of p18-Cyclin E on DNA repair

would be provided by showing that p18-Cyclin E slows down the kinetics of DNA repair

in vivo. This would be monitored by observing the appearance and disappearance of γ-

H2AX foci, as a measure of DNA damage and its repair. p18-Cyclin E stably expressing

293T cells would be irradiated or treated with etoposide and the presence of γ-H2AX would be determined by immunofluorescence or flow cytometry. If p18-Cyclin prevents the DNA repair, a persistence of the γ-H2AX foci in these cells as compared to control cells would be expected. A drawback in using the 293T cell system for this type of experiment could be the fact that the DNA damage signaling is not completely functional 149

in these cells due to the inactivation of p53 by the large T antigen. A different cellular system (e.g. normal diploid fibroblasts, epithelial cells or PBMC) in which the DNA damage signaling pathway is intact and which would also stably express p18-Cyclin E might be more indicated.

p18-Cyclin E impairs the recruitment of the XLF/XRCC4/Ligase IV heterocomplex to the Ku70/Ku80 heterodimer

In Chapter II I have shown that p18-Cyclin E impairs NHEJ DNA repair by preventing the recruitment of XLF/XRCC4/Ligase IV to the DNA repair complex at the damage site. However, recent studies have shown that XRCC4/Ligase IV can be recruited directly to Ku70/Ku80 in the absence DNA-PKcs (Costantini et al., 2007). Determining the ability of p18-Cyclin E to prevent the recruitment of XLF/XRCC4/Ligase IV using the experimental conditions used by Costantini et al. would provide conclusive mechanistic evidence regarding the influence of p18-Cyclin E in the final step of NHEJ. p18-Cyclin E stably expressing cells would be lysed according to the description in

Costantini et al. and Ku70/Ku80 would be immunoprecipitated followed by determination of Ligase IV and XRCC4 levels by immunoblotting. A reduction in the association between Ku70/Ku80 and XLF/XRCC4/Ligase IV would be expected. This experiment would determine unequivocally the mechanism by which p18-Cyclin E prevents NHEJ DNA repair.

150

Determination of p18-Cyclin E subcellular localization and its implications

Immunofluorescence studies showed that while Cyclin E localization is mainly in

the nucleus, where it exercises its well known role in cell cycle control and DNA

replication, p18-Cyclin E is both nuclear and cytosolic. Intriguingly, although present throughout the cytosol, preliminary data indicate that p18-Cyclin E is concentrated in peri-nuclear clusters resembling the Golgi apparatus (Figure 4.2). In contrast, a Cyclin

ER130A mutant that does not bind Cdk2 due to intramolecular destabilization localizes

mainly to the cytosol.

To determine whether, indeed, p18-Cyclin E localizes to the Golgi apparatus,

confocal microscopy will be used to examine the co-localization of p18-Cyclin E with

Golgin 97 (a unique protein specific to the Golgi apparatus) or a transfected Yellow

Fluorescence Protein bearing a Golgi localization signal. Furthermore, dynamics of the

p18-Cyclin E subcellular localization will be examined by time lapse confocal

microscopy using a p18-Cyclin E-GFP construct.

151

Figure 4.2 Localization of p18-Cyclin E-GFP, Cyclin E-GFP, and Cyclin ER130A-

GFP. Nuclei are stained with DAPI (blue). Nuclear and cytosolic localization of Cyclin E and Cyclin ER130A respectively, versus the Golgi-like formation localization of p18-

Cyclin E (green). Arrows indicate p18-Cyclin E clusters.

152

Characterize the mechanism of generation of p18-Cyclin E

The mechanism by which p18-Cyclin E is generated would be of particular interest as we have witnessed its appearance following treatment with DNA-damaging agents only in cells of hematopoietic origin. We examined the generation of p18-Cyclin E from Cyclin E in the NCI-H1299 non-small cell lung carcinoma cell line. A HA-Cyclin E stably expressing H1299 cell line was generated by transfection followed by selection with neomycin. These cells were either irradiated or treated with etoposide.

Unfortunately, in either case, the appearance of HA-p18-Cyclin E was not observed. The same experiment should be repeated in a hematopoietic cell line such as IM-9, Molt-4, or

Nalm-6, as it appears that due to yet unexplained mechanisms, p18-Cyclin E is generated only in cells of hematopoietic origin.

Another alternative of studying the circumstances in which p18-Cyclin E is being generated would be in a murine system. However, ionizing radiation and etoposide treatment failed to result in the generation of p18-Cyclin E. A careful analysis of the

Cyclin E amino-acid sequence led to the conclusion that the amino-acid residue where caspase-3-mediated cleavage is supposed to take place is mutated (Figure 4.3). A Cyclin

E sequence alignment across human, mouse, rat, rabbit, chicken, and frog indicated that only in human the LDVD sequence with the Asp275 (or 292 according to a longer isoform) residue is present. In all other species, the Asp275 residue is changed to Gly or

Cys. This explains why the generation of p18-Cyclin E was observed only in human cell lines. 153

Figure 4.3 Cyclin E sequence alignment across human, mouse, rat, rabbit, chicken,

and frog. The blue wire box identifies the Cyclin E caspase-3 cleavage site, LDVD. Only in humans, Cyclin E maintains the Asp275 (NM_05172 short isoform) or Asp292

(NM_001238 long isoform) residue, other species having this residues mutated to glycine. The sequences Homo sapiens (NM_001238), Mus musculus (NM_007633),

Rattus norvegicus (NM_001100821), Oryctolagus cunniculus (EU137106, partial sequence), Gallus gallus (NM_001031358), Xenopus laevis (NM_001087976) were aligned using ClustalW.

154

155

Furthermore, to test whether p18-Cyclin E is generated from the free or Cdk2-

bound Cyclin E, three expression constructs would be used: wt Cyclin E-HA, Cyclin

ED275A-HA (cleavage resistant mutant), and Cyclin ER130A-HA that does not bind Cdk2 due to intramolecular destabilization. Even though Cyclin E is known to shuttle from the

nucleus to the cytosol in a Crm1-independent manner (Jackman et al., 2002), its main

localization is in the nucleus, where it is imported after being synthesized and assembled with Cdk2 in the cytoplasm. Consequently, Cyclin ER130A is not expected to be retained in the nucleus and will be found mainly in the cytoplasm as also shown by immunocytochemistry (Figure 4.2). To determine from which pool of Cyclin E p18-

Cyclin E is generated, IM-9 cells stably expressing HA-Cyclin E or HA-Cyclin ER130A

will be irradiated and the generated fragments will be identified with anti-HA antibody. If

following IR, Cyclin E generates p18-Cyclin E and Cyclin ER130A does not generate it, p18-Cyclin E is most likely generated from cleavage of Cdk2-bound Cyclin E. If both

Cyclin E and Cyclin ER130A are able to generate p18-Cyclin E then the Cdk2-free pool of

Cyclin E is responsible for generating p18-Cyclin E. As negative control, the Cyclin E cleavage resistant mutant that is effective in blocking apoptosis (Mazumder et al., 2002) will be used.

The fact that the role of p18-Cyclin E is to commit the cell for apoptotic death by amplifying the apoptotic signal as well as by impairing DNA repair, warrants its use as a biomarker in determining the sensitivity of hematopoietic tumor cells to different chemotherapeutic approaches.

156

Study the Ku70/Hdm2-mediated p18-Cyclin E degradation

The ubiquitin E3 ligase activity of Hdm2, its recent shown interaction with Ku70

(Gama et al., 2008, unpublished) corroborated with the association between p18-Cyclin E and Ku70 raised the question of whether Hdm2 contributes to the degradation of p18-

Cyclin E. To unequivocally prove that Hdm2 is responsible for the ubiquitination of p18-

Cyclin E, Hdm2 will be either overexpressed or down-regulated using the siRNA technology and levels of p18-Cyclin E will be determined by immunoblotting.

Furthermore, nutlin, an inhibitor of Hdm2 will be used to block its activity in order to assess its effect on p18-Cyclin E (Wang and El-Deiry, 2008).

REFERENCES

Abraham, R. T. (2004). PI 3-kinase related kinases: 'big' players in stress-induced signaling pathways. DNA Repair (Amst) 3, 883-887.

Adachi, N., Ishino, T., Ishii, Y., Takeda, S., and Koyama, H. (2001). DNA ligase IV- deficient cells are more resistant to ionizing radiation in the absence of Ku70: Implications for DNA double-strand break repair. Proc Natl Acad Sci U S A 98, 12109- 12113.

Adams, J. (2004). The proteasome: a suitable antineoplastic target. Nat Rev Cancer 4, 349-360.

Adams, J., and Kauffman, M. (2004). Development of the proteasome inhibitor Velcade (Bortezomib). Cancer Invest 22, 304-311.

Aghajanian, C., Soignet, S., Dizon, D. S., Pien, C. S., Adams, J., Elliott, P. J., Sabbatini, P., Miller, V., Hensley, M. L., Pezzulli, S., et al. (2002). A phase I trial of the novel proteasome inhibitor PS341 in advanced solid tumor malignancies. Clin Cancer Res 8, 2505-2511.

Ahnesorg, P., Smith, P., and Jackson, S. P. (2006). XLF interacts with the XRCC4-DNA ligase IV complex to promote DNA nonhomologous end-joining. Cell 124, 301-313.

Ait-Si-Ali, S., Ramirez, S., Barre, F. X., Dkhissi, F., Magnaghi-Jaulin, L., Girault, J. A., Robin, P., Knibiehler, M., Pritchard, L. L., Ducommun, B., et al. (1998). Histone acetyltransferase activity of CBP is controlled by cycle-dependent kinases and oncoprotein E1A. Nature 396, 184-186.

Akama, Y., Yasui, W., Yokozaki, H., Kuniyasu, H., Kitahara, K., Ishikawa, T., and Tahara, E. (1995). Frequent amplification of the cyclin E gene in human gastric carcinomas. Jpn J Cancer Res 86, 617-621.

Akli, S., and Keyomarsi, K. (2003). Cyclin E and its low molecular weight forms in human cancer and as targets for cancer therapy. Cancer Biol Ther 2, S38-47.

Akli, S., Zheng, P. J., Multani, A. S., Wingate, H. F., Pathak, S., Zhang, N., Tucker, S. L., Chang, S., and Keyomarsi, K. (2004). Tumor-specific low molecular weight forms of cyclin E induce genomic instability and resistance to p21, p27, and antiestrogens in breast cancer. Cancer Res 64, 3198-3208.

157 158

Aleem, E., Kiyokawa, H., and Kaldis, P. (2005). Cdc2-cyclin E complexes regulate the G1/S phase transition. Nat Cell Biol 7, 831-836.

Alevizopoulos, K., Vlach, J., Hennecke, S., and Amati, B. (1997). Cyclin E and c-Myc promote cell proliferation in the presence of p16INK4a and of hypophosphorylated retinoblastoma family proteins. Embo J 16, 5322-5333.

Almasan, A., Linke, S. P., Paulson, T. G., Huang, L. C., and Wahl, G. M. (1995a). Genetic instability as a consequence of inappropriate entry into and progression through S-phase. Cancer Metastasis Rev 14, 59-73.

Almasan, A., Yin, Y., Kelly, R. E., Lee, E. Y., Bradley, A., Li, W., Bertino, J. R., and Wahl, G. M. (1995b). Deficiency of retinoblastoma protein leads to inappropriate S- phase entry, activation of E2F-responsive genes, and apoptosis. Proc Natl Acad Sci U S A 92, 5436-5440.

Alnemri, E. S., Livingston, D. J., Nicholson, D. W., Salvesen, G., Thornberry, N. A., Wong, W. W., and Yuan, J. (1996). Human ICE/CED-3 protease nomenclature. Cell 87, 171.

Anderson, C. W. (1993). DNA damage and the DNA-activated protein kinase. Trends Biochem Sci 18, 433-437.

Andreassen, P. R., Lohez, O. D., and Margolis, R. L. (2003). G2 and spindle assembly checkpoint adaptation, and tetraploidy arrest: implications for intrinsic and chemically induced genomic instability. Mutat Res 532, 245-253.

Ang, X. L., and Wade Harper, J. (2005). SCF-mediated protein degradation and cell cycle control. Oncogene 24, 2860-2870.

Antonsson, B., Montessuit, S., Lauper, S., Eskes, R., and Martinou, J. C. (2000). Bax oligomerization is required for channel-forming activity in liposomes and to trigger cytochrome c release from mitochondria. Biochem J 345 Pt 2, 271-278.

Arends, M. J., and Wyllie, A. H. (1991). Apoptosis: mechanisms and roles in pathology. Int Rev Exp Pathol 32, 223-254.

Audebert, M., Salles, B., and Calsou, P. (2004). Involvement of poly(ADP-ribose) polymerase-1 and XRCC1/DNA ligase III in an alternative route for DNA double-strand breaks rejoining. J Biol Chem 279, 55117-55126.

Bacsi, A., Kannan, S., Lee, M. S., Hazra, T. K., and Boldogh, I. (2005). Modulation of DNA-dependent protein kinase activity in chlorambucil-treated cells. Free Radic Biol Med 39, 1650-1659. 159

Bai, C., Sen, P., Hofmann, K., Ma, L., Goebl, M., Harper, J. W., and Elledge, S. J. (1996). SKP1 connects cell cycle regulators to the ubiquitin proteolysis machinery through a novel motif, the F-box. Cell 86, 263-274.

Bales, E., Mills, L., Milam, N., McGahren-Murray, M., Bandyopadhyay, D., Chen, D., Reed, J. A., Timchenko, N., van den Oord, J. J., Bar-Eli, M., et al. (2005). The low molecular weight cyclin E isoforms augment angiogenesis and metastasis of human melanoma cells in vivo. Cancer Res 65, 692-697.

Barnes, D. E., Stamp, G., Rosewell, I., Denzel, A., and Lindahl, T. (1998). Targeted disruption of the gene encoding DNA ligase IV leads to lethality in embryonic mice. Curr Biol 8, 1395-1398.

Baserga, R. (1985). The Biology of Cell Reproduction (Cambridge, Harvard University Press).

Basu, S., Rosenzweig, K. R., Youmell, M., and Price, B. D. (1998). The DNA-dependent protein kinase participates in the activation of NF kappa B following DNA damage. Biochem Biophys Res Commun 247, 79-83.

Bebenek, K., Garcia-Diaz, M., Blanco, L., and Kunkel, T. A. (2003). The frameshift infidelity of human DNA polymerase lambda. Implications for function. J Biol Chem 278, 34685-34690.

Berthet, C., Aleem, E., Coppola, V., Tessarollo, L., and Kaldis, P. (2003). Cdk2 knockout mice are viable. Curr Biol 13, 1775-1785.

Blattner, C., Sparks, A., and Lane, D. (1999). Transcription factor E2F-1 is upregulated in response to DNA damage in a manner analogous to that of p53. Mol Cell Biol 19, 3704-3713.

Block, W. D., Yu, Y., Merkle, D., Gifford, J. L., Ding, Q., Meek, K., and Lees-Miller, S. P. (2004). Autophosphorylation-dependent remodeling of the DNA-dependent protein kinase catalytic subunit regulates ligation of DNA ends. Nucleic Acids Res 32, 4351- 4357.

Boatright, K. M., Renatus, M., Scott, F. L., Sperandio, S., Shin, H., Pedersen, I. M., Ricci, J. E., Edris, W. A., Sutherlin, D. P., Green, D. R., and Salvesen, G. S. (2003). A unified model for apical caspase activation. Mol Cell 11, 529-541.

Boldin, M. P., Varfolomeev, E. E., Pancer, Z., Mett, I. L., Camonis, J. H., and Wallach, D. (1995). A novel protein that interacts with the death domain of Fas/APO1 contains a sequence motif related to the death domain. J Biol Chem 270, 7795-7798. 160

Boldogh, I., Roy, G., Lee, M. S., Bacsi, A., Hazra, T. K., Bhakat, K. K., Das, G. C., and Mitra, S. (2003). Reduced DNA double strand breaks in chlorambucil resistant cells are related to high DNA-PKcs activity and low oxidative stress. Toxicology 193, 137-152.

Boonstra, J. (2003). Progression through the G1-phase of the on-going cell cycle. J Cell Biochem 90, 244-252.

Bortner, D. M., and Rosenberg, M. P. (1997). Induction of mammary gland hyperplasia and carcinomas in transgenic mice expressing human cyclin E. Mol Cell Biol 17, 453- 459.

Botz, J., Zerfass-Thome, K., Spitkovsky, D., Delius, H., Vogt, B., Eilers, M., Hatzigeorgiou, A., and Jansen-Durr, P. (1996). Cell cycle regulation of the murine cyclin E gene depends on an E2F binding site in the promoter. Mol Cell Biol 16, 3401-3409.

Braig, M., Lee, S., Loddenkemper, C., Rudolph, C., Peters, A. H., Schlegelberger, B., Stein, H., Dorken, B., Jenuwein, T., and Schmitt, C. A. (2005). Oncogene-induced senescence as an initial barrier in lymphoma development. Nature 436, 660-665.

Brem, R., and Hall, J. (2005). XRCC1 is required for DNA single-strand break repair in human cells. Nucleic Acids Res 33, 2512-2520.

Bross, P. F., Kane, R., Farrell, A. T., Abraham, S., Benson, K., Brower, M. E., Bradley, S., Gobburu, J. V., Goheer, A., Lee, S. L., et al. (2004). Approval summary for bortezomib for injection in the treatment of multiple myeloma. Clin Cancer Res 10, 3954-3964.

Bryans, M., Valenzano, M. C., and Stamato, T. D. (1999). Absence of DNA ligase IV protein in XR-1 cells: evidence for stabilization by XRCC4. Mutat Res 433, 53-58.

Buck, D., Malivert, L., de Chasseval, R., Barraud, A., Fondaneche, M. C., Sanal, O., Plebani, A., Stephan, J. L., Hufnagel, M., le Deist, F., et al. (2006). Cernunnos, a novel nonhomologous end-joining factor, is mutated in human immunodeficiency with microcephaly. Cell 124, 287-299.

Bunn, P. A., Jr. (2004). The potential role of proteasome inhibitors in the treatment of lung cancer. Clin Cancer Res 10, 4263s-4265s.

Burma, S., Chen, B. P., Murphy, M., Kurimasa, A., and Chen, D. J. (2001). ATM phosphorylates histone H2AX in response to DNA double-strand breaks. J Biol Chem 276, 42462-42467.

Cain, K., Bratton, S. B., and Cohen, G. M. (2002). The Apaf-1 apoptosome: a large caspase-activating complex. Biochimie 84, 203-214. 161

Calhoun, E. S., Jones, J. B., Ashfaq, R., Adsay, V., Baker, S. J., Valentine, V., Hempen, P. M., Hilgers, W., Yeo, C. J., Hruban, R. H., and Kern, S. E. (2003). BRAF and FBXW7 (CDC4, FBW7, AGO, SEL10) mutations in distinct subsets of pancreatic cancer: potential therapeutic targets. Am J Pathol 163, 1255-1260.

Calsou, P., Delteil, C., Frit, P., Drouet, J., and Salles, B. (2003). Coordinated assembly of Ku and p460 subunits of the DNA-dependent protein kinase on DNA ends is necessary for XRCC4-ligase IV recruitment. J Mol Biol 326, 93-103.

Cande, C., Cohen, I., Daugas, E., Ravagnan, L., Larochette, N., Zamzami, N., and Kroemer, G. (2002). Apoptosis-inducing factor (AIF): a novel caspase-independent death effector released from mitochondria. Biochimie 84, 215-222.

Capp, J. P., Boudsocq, F., Bertrand, P., Laroche-Clary, A., Pourquier, P., Lopez, B. S., Cazaux, C., Hoffmann, J. S., and Canitrot, Y. (2006). The DNA polymerase lambda is required for the repair of non-compatible DNA double strand breaks by NHEJ in mammalian cells. Nucleic Acids Res 34, 2998-3007.

Carroll, P. E., Okuda, M., Horn, H. F., Biddinger, P., Stambrook, P. J., Gleich, L. L., Li, Y. Q., Tarapore, P., and Fukasawa, K. (1999). Centrosome hyperamplification in human cancer: chromosome instability induced by p53 mutation and/or overexpression. Oncogene 18, 1935-1944.

Casciola-Rosen, L. A., Anhalt, G. J., and Rosen, A. (1995). DNA-dependent protein kinase is one of a subset of autoantigens specifically cleaved early during apoptosis. J Exp Med 182, 1625-1634.

Chan, D. W., and Lees-Miller, S. P. (1996). The DNA-dependent protein kinase is inactivated by autophosphorylation of the catalytic subunit. J Biol Chem 271, 8936-8941.

Chance, B., Sies, H., and Boveris, A. (1979). Hydroperoxide metabolism in mammalian organs. Physiol Rev 59, 527-605.

Chappell, C., Hanakahi, L. A., Karimi-Busheri, F., Weinfeld, M., and West, S. C. (2002). Involvement of human polynucleotide kinase in double-strand break repair by non- homologous end joining. Embo J 21, 2827-2832.

Chauhan, D., Hideshima, T., Mitsiades, C., Richardson, P., and Anderson, K. C. (2005). Proteasome inhibitor therapy in multiple myeloma. Mol Cancer Ther 4, 686-692.

Chellappan, S. P., Hiebert, S., Mudryj, M., Horowitz, J. M., and Nevins, J. R. (1991). The E2F transcription factor is a cellular target for the RB protein. Cell 65, 1053-1061.

Chen, B. P., Chan, D. W., Kobayashi, J., Burma, S., Asaithamby, A., Morotomi-Yano, K., Botvinick, E., Qin, J., and Chen, D. J. (2005a). Cell cycle dependence of DNA- 162

dependent protein kinase phosphorylation in response to DNA double strand breaks. J Biol Chem 280, 14709-14715.

Chen, L., Trujillo, K., Sung, P., and Tomkinson, A. E. (2000a). Interactions of the DNA ligase IV-XRCC4 complex with DNA ends and the DNA-dependent protein kinase. J Biol Chem 275, 26196-26205.

Chen, Q., Gong, B., and Almasan, A. (2000b). Distinct stages of cytochrome c release from mitochondria: evidence for a feedback amplification loop linking caspase activation to mitochondrial dysfunction in genotoxic stress induced apoptosis. Cell Death Differ 7, 227-233.

Chen, Q., Gong, B., Mahmoud-Ahmed, A. S., Zhou, A., Hsi, E. D., Hussein, M., and Almasan, A. (2001). Apo2L/TRAIL and Bcl-2-related proteins regulate type I interferon- induced apoptosis in multiple myeloma. Blood 98, 2183-2192.

Chen, W., Lee, J., Cho, S. Y., and Fine, H. A. (2004). Proteasome-mediated destruction of the cyclin a/cyclin-dependent kinase 2 complex suppresses tumor cell growth in vitro and in vivo. Cancer Res 64, 3949-3957.

Chen, Y. N., Sharma, S. K., Ramsey, T. M., Jiang, L., Martin, M. S., Baker, K., Adams, P. D., Bair, K. W., and Kaelin, W. G., Jr. (1999). Selective killing of transformed cells by cyclin/cyclin-dependent kinase 2 antagonists. Proc Natl Acad Sci U S A 96, 4325-4329.

Chen, Z., Trotman, L. C., Shaffer, D., Lin, H. K., Dotan, Z. A., Niki, M., Koutcher, J. A., Scher, H. I., Ludwig, T., Gerald, W., et al. (2005b). Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis. Nature 436, 725-730.

Cheng, A., Kaldis, P., and Solomon, M. J. (2000). Dephosphorylation of human cyclin- dependent kinases by protein phosphatase type 2C alpha and beta 2 isoforms. J Biol Chem 275, 34744-34749.

Classon, M., and Harlow, E. (2002). The retinoblastoma tumour suppressor in development and cancer. Nat Rev Cancer 2, 910-917.

Clurman, B. E., Sheaff, R. J., Thress, K., Groudine, M., and Roberts, J. M. (1996). Turnover of cyclin E by the ubiquitin-proteasome pathway is regulated by cdk2 binding and cyclin phosphorylation. Genes Dev 10, 1979-1990.

Cohen, H. Y., Lavu, S., Bitterman, K. J., Hekking, B., Imahiyerobo, T. A., Miller, C., Frye, R., Ploegh, H., Kessler, B. M., and Sinclair, D. A. (2004). Acetylation of the C terminus of Ku70 by CBP and PCAF controls Bax-mediated apoptosis. Mol Cell 13, 627- 638. 163

Collado, M., Gil, J., Efeyan, A., Guerra, C., Schuhmacher, A. J., Barradas, M., Benguria, A., Zaballos, A., Flores, J. M., Barbacid, M., et al. (2005). Tumour biology: senescence in premalignant tumours. Nature 436, 642.

Collis, S. J., and DeWeese, T. L. (2004). Enhanced radiation response through directed molecular targeting approaches. Cancer Metastasis Rev 23, 277-292.

Convery, E., Shin, E. K., Ding, Q., Wang, W., Douglas, P., Davis, L. S., Nickoloff, J. A., Lees-Miller, S. P., and Meek, K. (2005). Inhibition of homologous recombination by variants of the catalytic subunit of the DNA-dependent protein kinase (DNA-PKcs). Proc Natl Acad Sci U S A 102, 1345-1350.

Cooray, P., Yuan, Y., Schoenwaelder, S. M., Mitchell, C. A., Salem, H. H., and Jackson, S. P. (1996). Focal adhesion kinase (pp125FAK) cleavage and regulation by calpain. Biochem J 318 ( Pt 1), 41-47.

Costantini, S., Woodbine, L., Andreoli, L., Jeggo, P. A., and Vindigni, A. (2007). Interaction of the Ku heterodimer with the DNA ligase IV/Xrcc4 complex and its regulation by DNA-PK. DNA Repair (Amst) 6, 712-722.

Critchlow, S. E., Bowater, R. P., and Jackson, S. P. (1997). Mammalian DNA double- strand break repair protein XRCC4 interacts with DNA ligase IV. Curr Biol 7, 588-598.

Critchlow, S. E., and Jackson, S. P. (1998). DNA end-joining: from yeast to man. Trends Biochem Sci 23, 394-398.

Cullinan, S. B., Gordan, J. D., Jin, J., Harper, J. W., and Diehl, J. A. (2004). The Keap1- BTB protein is an adaptor that bridges Nrf2 to a Cul3-based E3 ligase: oxidative stress sensing by a Cul3-Keap1 ligase. Mol Cell Biol 24, 8477-8486.

D'Urso, G., Marraccino, R. L., Marshak, D. R., and Roberts, J. M. (1990). Cell cycle control of DNA replication by a homologue from human cells of the p34cdc2 protein kinase. Science 250, 786-791.

Dai, Y., and Grant, S. (2003). Cyclin-dependent kinase inhibitors. Curr Opin Pharmacol 3, 362-370.

Danial, N. N., and Korsmeyer, S. J. (2004). Cell death: critical control points. Cell 116, 205-219.

Davies, T. G., Bentley, J., Arris, C. E., Boyle, F. T., Curtin, N. J., Endicott, J. A., Gibson, A. E., Golding, B. T., Griffin, R. J., Hardcastle, I. R., et al. (2002a). Structure-based design of a potent purine-based cyclin-dependent kinase inhibitor. Nat Struct Biol 9, 745- 749. 164

Davies, T. G., Pratt, D. J., Endicott, J. A., Johnson, L. N., and Noble, M. E. (2002b). Structure-based design of cyclin-dependent kinase inhibitors. Pharmacol Ther 93, 125- 133.

De Bont, R., and van Larebeke, N. (2004). Endogenous DNA damage in humans: a review of quantitative data. Mutagenesis 19, 169-185.

Dealy, M. J., Nguyen, K. V., Lo, J., Gstaiger, M., Krek, W., Elson, D., Arbeit, J., Kipreos, E. T., and Johnson, R. S. (1999). Loss of Cul1 results in early embryonic lethality and dysregulation of cyclin E. Nat Genet 23, 245-248.

DeFazio, L. G., Stansel, R. M., Griffith, J. D., and Chu, G. (2002). Synapsis of DNA ends by DNA-dependent protein kinase. Embo J 21, 3192-3200.

Denault, J. B., and Salvesen, G. S. (2002). Caspases: keys in the ignition of cell death. Chem Rev 102, 4489-4500.

Desagher, S., Osen-Sand, A., Nichols, A., Eskes, R., Montessuit, S., Lauper, S., Maundrell, K., Antonsson, B., and Martinou, J. C. (1999). Bid-induced conformational change of Bax is responsible for mitochondrial cytochrome c release during apoptosis. J Cell Biol 144, 891-901.

Deshaies, R. J. (1999). SCF and Cullin/Ring H2-based ubiquitin ligases. Annu Rev Cell Dev Biol 15, 435-467.

Deshpande, R. A., and Wilson, T. E. (2007). Modes of interaction among yeast Nej1, Lif1 and Dnl4 proteins and comparison to human XLF, XRCC4 and Lig4. DNA Repair (Amst) 6, 1507-1516.

DiBiase, S. J., Zeng, Z. C., Chen, R., Hyslop, T., Curran, W. J., Jr., and Iliakis, G. (2000). DNA-dependent protein kinase stimulates an independently active, nonhomologous, end- joining apparatus. Cancer Res 60, 1245-1253.

Donepudi, M., Mac Sweeney, A., Briand, C., and Grutter, M. G. (2003). Insights into the regulatory mechanism for caspase-8 activation. Mol Cell 11, 543-549.

Dong, Y., Sui, L., Tai, Y., Sugimoto, K., Hirao, T., and Tokuda, M. (2000). Prognostic significance of cyclin E overexpression in laryngeal squamous cell carcinomas. Clin Cancer Res 6, 4253-4258.

Donnellan, R., and Chetty, R. (1999). Cyclin E in human cancers. Faseb J 13, 773-780.

Donzelli, M., and Draetta, G. F. (2003). Regulating mammalian checkpoints through inactivation. EMBO Rep 4, 671-677. 165

Douglas, P., Moorhead, G. B., Ye, R., and Lees-Miller, S. P. (2001). Protein phosphatases regulate DNA-dependent protein kinase activity. J Biol Chem 276, 18992- 18998.

Dowdy, S. F., Hinds, P. W., Louie, K., Reed, S. I., Arnold, A., and Weinberg, R. A. (1993). Physical interaction of the retinoblastoma protein with human D cyclins. Cell 73, 499-511.

Drexler, H. C. (1997). Activation of the cell death program by inhibition of proteasome function. Proc Natl Acad Sci U S A 94, 855-860.

Du, C., Fang, M., Li, Y., Li, L., and Wang, X. (2000). Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell 102, 33-42.

Dulic, V., Lees, E., and Reed, S. I. (1992). Association of human cyclin E with a periodic G1-S phase protein kinase. Science 257, 1958-1961.

Duronio, R. J., Brook, A., Dyson, N., and O'Farrell, P. H. (1996). E2F-induced S phase requires cyclin E. Genes Dev 10, 2505-2513.

Dyson, N. (1998). The regulation of E2F by pRB-family proteins. Genes Dev 12, 2245- 2262.

Earnshaw, W. C., Martins, L. M., and Kaufmann, S. H. (1999). Mammalian caspases: structure, activation, substrates, and functions during apoptosis. Annu Rev Biochem 68, 383-424.

Ekholm-Reed, S., Spruck, C. H., Sangfelt, O., van Drogen, F., Mueller-Holzner, E., Widschwendter, M., Zetterberg, A., and Reed, S. I. (2004). Mutation of hCDC4 leads to cell cycle deregulation of cyclin E in cancer. Cancer Res 64, 795-800.

Ekholm, S. V., Zickert, P., Reed, S. I., and Zetterberg, A. (2001). Accumulation of cyclin E is not a prerequisite for passage through the restriction point. Mol Cell Biol 21, 3256- 3265.

Elledge, S. J. (1996). Cell cycle checkpoints: preventing an identity crisis. Science 274, 1664-1672.

Elliott, B., Richardson, C., Winderbaum, J., Nickoloff, J. A., and Jasin, M. (1998). Gene conversion tracts from double-strand break repair in mammalian cells. Mol Cell Biol 18, 93-101. 166

Enari, M., Sakahira, H., Yokoyama, H., Okawa, K., Iwamatsu, A., and Nagata, S. (1998). A caspase-activated DNase that degrades DNA during apoptosis, and its inhibitor ICAD. Nature 391, 43-50.

Erlandsson, F., Wahlby, C., Ekholm-Reed, S., Hellstrom, A. C., Bengtsson, E., and Zetterberg, A. (2003). Abnormal expression pattern of cyclin E in tumour cells. Int J Cancer 104, 369-375.

Erlanson, M., and Landberg, G. (2001). Prognostic implications of p27 and cyclin E protein contents in malignant lymphomas. Leuk Lymphoma 40, 461-470.

Erlanson, M., Portin, C., Linderholm, B., Lindh, J., Roos, G., and Landberg, G. (1998). Expression of cyclin E and the cyclin-dependent kinase inhibitor p27 in malignant lymphomas-prognostic implications. Blood 92, 770-777.

Evan, G. I., and Vousden, K. H. (2001). Proliferation, cell cycle and apoptosis in cancer. Nature 411, 342-348.

Evans, T., Rosenthal, E. T., Youngblom, J., Distel, D., and Hunt, T. (1983). Cyclin: a protein specified by maternal mRNA in sea urchin eggs that is destroyed at each cleavage division. Cell 33, 389-396.

Ewen, M. E. (2000). Where the cell cycle and histones meet. Genes Dev 14, 2265-2270.

Facchinetti, M. M., De Siervi, A., Toskos, D., and Senderowicz, A. M. (2004). UCN-01- induced cell cycle arrest requires the transcriptional induction of p21(waf1/cip1) by activation of mitogen-activated protein/extracellular signal-regulated kinase kinase/extracellular signal-regulated kinase pathway. Cancer Res 64, 3629-3637.

Fadeel, B., and Orrenius, S. (2005). Apoptosis: a basic biological phenomenon with wide-ranging implications in human disease. J Intern Med 258, 479-517.

Falck, J., Mailand, N., Syljuasen, R. G., Bartek, J., and Lukas, J. (2001). The ATM- Chk2-Cdc25A checkpoint pathway guards against radioresistant DNA synthesis. Nature 410, 842-847.

Ferguson, D. O., and Alt, F. W. (2001). DNA double strand break repair and chromosomal translocation: lessons from animal models. Oncogene 20, 5572-5579.

Foster, R. E., Nnakwe, C., Woo, L., and Frank, K. M. (2006). Monoubiquitination of the nonhomologous end joining protein XRCC4. Biochem Biophys Res Commun 341, 175- 183. 167

Franco, S. J., Rodgers, M. A., Perrin, B. J., Han, J., Bennin, D. A., Critchley, D. R., and Huttenlocher, A. (2004). Calpain-mediated proteolysis of talin regulates adhesion dynamics. Nat Cell Biol 6, 977-983.

Frank, K. M., Sharpless, N. E., Gao, Y., Sekiguchi, J. M., Ferguson, D. O., Zhu, C., Manis, J. P., Horner, J., DePinho, R. A., and Alt, F. W. (2000). DNA ligase IV deficiency in mice leads to defective neurogenesis and embryonic lethality via the p53 pathway. Mol Cell 5, 993-1002.

Fugmann, S. D., Lee, A. I., Shockett, P. E., Villey, I. J., and Schatz, D. G. (2000). The RAG proteins and V(D)J recombination: complexes, ends, and transposition. Annu Rev Immunol 18, 495-527.

Gali-Muhtasib, H., and Bakkar, N. (2002). Modulating cell cycle: current applications and prospects for future drug development. Curr Cancer Drug Targets 2, 309-336.

Gama, V., Yoshida, T., Gomez, J. A., Basile, D. P., Mayo, L. D., Haas, A. L., and Matsuyama, S. (2006). Involvement of the ubiquitin pathway in decreasing Ku70 levels in response to drug-induced apoptosis. Exp Cell Res 312, 488-499.

Gao, Y., Sun, Y., Frank, K. M., Dikkes, P., Fujiwara, Y., Seidl, K. J., Sekiguchi, J. M., Rathbun, G. A., Swat, W., Wang, J., et al. (1998). A critical role for DNA end-joining proteins in both lymphogenesis and neurogenesis. Cell 95, 891-902.

Geisen, C., and Moroy, T. (2002). The oncogenic activity of cyclin E is not confined to Cdk2 activation alone but relies on several other, distinct functions of the protein. J Biol Chem 277, 39909-39918.

Gellert, M. (2002). V(D)J recombination: RAG proteins, repair factors, and regulation. Annu Rev Biochem 71, 101-132.

Geng, Y., Eaton, E. N., Picon, M., Roberts, J. M., Lundberg, A. S., Gifford, A., Sardet, C., and Weinberg, R. A. (1996). Regulation of cyclin E transcription by E2Fs and retinoblastoma protein. Oncogene 12, 1173-1180.

Geng, Y., Lee, Y. M., Welcker, M., Swanger, J., Zagozdzon, A., Winer, J. D., Roberts, J. M., Kaldis, P., Clurman, B. E., and Sicinski, P. (2007). Kinase-independent function of cyclin E. Mol Cell 25, 127-139.

Geng, Y., Whoriskey, W., Park, M. Y., Bronson, R. T., Medema, R. H., Li, T., Weinberg, R. A., and Sicinski, P. (1999). Rescue of cyclin D1 deficiency by knockin cyclin E. Cell 97, 767-777. 168

Geng, Y., Yu, Q., Sicinska, E., Das, M., Schneider, J. E., Bhattacharya, S., Rideout, W. M., Bronson, R. T., Gardner, H., and Sicinski, P. (2003). Cyclin E ablation in the mouse. Cell 114, 431-443.

Glotzer, M. (1995). Cell cycle. The only way out of mitosis. Curr Biol 5, 970-972.

Gomez, J. A., Gama, V., Yoshida, T., Sun, W., Hayes, P., Leskov, K., Boothman, D., and Matsuyama, S. (2007). Bax-inhibiting peptides derived from Ku70 and cell-penetrating pentapeptides. Biochem Soc Trans 35, 797-801.

Gong, B., and Almasan, A. (2000). Apo2 ligand/TNF-related apoptosis-inducing ligand and death receptor 5 mediate the apoptotic signaling induced by ionizing radiation in leukemic cells. Cancer Res 60, 5754-5760.

Gong, B., Chen, Q., Endlich, B., Mazumder, S., and Almasan, A. (1999). Ionizing radiation-induced, Bax-mediated cell death is dependent on activation of cysteine and serine proteases. Cell Growth Differ 10, 491-502.

Goodarzi, A. A., Yu, Y., Riballo, E., Douglas, P., Walker, S. A., Ye, R., Harer, C., Marchetti, C., Morrice, N., Jeggo, P. A., and Lees-Miller, S. P. (2006). DNA-PK autophosphorylation facilitates Artemis endonuclease activity. Embo J 25, 3880-3889.

Graham, K. L., Gustin, K. E., Rivera, C., Kuyumcu-Martinez, N. M., Choe, S. S., Lloyd, R. E., Sarnow, P., and Utz, P. J. (2004). Proteolytic cleavage of the catalytic subunit of DNA-dependent protein kinase during poliovirus infection. J Virol 78, 6313-6321.

Grawunder, U., Wilm, M., Wu, X., Kulesza, P., Wilson, T. E., Mann, M., and Lieber, M. R. (1997). Activity of DNA ligase IV stimulated by complex formation with XRCC4 protein in mammalian cells. Nature 388, 492-495.

Grawunder, U., Zimmer, D., Kulesza, P., and Lieber, M. R. (1998). Requirement for an interaction of XRCC4 with DNA ligase IV for wild-type V(D)J recombination and DNA double-strand break repair in vivo. J Biol Chem 273, 24708-24714.

Green, D. R., and Evan, G. I. (2002). A matter of life and death. Cancer Cell 1, 19-30.

Griffiths, G. J., Dubrez, L., Morgan, C. P., Jones, N. A., Whitehouse, J., Corfe, B. M., Dive, C., and Hickman, J. A. (1999). Cell damage-induced conformational changes of the pro-apoptotic protein Bak in vivo precede the onset of apoptosis. J Cell Biol 144, 903- 914.

Gu, J., Lu, H., Tsai, A. G., Schwarz, K., and Lieber, M. R. (2007). Single-stranded DNA ligation and XLF-stimulated incompatible DNA end ligation by the XRCC4-DNA ligase IV complex: influence of terminal DNA sequence. Nucleic Acids Res 35, 5755-5762. 169

Gudas, J. M., Payton, M., Thukral, S., Chen, E., Bass, M., Robinson, M. O., and Coats, S. (1999). Cyclin E2, a novel G1 cyclin that binds Cdk2 and is aberrantly expressed in human cancers. Mol Cell Biol 19, 612-622.

Gullo, C., Au, M., Feng, G., and Teoh, G. (2006). The biology of Ku and its potential oncogenic role in cancer. Biochim Biophys Acta 1765, 223-234.

Gupta-Rossi, N., Le Bail, O., Gonen, H., Brou, C., Logeat, F., Six, E., Ciechanover, A., and Israel, A. (2001). Functional interaction between SEL-10, an F-box protein, and the nuclear form of activated Notch1 receptor. J Biol Chem 276, 34371-34378.

Haber, J. E. (1995). In vivo biochemistry: physical monitoring of recombination induced by site-specific endonucleases. Bioessays 17, 609-620.

Hartwell, L. H., and Weinert, T. A. (1989). Checkpoints: controls that ensure the order of cell cycle events. Science 246, 629-634.

Haupt, Y., Maya, R., Kazaz, A., and Oren, M. (1997). Mdm2 promotes the rapid degradation of p53. Nature 387, 296-299.

Hayden, P. J., Mitsiades, C. S., Anderson, K. C., and Richardson, P. G. (2007). Novel therapies in myeloma. Curr Opin Hematol 14, 609-615.

He, F., Li, L., Kim, D., Wen, B., Deng, X., Gutin, P. H., Ling, C. C., and Li, G. C. (2007). Adenovirus-mediated expression of a dominant negative Ku70 fragment radiosensitizes human tumor cells under aerobic and hypoxic conditions. Cancer Res 67, 634-642.

Hedberg, Y., Davoodi, E., Ljungberg, B., Roos, G., and Landberg, G. (2002). Cyclin E and p27 protein content in human renal cell carcinoma: clinical outcome and associations with cyclin D. Int J Cancer 102, 601-607.

Hegde, R., Srinivasula, S. M., Zhang, Z., Wassell, R., Mukattash, R., Cilenti, L., DuBois, G., Lazebnik, Y., Zervos, A. S., Fernandes-Alnemri, T., and Alnemri, E. S. (2002). Identification of Omi/HtrA2 as a mitochondrial apoptotic serine protease that disrupts inhibitor of apoptosis protein-caspase interaction. J Biol Chem 277, 432-438.

Hengartner, M. O. (2000). The biochemistry of apoptosis. Nature 407, 770-776.

Herrera, R. E., Chen, F., and Weinberg, R. A. (1996). Increased histone H1 phosphorylation and relaxed chromatin structure in Rb-deficient fibroblasts. Proc Natl Acad Sci U S A 93, 11510-11515. 170

Herrmann, J. L., Briones, F., Jr., Brisbay, S., Logothetis, C. J., and McDonnell, T. J. (1998). Prostate carcinoma cell death resulting from inhibition of proteasome activity is independent of functional Bcl-2 and p53. Oncogene 17, 2889-2899.

Hershko, A., and Ciechanover, A. (1998). The ubiquitin system. Annu Rev Biochem 67, 425-479.

Hideshima, T., Mitsiades, C., Akiyama, M., Hayashi, T., Chauhan, D., Richardson, P., Schlossman, R., Podar, K., Munshi, N. C., Mitsiades, N., and Anderson, K. C. (2003). Molecular mechanisms mediating antimyeloma activity of proteasome inhibitor PS-341. Blood 101, 1530-1534. Epub 2002 Sep 1526.

Hoeijmakers, J. H. (2001). Genome maintenance mechanisms for preventing cancer. Nature 411, 366-374.

Hoffmann, I., Draetta, G., and Karsenti, E. (1994). Activation of the phosphatase activity of human cdc25A by a cdk2-cyclin E dependent phosphorylation at the G1/S transition. Embo J 13, 4302-4310.

Hsu, H. L., Yannone, S. M., and Chen, D. J. (2002). Defining interactions between DNA- PK and ligase IV/XRCC4. DNA Repair (Amst) 1, 225-235.

Hsu, Y. T., Wolter, K. G., and Youle, R. J. (1997). Cytosol-to-membrane redistribution of Bax and Bcl-X(L) during apoptosis. Proc Natl Acad Sci U S A 94, 3668-3672.

Hsu, Y. T., and Youle, R. J. (1998). Bax in murine thymus is a soluble monomeric protein that displays differential detergent-induced conformations. J Biol Chem 273, 10777-10783.

Hua, X. H., Yan, H., and Newport, J. (1997). A role for Cdk2 kinase in negatively regulating DNA replication during S phase of the cell cycle. J Cell Biol 137, 183-192.

Hubalek, M. M., Widschwendter, A., Erdel, M., Gschwendtner, A., Fiegl, H. M., Muller, H. M., Goebel, G., Mueller-Holzner, E., Marth, C., Spruck, C. H., et al. (2004). Cyclin E dysregulation and chromosomal instability in endometrial cancer. Oncogene 23, 4187- 4192.

Hunter, T., and Pines, J. (1994). Cyclins and cancer. II: Cyclin D and CDK inhibitors come of age. Cell 79, 573-582.

Hutchins, J. B., and Barger, S. W. (1998). Why neurons die: cell death in the nervous system. Anat Rec 253, 79-90.

Hwang, H. C., and Clurman, B. E. (2005). Cyclin E in normal and neoplastic cell cycles. Oncogene 24, 2776-2786. 171

Jackman, M., Kubota, Y., den Elzen, N., Hagting, A., and Pines, J. (2002). Cyclin A- and cyclin E-Cdk complexes shuttle between the nucleus and the cytoplasm. Mol Biol Cell 13, 1030-1045.

Jackson, A. L., and Loeb, L. A. (2001). The contribution of endogenous sources of DNA damage to the multiple mutations in cancer. Mutat Res 477, 7-21.

Jackson, P. K., Chevalier, S., Philippe, M., and Kirschner, M. W. (1995). Early events in DNA replication require cyclin E and are blocked by p21CIP1. J Cell Biol 130, 755-769.

Jackson, S. P. (2001). Detecting, signalling and repairing DNA double-strand breaks. Biochem Soc Trans 29, 655-661.

Janiak, F., Leber, B., and Andrews, D. W. (1994). Assembly of Bcl-2 into microsomal and outer mitochondrial membranes. J Biol Chem 269, 9842-9849.

Jeggo, P. A. (1998). DNA breakage and repair. Adv Genet 38, 185-218.

Jentsch, S. (1992). The ubiquitin-conjugation system. Annu Rev Genet 26, 179-207.

Jin, J., Shirogane, T., Xu, L., Nalepa, G., Qin, J., Elledge, S. J., and Harper, J. W. (2003). SCFbeta-TRCP links Chk1 signaling to degradation of the Cdc25A protein phosphatase. Genes Dev 17, 3062-3074.

Jin, Y. H., Yoo, K. J., Lee, Y. H., and Lee, S. K. (2000). Caspase 3-mediated cleavage of p21WAF1/CIP1 associated with the cyclin A-cyclin-dependent kinase 2 complex is a prerequisite for apoptosis in SK-HEP-1 cells. J Biol Chem 275, 30256-30263.

Johnston, L. H. (1990). Periodic events in the cell cycle. Curr Opin Cell Biol 2, 274-279.

Joza, N., Susin, S. A., Daugas, E., Stanford, W. L., Cho, S. K., Li, C. Y., Sasaki, T., Elia, A. J., Cheng, H. Y., Ravagnan, L., et al. (2001). Essential role of the mitochondrial apoptosis-inducing factor in programmed cell death. Nature 410, 549-554.

Karsunky, H., Geisen, C., Schmidt, T., Haas, K., Zevnik, B., Gau, E., and Moroy, T. (1999). Oncogenic potential of cyclin E in T-cell lymphomagenesis in transgenic mice: evidence for cooperation between cyclin E and Ras but not Myc. Oncogene 18, 7816- 7824.

Kaufmann, S. H., and Hengartner, M. O. (2001). Programmed cell death: alive and well in the new millennium. Trends Cell Biol 11, 526-534.

Keeney, S. (2001). Mechanism and control of meiotic recombination initiation. Curr Top Dev Biol 52, 1-53. 172

Keyomarsi, K., Conte, D., Jr., Toyofuku, W., and Fox, M. P. (1995). Deregulation of cyclin E in breast cancer. Oncogene 11, 941-950.

Keyomarsi, K., O'Leary, N., Molnar, G., Lees, E., Fingert, H. J., and Pardee, A. B. (1994). Cyclin E, a potential prognostic marker for breast cancer. Cancer Res 54, 380- 385.

Keyomarsi, K., and Pardee, A. B. (1993). Redundant cyclin overexpression and gene amplification in breast cancer cells. Proc Natl Acad Sci U S A 90, 1112-1116.

Keyomarsi, K., Tucker, S. L., Buchholz, T. A., Callister, M., Ding, Y., Hortobagyi, G. N., Bedrosian, I., Knickerbocker, C., Toyofuku, W., Lowe, M., et al. (2002). Cyclin E and survival in patients with breast cancer. N Engl J Med 347, 1566-1575.

Khanna, K. K., and Jackson, S. P. (2001). DNA double-strand breaks: signaling, repair and the cancer connection. Nat Genet 27, 247-254.

Kischkel, F. C., Hellbardt, S., Behrmann, I., Germer, M., Pawlita, M., Krammer, P. H., and Peter, M. E. (1995). Cytotoxicity-dependent APO-1 (Fas/CD95)-associated proteins form a death-inducing signaling complex (DISC) with the receptor. Embo J 14, 5579- 5588.

Kischkel, F. C., Lawrence, D. A., Chuntharapai, A., Schow, P., Kim, K. J., and Ashkenazi, A. (2000). Apo2L/TRAIL-dependent recruitment of endogenous FADD and caspase-8 to death receptors 4 and 5. Immunity 12, 611-620.

Koch, C. A., Agyei, R., Galicia, S., Metalnikov, P., O'Donnell, P., Starostine, A., Weinfeld, M., and Durocher, D. (2004). Xrcc4 physically links DNA end processing by polynucleotide kinase to DNA ligation by DNA ligase IV. Embo J 23, 3874-3885.

Koepp, D. M., Schaefer, L. K., Ye, X., Keyomarsi, K., Chu, C., Harper, J. W., and Elledge, S. J. (2001). Phosphorylation-dependent ubiquitination of cyclin E by the SCFFbw7 ubiquitin ligase. Science 294, 173-177.

Koff, A., Cross, F., Fisher, A., Schumacher, J., Leguellec, K., Philippe, M., and Roberts, J. M. (1991). Human cyclin E, a new cyclin that interacts with two members of the CDC2 gene family. Cell 66, 1217-1228.

Koff, A., Giordano, A., Desai, D., Yamashita, K., Harper, J. W., Elledge, S., Nishimoto, T., Morgan, D. O., Franza, B. R., and Roberts, J. M. (1992). Formation and activation of a cyclin E-cdk2 complex during the G1 phase of the human cell cycle. Science 257, 1689-1694. 173

Koike, M., and Koike, A. (2005). The Ku70-binding site of Ku80 is required for the stabilization of Ku70 in the cytoplasm, for the nuclear translocation of Ku80, and for Ku80-dependent DNA repair. Exp Cell Res 305, 266-276.

Krek, W., Xu, G., and Livingston, D. M. (1995). Cyclin A-kinase regulation of E2F-1 DNA binding function underlies suppression of an S phase checkpoint. Cell 83, 1149- 1158.

Krude, T., Jackman, M., Pines, J., and Laskey, R. A. (1997). Cyclin/Cdk-dependent initiation of DNA replication in a human cell-free system. Cell 88, 109-119.

Krueger, A., Baumann, S., Krammer, P. H., and Kirchhoff, S. (2001). FLICE-inhibitory proteins: regulators of death receptor-mediated apoptosis. Mol Cell Biol 21, 8247-8254.

Kubbutat, M. H., Jones, S. N., and Vousden, K. H. (1997). Regulation of p53 stability by Mdm2. Nature 387, 299-303.

Kuerbitz, S. J., Plunkett, B. S., Walsh, W. V., and Kastan, M. B. (1992). Wild-type p53 is a determinant following irradiation. Proc Natl Acad Sci U S A 89, 7491-7495.

Kuhling, H., Alm, P., Olsson, H., Ferno, M., Baldetorp, B., Parwaresch, R., and Rudolph, P. (2003). Expression of cyclins E, A, and B, and prognosis in lymph node-negative breast cancer. J Pathol 199, 424-431.

Kysela, B., Chovanec, M., and Jeggo, P. A. (2005). Phosphorylation of linker histones by DNA-dependent protein kinase is required for DNA ligase IV-dependent ligation in the presence of histone H1. Proc Natl Acad Sci U S A 102, 1877-1882.

Kysela, B., Doherty, A. J., Chovanec, M., Stiff, T., Ameer-Beg, S. M., Vojnovic, B., Girard, P. M., and Jeggo, P. A. (2003). Ku stimulation of DNA ligase IV-dependent ligation requires inward movement along the DNA molecule. J Biol Chem 278, 22466- 22474.

Lam, E. W., and La Thangue, N. B. (1994). DP and E2F proteins: coordinating transcription with cell cycle progression. Curr Opin Cell Biol 6, 859-866.

Lane, M. E., Yu, B., Rice, A., Lipson, K. E., Liang, C., Sun, L., Tang, C., McMahon, G., Pestell, R. G., and Wadler, S. (2001). A novel cdk2-selective inhibitor, SU9516, induces apoptosis in colon carcinoma cells. Cancer Res 61, 6170-6177.

Lauper, N., Beck, A. R., Cariou, S., Richman, L., Hofmann, K., Reith, W., Slingerland, J. M., and Amati, B. (1998). Cyclin E2: a novel CDK2 partner in the late G1 and S phases of the mammalian cell cycle. Oncogene 17, 2637-2643. 174

Le Cam, L., Polanowska, J., Fabbrizio, E., Olivier, M., Philips, A., Ng Eaton, E., Classon, M., Geng, Y., and Sardet, C. (1999). Timing of cyclin E gene expression depends on the regulated association of a bipartite repressor element with a novel E2F complex. Embo J 18, 1878-1890.

Leber, R., Wise, T. W., Mizuta, R., and Meek, K. (1998). The XRCC4 gene product is a target for and interacts with the DNA-dependent protein kinase. J Biol Chem 273, 1794- 1801.

Lee, M. H., and Yang, H. Y. (2003). Regulators of G1 cyclin-dependent kinases and cancers. Cancer Metastasis Rev 22, 435-449.

Lees-Miller, S. P., and Meek, K. (2003). Repair of DNA double strand breaks by non- homologous end joining. Biochimie 85, 1161-1173.

Leist, M., and Jaattela, M. (2001). Four deaths and a funeral: from caspases to alternative mechanisms. Nat Rev Mol Cell Biol 2, 589-598.

Levine, A. J. (1997). p53, the cellular gatekeeper for growth and division. Cell 88, 323- 331.

Levkau, B., Koyama, H., Raines, E. W., Clurman, B. E., Herren, B., Orth, K., Roberts, J. M., and Ross, R. (1998). Cleavage of p21Cip1/Waf1 and p27Kip1 mediates apoptosis in endothelial cells through activation of Cdk2: role of a caspase cascade. Mol Cell 1, 553- 563.

Lew, D. J., Dulic, V., and Reed, S. I. (1991). Isolation of three novel human cyclins by rescue of G1 cyclin (Cln) function in yeast. Cell 66, 1197-1206.

Lew, D. J., and Kornbluth, S. (1996). Regulatory roles of cyclin dependent kinase phosphorylation in cell cycle control. Curr Opin Cell Biol 8, 795-804.

Li, G. C., He, F., Shao, X., Urano, M., Shen, L., Kim, D., Borrelli, M., Leibel, S. A., Gutin, P. H., and Ling, C. C. (2003). Adenovirus-mediated heat-activated antisense Ku70 expression radiosensitizes tumor cells in vitro and in vivo. Cancer Res 63, 3268-3274.

Li, J. Q., Miki, H., Ohmori, M., Wu, F., and Funamoto, Y. (2001a). Expression of cyclin E and cyclin-dependent kinase 2 correlates with metastasis and prognosis in colorectal carcinoma. Hum Pathol 32, 945-953.

Li, L. Y., Luo, X., and Wang, X. (2001b). Endonuclease G is an apoptotic DNase when released from mitochondria. Nature 412, 95-99. 175

Li, P., Nijhawan, D., Budihardjo, I., Srinivasula, S. M., Ahmad, M., Alnemri, E. S., and Wang, X. (1997). Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell 91, 479-489.

Li, Y., Chirgadze, D. Y., Bolanos-Garcia, V. M., Sibanda, B. L., Davies, O. R., Ahnesorg, P., Jackson, S. P., and Blundell, T. L. (2008). Crystal structure of human XLF/Cernunnos reveals unexpected differences from XRCC4 with implications for NHEJ. Embo J 27, 290-300.

Libertini, S. J., Robinson, B. S., Dhillon, N. K., Glick, D., George, M., Dandekar, S., Gregg, J. P., Sawai, E., and Mudryj, M. (2005). Cyclin E both regulates and is regulated by calpain 2, a protease associated with metastatic breast cancer phenotype. Cancer Res 65, 10700-10708.

Lieber, M. R. (1999). The biochemistry and biological significance of nonhomologous DNA end joining: an essential repair process in multicellular eukaryotes. Genes Cells 4, 77-85.

Lieber, M. R., Ma, Y., Pannicke, U., and Schwarz, K. (2003). Mechanism and regulation of human non-homologous DNA end-joining. Nat Rev Mol Cell Biol 4, 712-720.

Lindahl, T., and Nyberg, B. (1972). Rate of depurination of native deoxyribonucleic acid. Biochemistry 11, 3610-3618.

Lingle, W. L., Lutz, W. H., Ingle, J. N., Maihle, N. J., and Salisbury, J. L. (1998). Centrosome hypertrophy in human breast tumors: implications for genomic stability and cell polarity. Proc Natl Acad Sci U S A 95, 2950-2955.

Loden, M., Stighall, M., Nielsen, N. H., Roos, G., Emdin, S. O., Ostlund, H., and Landberg, G. (2002). The cyclin D1 high and cyclin E high subgroups of breast cancer: separate pathways in tumorogenesis based on pattern of genetic aberrations and inactivation of the pRb node. Oncogene 21, 4680-4690.

Lukas, J., Herzinger, T., Hansen, K., Moroni, M. C., Resnitzky, D., Helin, K., Reed, S. I., and Bartek, J. (1997). Cyclin E-induced S phase without activation of the pRb/E2F pathway. Genes Dev 11, 1479-1492.

Ma, T., Van Tine, B. A., Wei, Y., Garrett, M. D., Nelson, D., Adams, P. D., Wang, J., Qin, J., Chow, L. T., and Harper, J. W. (2000). Cell cycle-regulated phosphorylation of p220(NPAT) by cyclin E/Cdk2 in Cajal bodies promotes histone gene transcription. Genes Dev 14, 2298-2313.

Ma, Y., Lu, H., Schwarz, K., and Lieber, M. R. (2005a). Repair of double-strand DNA breaks by the human nonhomologous DNA end joining pathway: the iterative processing model. Cell Cycle 4, 1193-1200. 176

Ma, Y., Lu, H., Tippin, B., Goodman, M. F., Shimazaki, N., Koiwai, O., Hsieh, C. L., Schwarz, K., and Lieber, M. R. (2004). A biochemically defined system for mammalian nonhomologous DNA end joining. Mol Cell 16, 701-713.

Ma, Y., Pannicke, U., Lu, H., Niewolik, D., Schwarz, K., and Lieber, M. R. (2005b). The DNA-dependent protein kinase catalytic subunit phosphorylation sites in human Artemis. J Biol Chem 280, 33839-33846.

Ma, Y., Pannicke, U., Schwarz, K., and Lieber, M. R. (2002). Hairpin opening and overhang processing by an Artemis/DNA-dependent protein kinase complex in nonhomologous end joining and V(D)J recombination. Cell 108, 781-794.

Mahajan, K. N., Nick McElhinny, S. A., Mitchell, B. S., and Ramsden, D. A. (2002). Association of DNA polymerase mu (pol mu) with Ku and ligase IV: role for pol mu in end-joining double-strand break repair. Mol Cell Biol 22, 5194-5202.

Marangoni, E., Le Romancer, M., Foray, N., Muller, C., Douc-Rasy, S., Vaganay, S., Abdulkarim, B., Barrois, M., Calsou, P., Bernier, J., et al. (2000). Transfer of Ku86 RNA antisense decreases the radioresistance of human fibroblasts. Cancer Gene Ther 7, 339- 346.

Mari, P. O., Florea, B. I., Persengiev, S. P., Verkaik, N. S., Bruggenwirth, H. T., Modesti, M., Giglia-Mari, G., Bezstarosti, K., Demmers, J. A., Luider, T. M., et al. (2006). Dynamic assembly of end-joining complexes requires interaction between Ku70/80 and XRCC4. Proc Natl Acad Sci U S A 103, 18597-18602.

Martin, L. G., Demers, G. W., and Galloway, D. A. (1998). Disruption of the G1/S transition in human papillomavirus type 16 E7-expressing human cells is associated with altered regulation of cyclin E. J Virol 72, 975-985.

Martins, L. M., Iaccarino, I., Tenev, T., Gschmeissner, S., Totty, N. F., Lemoine, N. R., Savopoulos, J., Gray, C. W., Creasy, C. L., Dingwall, C., and Downward, J. (2002). The serine protease Omi/HtrA2 regulates apoptosis by binding XIAP through a reaper-like motif. J Biol Chem 277, 439-444.

Masui, Y., and Markert, C. L. (1971). Cytoplasmic control of nuclear behavior during meiotic maturation of frog oocytes. J Exp Zool 177, 129-145.

Matsumoto, Y., and Maller, J. L. (2004). A centrosomal localization signal in cyclin E required for Cdk2-independent S phase entry. Science 306, 885-888.

Matsumoto, Y., Suzuki, N., Namba, N., Umeda, N., Ma, X. J., Morita, A., Tomita, M., Enomoto, A., Serizawa, S., Hirano, K., et al. (2000). Cleavage and phosphorylation of XRCC4 protein induced by X-irradiation. FEBS Lett 478, 67-71. 177

Matsushime, H., Roussel, M. F., Ashmun, R. A., and Sherr, C. J. (1991). Colony- stimulating factor 1 regulates novel cyclins during the G1 phase of the cell cycle. Cell 65, 701-713.

Mazumder, S., DuPree, E. L., and Almasan, A. (2004). A dual role of cyclin E in cell proliferation and apoptosis may provide a target for cancer therapy. Curr Cancer Drug Targets 4, 65-75.

Mazumder, S., Gong, B., and Almasan, A. (2000). Cyclin E induction by genotoxic stress leads to apoptosis of hematopoietic cells. Oncogene 19, 2828-2835.

Mazumder, S., Gong, B., Chen, Q., Drazba, J. A., Buchsbaum, J. C., and Almasan, A. (2002). Proteolytic cleavage of cyclin E leads to inactivation of associated kinase activity and amplification of apoptosis in hematopoietic cells. Mol Cell Biol 22, 2398-2409.

Mazumder, S., Plesca, D., and Almasan, A. (2007a). A jekyll and hyde role of cyclin E in the genotoxic stress response: switching from cell cycle control to apoptosis regulation. Cell Cycle 6, 1437-1442. Epub 2007 May 1410.

Mazumder, S., Plesca, D., Kinter, M., and Almasan, A. (2007b). Interaction of a Cyclin E Fragment with Ku70 Regulates Bax-mediated Apoptosis. Mol Cell Biol 26, 26.

McEvoy, J. D., Kossatz, U., Malek, N., and Singer, J. D. (2007). Constitutive turnover of cyclin E by Cul3 maintains quiescence. Mol Cell Biol 27, 3651-3666.

McVey, D., Brizuela, L., Mohr, I., Marshak, D. R., Gluzman, Y., and Beach, D. (1989). Phosphorylation of large tumour antigen by cdc2 stimulates SV40 DNA replication. Nature 341, 503-507.

Mendoza, N., Fong, S., Marsters, J., Koeppen, H., Schwall, R., and Wickramasinghe, D. (2003). Selective cyclin-dependent kinase 2/cyclin A antagonists that differ from ATP site inhibitors block tumor growth. Cancer Res 63, 1020-1024.

Merkle, D., Douglas, P., Moorhead, G. B., Leonenko, Z., Yu, Y., Cramb, D., Bazett- Jones, D. P., and Lees-Miller, S. P. (2002). The DNA-dependent protein kinase interacts with DNA to form a protein-DNA complex that is disrupted by phosphorylation. Biochemistry 41, 12706-12714.

Michaloglou, C., Vredeveld, L. C., Soengas, M. S., Denoyelle, C., Kuilman, T., van der Horst, C. M., Majoor, D. M., Shay, J. W., Mooi, W. J., and Peeper, D. S. (2005). BRAFE600-associated senescence-like cell cycle arrest of human naevi. Nature 436, 720- 724.

Micheau, O., and Tschopp, J. (2003). Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes. Cell 114, 181-190. 178

Mills, K. D., Ferguson, D. O., and Alt, F. W. (2003). The role of DNA breaks in genomic instability and tumorigenesis. Immunol Rev 194, 77-95.

Minshull, J., Golsteyn, R., Hill, C. S., and Hunt, T. (1990). The A- and B-type cyclin associated cdc2 kinases in Xenopus turn on and off at different times in the cell cycle. Embo J 9, 2865-2875.

Mitchison, J. M. (1971). The Biology of the Cell Cycle (Cambridge, UK, Cambridge University Press).

Moberg, K. H., Bell, D. W., Wahrer, D. C., Haber, D. A., and Hariharan, I. K. (2001). Archipelago regulates Cyclin E levels in Drosophila and is mutated in human cancer cell lines. Nature 413, 311-316.

Modesti, M., Hesse, J. E., and Gellert, M. (1999). DNA binding of Xrcc4 protein is associated with V(D)J recombination but not with stimulation of DNA ligase IV activity. Embo J 18, 2008-2018.

Monaco, E. A., 3rd, Beaman-Hall, C. M., Mathur, A., and Vallano, M. L. (2004). Roscovitine, olomoucine, purvalanol: inducers of apoptosis in maturing cerebellar granule neurons. Biochem Pharmacol 67, 1947-1964.

Morgan, D. O. (1996). The dynamics of cyclin dependent kinase structure. Curr Opin Cell Biol 8, 767-772.

Moroy, T., and Geisen, C. (2004). Cyclin E. Int J Biochem Cell Biol 36, 1424-1439.

Mudryj, M., Devoto, S. H., Hiebert, S. W., Hunter, T., Pines, J., and Nevins, J. R. (1991). Cell cycle regulation of the E2F transcription factor involves an interaction with cyclin A. Cell 65, 1243-1253.

Murray, A., Hunt, T. (1993). The Cell Cycle (Oxford, UK, Oxford University Press).

Murray, A. W. (2004). Recycling the cell cycle: cyclins revisited. Cell 116, 221-234.

Murray, A. W., and Kirschner, M. W. (1989). Dominoes and clocks: the union of two views of the cell cycle. Science 246, 614-621.

Murray, A. W., and Kirschner, M. W. (1991). What controls the cell cycle? Sci Am 264, 56-63.

Mussman, J. G., Horn, H. F., Carroll, P. E., Okuda, M., Tarapore, P., Donehower, L. A., and Fukasawa, K. (2000). Synergistic induction of centrosome hyperamplification by loss of p53 and cyclin E overexpression. Oncogene 19, 1635-1646. 179

Muzio, M., Chinnaiyan, A. M., Kischkel, F. C., O'Rourke, K., Shevchenko, A., Ni, J., Scaffidi, C., Bretz, J. D., Zhang, M., Gentz, R., et al. (1996). FLICE, a novel FADD- homologous ICE/CED-3-like protease, is recruited to the CD95 (Fas/APO-1) death-- inducing signaling complex. Cell 85, 817-827.

Nagata, S., Nagase, H., Kawane, K., Mukae, N., and Fukuyama, H. (2003). Degradation of chromosomal DNA during apoptosis. Cell Death Differ 10, 108-116.

Nakano, K., and Vousden, K. H. (2001). PUMA, a novel proapoptotic gene, is induced by p53. Mol Cell 7, 683-694.

Nash, P., Tang, X., Orlicky, S., Chen, Q., Gertler, F. B., Mendenhall, M. D., Sicheri, F., Pawson, T., and Tyers, M. (2001). Multisite phosphorylation of a CDK inhibitor sets a threshold for the onset of DNA replication. Nature 414, 514-521.

Nasmyth, K. (2001). A prize for proliferation. Cell 107, 689-701.

Nevaldine, B., Longo, J. A., and Hahn, P. J. (1997). The scid defect results in much slower repair of DNA double-strand breaks but not high levels of residual breaks. Radiat Res 147, 535-540.

Nevins, J. R. (1992). E2F: a link between the Rb tumor suppressor protein and viral oncoproteins. Science 258, 424-429.

Nick McElhinny, S. A., Snowden, C. M., McCarville, J., and Ramsden, D. A. (2000). Ku recruits the XRCC4-ligase IV complex to DNA ends. Mol Cell Biol 20, 2996-3003.

Nielsen, N. H., Arnerlov, C., Emdin, S. O., and Landberg, G. (1996). Cyclin E overexpression, a negative prognostic factor in breast cancer with strong correlation to oestrogen receptor status. Br J Cancer 74, 874-880.

Noguchi, T., Dobashi, Y., Minehara, H., Itoman, M., and Kameya, T. (2000). Involvement of cyclins in cell proliferation and their clinical implications in soft tissue smooth muscle tumors. Am J Pathol 156, 2135-2147.

Nomura, M., Shimizu, S., Ito, T., Narita, M., Matsuda, H., and Tsujimoto, Y. (1999). Apoptotic cytosol facilitates Bax translocation to mitochondria that involves cytosolic factor regulated by Bcl-2. Cancer Res 59, 5542-5548.

O'Connor, L., Huang, D. C., O'Reilly, L. A., and Strasser, A. (2000). Apoptosis and cell division. Curr Opin Cell Biol 12, 257-263.

O'Driscoll, M., Gennery, A. R., Seidel, J., Concannon, P., and Jeggo, P. A. (2004). An overview of three new disorders associated with genetic instability: LIG4 syndrome, RS- SCID and ATR-Seckel syndrome. DNA Repair (Amst) 3, 1227-1235. 180

O'Driscoll, M., and Jeggo, P. A. (2006). The role of double-strand break repair - insights from human genetics. Nat Rev Genet 7, 45-54.

Oda, E., Ohki, R., Murasawa, H., Nemoto, J., Shibue, T., Yamashita, T., Tokino, T., Taniguchi, T., and Tanaka, N. (2000). Noxa, a BH3-only member of the Bcl-2 family and candidate mediator of p53-induced apoptosis. Science 288, 1053-1058.

Ohashi, R., Gao, C., Miyazaki, M., Hamazaki, K., Tsuji, T., Inoue, Y., Uemura, T., Hirai, R., Shimizu, N., and Namba, M. (2001). Enhanced expression of cyclin E and cyclin A in human hepatocellular carcinomas. Anticancer Res 21, 657-662.

Ohtani, K., DeGregori, J., and Nevins, J. R. (1995). Regulation of the cyclin E gene by transcription factor E2F1. Proc Natl Acad Sci U S A 92, 12146-12150.

Ohtsubo, M., and Roberts, J. M. (1993). Cyclin-dependent regulation of G1 in mammalian fibroblasts. Science 259, 1908-1912.

Ohtsubo, M., Theodoras, A. M., Schumacher, J., Roberts, J. M., and Pagano, M. (1995). Human cyclin E, a nuclear protein essential for the G1-to-S phase transition. Mol Cell Biol 15, 2612-2624.

Okuda, M., Horn, H. F., Tarapore, P., Tokuyama, Y., Smulian, A. G., Chan, P. K., Knudsen, E. S., Hofmann, I. A., Snyder, J. D., Bove, K. E., and Fukasawa, K. (2000). Nucleophosmin/B23 is a target of CDK2/cyclin E in centrosome duplication. Cell 103, 127-140.

Oltvai, Z. N., Milliman, C. L., and Korsmeyer, S. J. (1993). Bcl-2 heterodimerizes in vivo with a conserved homolog, Bax, that accelerates programmed cell death. Cell 74, 609- 619.

Orlicky, S., Tang, X., Willems, A., Tyers, M., and Sicheri, F. (2003). Structural basis for phosphodependent substrate selection and orientation by the SCFCdc4 ubiquitin ligase. Cell 112, 243-256.

Ortega, S., Prieto, I., Odajima, J., Martin, A., Dubus, P., Sotillo, R., Barbero, J. L., Malumbres, M., and Barbacid, M. (2003). Cyclin-dependent kinase 2 is essential for meiosis but not for mitotic cell division in mice. Nat Genet 35, 25-31.

Pagano, M., Draetta, G., and Jansen-Durr, P. (1992). Association of cdk2 kinase with the transcription factor E2F during S phase. Science 255, 1144-1147.

Pahler, J. C., Ruiz, S., Niemer, I., Calvert, L. R., Andreeff, M., Keating, M., Faderl, S., and McConkey, D. J. (2003). Effects of the proteasome inhibitor, bortezomib, on apoptosis in isolated lymphocytes obtained from patients with chronic lymphocytic leukemia. Clin Cancer Res 9, 4570-4577. 181

Pan, Z. Q., Amin, A. A., Gibbs, E., Niu, H., and Hurwitz, J. (1994). Phosphorylation of the p34 subunit of human single-stranded-DNA-binding protein in cyclin A-activated G1 extracts is catalyzed by cdk-cyclin A complex and DNA-dependent protein kinase. Proc Natl Acad Sci U S A 91, 8343-8347.

Papandreou, C. N., Daliani, D. D., Nix, D., Yang, H., Madden, T., Wang, X., Pien, C. S., Millikan, R. E., Tu, S. M., Pagliaro, L., et al. (2004). Phase I trial of the proteasome inhibitor bortezomib in patients with advanced solid tumors with observations in androgen-independent prostate cancer. J Clin Oncol 22, 2108-2121.

Paques, F., and Haber, J. E. (1999). Multiple pathways of recombination induced by double-strand breaks in Saccharomyces cerevisiae. Microbiol Mol Biol Rev 63, 349-404.

Pardee, A. B. (1989). G1 events and regulation of cell proliferation. Science 246, 603- 608.

Parisi, T., Beck, A. R., Rougier, N., McNeil, T., Lucian, L., Werb, Z., and Amati, B. (2003). Cyclins E1 and E2 are required for endoreplication in placental trophoblast giant cells. Embo J 22, 4794-4803.

Parker, L. L., Sylvestre, P. J., Byrnes, M. J., 3rd, Liu, F., and Piwnica-Worms, H. (1995). Identification of a 95-kDa -like tyrosine kinase in HeLa cells. Proc Natl Acad Sci U S A 92, 9638-9642.

Paull, T. T., Rogakou, E. P., Yamazaki, V., Kirchgessner, C. U., Gellert, M., and Bonner, W. M. (2000). A critical role for histone H2AX in recruitment of repair factors to nuclear foci after DNA damage. Curr Biol 10, 886-895.

Peter, M. E., and Krammer, P. H. (2003). The CD95(APO-1/Fas) DISC and beyond. Cell Death Differ 10, 26-35.

Petrini, J. H., and Stracker, T. H. (2003). The cellular response to DNA double-strand breaks: defining the sensors and mediators. Trends Cell Biol 13, 458-462.

Pickart, C. M., and Rose, I. A. (1985). Functional heterogeneity of ubiquitin carrier proteins. J Biol Chem 260, 1573-1581.

Pierce, A. J., Hu, P., Han, M., Ellis, N., and Jasin, M. (2001). Ku DNA end-binding protein modulates homologous repair of double-strand breaks in mammalian cells. Genes Dev 15, 3237-3242.

Poinsignon, C., de Chasseval, R., Soubeyrand, S., Moshous, D., Fischer, A., Hache, R. J., and de Villartay, J. P. (2004). Phosphorylation of Artemis following irradiation-induced DNA damage. Eur J Immunol 34, 3146-3155. 182

Polanowska, J., Fabbrizio, E., Le Cam, L., Trouche, D., Emiliani, S., Herrera, R., and Sardet, C. (2001). The periodic down regulation of Cyclin E gene expression from exit of mitosis to end of G(1) is controlled by a deacetylase- and E2F-associated bipartite repressor element. Oncogene 20, 4115-4127.

Poon, R. Y., and Hunter, T. (1995). Dephosphorylation of Cdk2 Thr160 by the cyclin- dependent kinase-interacting phosphatase KAP in the absence of cyclin. Science 270, 90- 93.

Poon, R. Y., and Hunter, T. (1998). Expression of a novel form of p21Cip1/Waf1 in UV- irradiated and transformed cells. Oncogene 16, 1333-1343.

Porter, D. C., Zhang, N., Danes, C., McGahren, M. J., Harwell, R. M., Faruki, S., and Keyomarsi, K. (2001). Tumor-specific proteolytic processing of cyclin E generates hyperactive lower-molecular-weight forms. Mol Cell Biol 21, 6254-6269.

Porter, P. L., Malone, K. E., Heagerty, P. J., Alexander, G. M., Gatti, L. A., Firpo, E. J., Daling, J. R., and Roberts, J. M. (1997). Expression of cell-cycle regulators p27Kip1 and cyclin E, alone and in combination, correlate with survival in young breast cancer patients. Nat Med 3, 222-225.

Pruschy, M., Wirbelauer, C., Glanzmann, C., Bodis, S., and Krek, W. (1999). E2F-1 has properties of a radiosensitizer and its regulation by cyclin A kinase is required for cell survival of fibrosarcoma cells lacking p53. Cell Growth Differ 10, 141-146.

Puthalakath, H., Huang, D. C., O'Reilly, L. A., King, S. M., and Strasser, A. (1999). The proapoptotic activity of the Bcl-2 family member Bim is regulated by interaction with the dynein motor complex. Mol Cell 3, 287-296.

Puthalakath, H., Villunger, A., O'Reilly, L. A., Beaumont, J. G., Coultas, L., Cheney, R. E., Huang, D. C., and Strasser, A. (2001). Bmf: a proapoptotic BH3-only protein regulated by interaction with the myosin V actin motor complex, activated by anoikis. Science 293, 1829-1832.

Raghavan, S. C., Kirsch, I. R., and Lieber, M. R. (2001). Analysis of the V(D)J recombination efficiency at lymphoid chromosomal translocation breakpoints. J Biol Chem 276, 29126-29133.

Rajagopalan, H., Jallepalli, P. V., Rago, C., Velculescu, V. E., Kinzler, K. W., Vogelstein, B., and Lengauer, C. (2004). Inactivation of hCDC4 can cause chromosomal instability. Nature 428, 77-81.

Rathmell, J. C., and Thompson, C. B. (2002). Pathways of apoptosis in lymphocyte development, homeostasis, and disease. Cell 109 Suppl, S97-107. 183

Reddy, Y. V., Ding, Q., Lees-Miller, S. P., Meek, K., and Ramsden, D. A. (2004). Non- homologous end joining requires that the DNA-PK complex undergo an autophosphorylation-dependent rearrangement at DNA ends. J Biol Chem 279, 39408- 39413.

Redon, C., Pilch, D., Rogakou, E., Sedelnikova, O., Newrock, K., and Bonner, W. (2002). Histone H2A variants H2AX and H2AZ. Curr Opin Genet Dev 12, 162-169.

Reed, S. I. (1996). Cyclin E: in mid-cycle. Biochim Biophys Acta 1287, 151-153.

Reed, S. I. (2003). Ratchets and clocks: the cell cycle, ubiquitylation and protein turnover. Nat Rev Mol Cell Biol 4, 855-864.

Resnitzky, D., Gossen, M., Bujard, H., and Reed, S. I. (1994). Acceleration of the G1/S phase transition by expression of cyclins D1 and E with an inducible system. Mol Cell Biol 14, 1669-1679.

Resnitzky, D., and Reed, S. I. (1995). Different roles for cyclins D1 and E in regulation of the G1-to-S transition. Mol Cell Biol 15, 3463-3469.

Richardson, C., Moynahan, M. E., and Jasin, M. (1998). Double-strand break repair by interchromosomal recombination: suppression of chromosomal translocations. Genes Dev 12, 3831-3842.

Richardson, P. G., Barlogie, B., Berenson, J., Singhal, S., Jagannath, S., Irwin, D., Rajkumar, S. V., Srkalovic, G., Alsina, M., Alexanian, R., et al. (2003). A phase 2 study of bortezomib in relapsed, refractory myeloma. N Engl J Med 348, 2609-2617.

Rios-Doria, J., Day, K. C., Kuefer, R., Rashid, M. G., Chinnaiyan, A. M., Rubin, M. A., and Day, M. L. (2003). The role of calpain in the proteolytic cleavage of E-cadherin in prostate and mammary epithelial cells. J Biol Chem 278, 1372-1379.

Rios-Doria, J., Kuefer, R., Ethier, S. P., and Day, M. L. (2004). Cleavage of beta-catenin by calpain in prostate and mammary tumor cells. Cancer Res 64, 7237-7240.

Roberts, J. M., and Sherr, C. J. (2003). Bared essentials of CDK2 and cyclin E. Nat Genet 35, 9-10.

Rodriguez, J., and Lazebnik, Y. (1999). Caspase-9 and APAF-1 form an active holoenzyme. Genes Dev 13, 3179-3184.

Rogakou, E. P., Pilch, D. R., Orr, A. H., Ivanova, V. S., and Bonner, W. M. (1998). DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139. J Biol Chem 273, 5858-5868. 184

Rooney, S., Chaudhuri, J., and Alt, F. W. (2004). The role of the non-homologous end- joining pathway in lymphocyte development. Immunol Rev 200, 115-131.

Roth, D. B., and Wilson, J. H. (1986). Nonhomologous recombination in mammalian cells: role for short sequence homologies in the joining reaction. Mol Cell Biol 6, 4295- 4304.

Russo, A., Fratto, M. E., Bazan, V., Schiro, V., Agnese, V., Cicero, G., Vincenzi, B., Tonini, G., and Santini, D. (2007). Targeting apoptosis in solid tumors: the role of bortezomib from preclinical to clinical evidence. Expert Opin Ther Targets 11, 1571- 1586.

Salvesen, G. S., and Duckett, C. S. (2002). IAP proteins: blocking the road to death's door. Nat Rev Mol Cell Biol 3, 401-410.

Sandhu, C., and Slingerland, J. (2000). Deregulation of the cell cycle in cancer. Cancer Detect Prev 24, 107-118.

Sauer, K., and Lehner, C. F. (1995). The role of cyclin E in the regulation of entry into S phase. Prog Cell Cycle Res 1, 125-139.

Scaffidi, C., Fulda, S., Srinivasan, A., Friesen, C., Li, F., Tomaselli, K. J., Debatin, K. M., Krammer, P. H., and Peter, M. E. (1998). Two CD95 (APO-1/Fas) signaling pathways. Embo J 17, 1675-1687.

Schild-Poulter, C., Shih, A., Tantin, D., Yarymowich, N. C., Soubeyrand, S., Sharp, P. A., and Hache, R. J. (2007). DNA-PK phosphorylation sites on Oct-1 promote cell survival following DNA damage. Oncogene 26, 3980-3988.

Schild-Poulter, C., Shih, A., Yarymowich, N. C., and Hache, R. J. (2003). Down- regulation of histone H2B by DNA-dependent protein kinase in response to DNA damage through modulation of octamer transcription factor 1. Cancer Res 63, 7197-7205.

Schraml, P., Bucher, C., Bissig, H., Nocito, A., Haas, P., Wilber, K., Seelig, S., Kononen, J., Mihatsch, M. J., Dirnhofer, S., and Sauter, G. (2003). Cyclin E overexpression and amplification in human tumours. J Pathol 200, 375-382.

Schwarz, K., Ma, Y., Pannicke, U., and Lieber, M. R. (2003). Human severe combined immune deficiency and DNA repair. Bioessays 25, 1061-1070.

Seghezzi, W., Chua, K., Shanahan, F., Gozani, O., Reed, R., and Lees, E. (1998). Cyclin E associates with components of the pre-mRNA splicing machinery in mammalian cells. Mol Cell Biol 18, 4526-4536. 185

Senderowicz, A. M. (2003). Small-molecule cyclin-dependent kinase modulators. Oncogene 22, 6609-6620.

Shanahan, F., Seghezzi, W., Parry, D., Mahony, D., and Lees, E. (1999). Cyclin E associates with BAF155 and BRG1, components of the mammalian SWI-SNF complex, and alters the ability of BRG1 to induce growth arrest. Mol Cell Biol 19, 1460-1469.

Sheaff, R. J., Groudine, M., Gordon, M., Roberts, J. M., and Clurman, B. E. (1997). Cyclin E-CDK2 is a regulator of p27Kip1. Genes Dev 11, 1464-1478.

Sherr, C. J. (1996). Cancer cell cycles. Science 274, 1672-1677.

Sherr, C. J., and McCormick, F. (2002). The RB and p53 pathways in cancer. Cancer Cell 2, 103-112.

Sherr, C. J., and Roberts, J. M. (1995). Inhibitors of mammalian G1 cyclin-dependent kinases. Genes Dev 9, 1149-1163.

Sherr, C. J., and Roberts, J. M. (1999). CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev 13, 1501-1512.

Shi, L., Qiu, D., Zhao, G., Corthesy, B., Lees-Miller, S., Reeves, W. H., and Kao, P. N. (2007). Dynamic binding of Ku80, Ku70 and NF90 to the IL-2 promoter in vivo in activated T-cells. Nucleic Acids Res 35, 2302-2310.

Shiloh, Y. (2003). ATM and related protein kinases: safeguarding genome integrity. Nat Rev Cancer 3, 155-168.

Singer, J. D., Gurian-West, M., Clurman, B., and Roberts, J. M. (1999). Cullin-3 targets cyclin E for ubiquitination and controls S phase in mammalian cells. Genes Dev 13, 2375-2387.

Skowyra, D., Craig, K. L., Tyers, M., Elledge, S. J., and Harper, J. W. (1997). F-box proteins are receptors that recruit phosphorylated substrates to the SCF ubiquitin-ligase complex. Cell 91, 209-219.

Smith, G. C., and Jackson, S. P. (1999). The DNA-dependent protein kinase. Genes Dev 13, 916-934.

Smits, V. A., and Medema, R. H. (2001). Checking out the G(2)/M transition. Biochim Biophys Acta 1519, 1-12.

Sobol, R. W. (2008). CHIPping away at base excision repair. Mol Cell 29, 413-415. 186

Span, P. N., Tjan-Heijnen, V. C., Manders, P., Beex, L. V., and Sweep, C. G. (2003). Cyclin-E is a strong predictor of endocrine therapy failure in human breast cancer. Oncogene 22, 4898-4904.

Sprick, M. R., Weigand, M. A., Rieser, E., Rauch, C. T., Juo, P., Blenis, J., Krammer, P. H., and Walczak, H. (2000). FADD/MORT1 and caspase-8 are recruited to TRAIL receptors 1 and 2 and are essential for apoptosis mediated by TRAIL receptor 2. Immunity 12, 599-609.

Spruck, C. H., Strohmaier, H., Sangfelt, O., Muller, H. M., Hubalek, M., Muller-Holzner, E., Marth, C., Widschwendter, M., and Reed, S. I. (2002). hCDC4 gene mutations in endometrial cancer. Cancer Res 62, 4535-4539.

Spruck, C. H., Won, K. A., and Reed, S. I. (1999). Deregulated cyclin E induces chromosome instability. Nature 401, 297-300.

Srinivasula, S. M., Hegde, R., Saleh, A., Datta, P., Shiozaki, E., Chai, J., Lee, R. A., Robbins, P. D., Fernandes-Alnemri, T., Shi, Y., and Alnemri, E. S. (2001). A conserved XIAP-interaction motif in caspase-9 and Smac/DIABLO regulates caspase activity and apoptosis. Nature 410, 112-116.

Stabach, P. R., Cianci, C. D., Glantz, S. B., Zhang, Z., and Morrow, J. S. (1997). Site- directed mutagenesis of alpha II spectrin at codon 1175 modulates its mu-calpain susceptibility. Biochemistry 36, 57-65.

Strasser, A., O'Connor, L., and Dixit, V. M. (2000). Apoptosis signaling. Annu Rev Biochem 69, 217-245.

Strohmaier, H., Spruck, C. H., Kaiser, P., Won, K. A., Sangfelt, O., and Reed, S. I. (2001). Human F-box protein hCdc4 targets cyclin E for proteolysis and is mutated in a breast cancer cell line. Nature 413, 316-322.

Subramanian, C., Opipari, A. W., Jr., Bian, X., Castle, V. P., and Kwok, R. P. (2005). Ku70 acetylation mediates neuroblastoma cell death induced by histone deacetylase inhibitors. Proc Natl Acad Sci U S A 102, 4842-4847.

Sugimoto, M., Martin, N., Wilks, D. P., Tamai, K., Huot, T. J., Pantoja, C., Okumura, K., Serrano, M., and Hara, E. (2002). Activation of cyclin D1-kinase in murine fibroblasts lacking both p21(Cip1) and p27(Kip1). Oncogene 21, 8067-8074.

Susin, S. A., Lorenzo, H. K., Zamzami, N., Marzo, I., Snow, B. E., Brothers, G. M., Mangion, J., Jacotot, E., Costantini, P., Loeffler, M., et al. (1999). Molecular characterization of mitochondrial apoptosis-inducing factor. Nature 397, 441-446. 187

Taghian, D. G., and Nickoloff, J. A. (1997). Chromosomal double-strand breaks induce gene conversion at high frequency in mammalian cells. Mol Cell Biol 17, 6386-6393.

Takahashi, R., Deveraux, Q., Tamm, I., Welsh, K., Assa-Munt, N., Salvesen, G. S., and Reed, J. C. (1998). A single BIR domain of XIAP sufficient for inhibiting caspases. J Biol Chem 273, 7787-7790.

Takata, M., Sasaki, M. S., Sonoda, E., Morrison, C., Hashimoto, M., Utsumi, H., Yamaguchi-Iwai, Y., Shinohara, A., and Takeda, S. (1998). Homologous recombination and non-homologous end-joining pathways of DNA double-strand break repair have overlapping roles in the maintenance of chromosomal integrity in vertebrate cells. Embo J 17, 5497-5508.

Tetzlaff, M. T., Yu, W., Li, M., Zhang, P., Finegold, M., Mahon, K., Harper, J. W., Schwartz, R. J., and Elledge, S. J. (2004). Defective cardiovascular development and elevated cyclin E and Notch proteins in mice lacking the Fbw7 F-box protein. Proc Natl Acad Sci U S A 101, 3338-3345.

Thornberry, N. A., and Lazebnik, Y. (1998). Caspases: enemies within. Science 281, 1312-1316.

Tomkinson, A. E., Vijayakumar, S., Pascal, J. M., and Ellenberger, T. (2006). DNA ligases: structure, reaction mechanism, and function. Chem Rev 106, 687-699.

Tsai, C. J., Kim, S. A., and Chu, G. (2007). Cernunnos/XLF promotes the ligation of mismatched and noncohesive DNA ends. Proc Natl Acad Sci U S A 104, 7851-7856.

Tsai, L. H., Lees, E., Faha, B., Harlow, E., and Riabowol, K. (1993). The cdk2 kinase is required for the G1-to-S transition in mammalian cells. Oncogene 8, 1593-1602.

Tsujimoto, Y., Finger, L. R., Yunis, J., Nowell, P. C., and Croce, C. M. (1984). Cloning of the chromosome breakpoint of neoplastic B cells with the t(14;18) chromosome translocation. Science 226, 1097-1099.

Tsunematsu, R., Nakayama, K., Oike, Y., Nishiyama, M., Ishida, N., Hatakeyama, S., Bessho, Y., Kageyama, R., Suda, T., and Nakayama, K. I. (2004). Mouse Fbw7/Sel- 10/Cdc4 is required for notch degradation during vascular development. J Biol Chem 279, 9417-9423.

Turchi, J. J., Henkels, K. M., and Zhou, Y. (2000). Cisplatin-DNA adducts inhibit translocation of the Ku subunits of DNA-PK. Nucleic Acids Res 28, 4634-4641.

Uziel, T., Lerenthal, Y., Moyal, L., Andegeko, Y., Mittelman, L., and Shiloh, Y. (2003). Requirement of the MRN complex for ATM activation by DNA damage. Embo J 22, 5612-5621. 188

Valerie, K., and Povirk, L. F. (2003). Regulation and mechanisms of mammalian double- strand break repair. Oncogene 22, 5792-5812.

van den Heuvel, S., and Harlow, E. (1993). Distinct roles for cyclin-dependent kinases in cell cycle control. Science 262, 2050-2054.

van der Voorn, L., and Ploegh, H. L. (1992). The WD-40 repeat. FEBS Lett 307, 131- 134.

van Drogen, F., Sangfelt, O., Malyukova, A., Matskova, L., Yeh, E., Means, A. R., and Reed, S. I. (2006). Ubiquitylation of cyclin E requires the sequential function of SCF complexes containing distinct hCdc4 isoforms. Mol Cell 23, 37-48.

van Gent, D. C., Hoeijmakers, J. H., and Kanaar, R. (2001). Chromosomal stability and the DNA double-stranded break connection. Nat Rev Genet 2, 196-206.

Vaux, D. L., and Strasser, A. (1996). The molecular biology of apoptosis. Proc Natl Acad Sci U S A 93, 2239-2244.

Verhagen, A. M., Ekert, P. G., Pakusch, M., Silke, J., Connolly, L. M., Reid, G. E., Moritz, R. L., Simpson, R. J., and Vaux, D. L. (2000). Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins. Cell 102, 43-53.

Vlach, J., Hennecke, S., and Amati, B. (1997). Phosphorylation-dependent degradation of the cyclin-dependent kinase inhibitor p27. Embo J 16, 5334-5344.

Walker, J. R., Corpina, R. A., and Goldberg, J. (2001). Structure of the Ku heterodimer bound to DNA and its implications for double-strand break repair. Nature 412, 607-614.

Wang, H., Wang, M., Wang, H., Bocker, W., and Iliakis, G. (2005). Complex H2AX phosphorylation patterns by multiple kinases including ATM and DNA-PK in human cells exposed to ionizing radiation and treated with kinase inhibitors. J Cell Physiol 202, 492-502.

Wang, H., Zeng, Z. C., Bui, T. A., Sonoda, E., Takata, M., Takeda, S., and Iliakis, G. (2001a). Efficient rejoining of radiation-induced DNA double-strand breaks in vertebrate cells deficient in genes of the RAD52 epistasis group. Oncogene 20, 2212-2224.

Wang, H., Zeng, Z. C., Perrault, A. R., Cheng, X., Qin, W., and Iliakis, G. (2001b). Genetic evidence for the involvement of DNA ligase IV in the DNA-PK-dependent pathway of non-homologous end joining in mammalian cells. Nucleic Acids Res 29, 1653-1660. 189

Wang, M., Wu, W., Wu, W., Rosidi, B., Zhang, L., Wang, H., and Iliakis, G. (2006). PARP-1 and Ku compete for repair of DNA double strand breaks by distinct NHEJ pathways. Nucleic Acids Res 34, 6170-6182.

Wang, S., Guo, M., Ouyang, H., Li, X., Cordon-Cardo, C., Kurimasa, A., Chen, D. J., Fuks, Z., Ling, C. C., and Li, G. C. (2000). The catalytic subunit of DNA-dependent protein kinase selectively regulates p53-dependent apoptosis but not cell-cycle arrest. Proc Natl Acad Sci U S A 97, 1584-1588.

Wang, W., and El-Deiry, W. S. (2008). Restoration of p53 to limit tumor growth. Curr Opin Oncol 20, 90-96.

Wang, X. D., Rosales, J. L., Magliocco, A., Gnanakumar, R., and Lee, K. Y. (2003). Cyclin E in breast tumors is cleaved into its low molecular weight forms by calpain. Oncogene 22, 769-774.

Wang, Y., Penfold, S., Tang, X., Hattori, N., Riley, P., Harper, J. W., Cross, J. C., and Tyers, M. (1999). Deletion of the Cul1 gene in mice causes arrest in early embryogenesis and accumulation of cyclin E. Curr Biol 9, 1191-1194.

Wei, M. C., Lindsten, T., Mootha, V. K., Weiler, S., Gross, A., Ashiya, M., Thompson, C. B., and Korsmeyer, S. J. (2000). tBID, a membrane-targeted death ligand, oligomerizes BAK to release cytochrome c. Genes Dev 14, 2060-2071.

Wei, W., Jin, J., Schlisio, S., Harper, J. W., and Kaelin, W. G., Jr. (2005). The v-Jun point mutation allows c-Jun to escape GSK3-dependent recognition and destruction by the Fbw7 ubiquitin ligase. Cancer Cell 8, 25-33.

Weinberg, R. A. (1995). The retinoblastoma protein and cell cycle control. Cell 81, 323- 330.

Welcker, M., and Clurman, B. E. (2005). The SV40 large T antigen contains a decoy phosphodegron that mediates its interactions with Fbw7/hCdc4. J Biol Chem 280, 7654- 7658.

Welcker, M., and Clurman, B. E. (2008). FBW7 ubiquitin ligase: a tumour suppressor at the crossroads of cell division, growth and differentiation. Nat Rev Cancer 8, 83-93.

Welcker, M., Orian, A., Grim, J. E., Eisenman, R. N., and Clurman, B. E. (2004). A nucleolar isoform of the Fbw7 ubiquitin ligase regulates c-Myc and cell size. Curr Biol 14, 1852-1857.

Welcker, M., Singer, J., Loeb, K. R., Grim, J., Bloecher, A., Gurien-West, M., Clurman, B. E., and Roberts, J. M. (2003). Multisite phosphorylation by Cdk2 and GSK3 controls cyclin E degradation. Mol Cell 12, 381-392. 190

West, R. B., Yaneva, M., and Lieber, M. R. (1998). Productive and nonproductive complexes of Ku and DNA-dependent protein kinase at DNA termini. Mol Cell Biol 18, 5908-5920.

West, S. C. (2003). Molecular views of recombination proteins and their control. Nat Rev Mol Cell Biol 4, 435-445.

Weterings, E., and van Gent, D. C. (2004). The mechanism of non-homologous end- joining: a synopsis of synapsis. DNA Repair (Amst) 3, 1425-1435.

Weterings, E., Verkaik, N. S., Bruggenwirth, H. T., Hoeijmakers, J. H., and van Gent, D. C. (2003). The role of DNA dependent protein kinase in synapsis of DNA ends. Nucleic Acids Res 31, 7238-7246.

Whittaker, S. R., Walton, M. I., Garrett, M. D., and Workman, P. (2004). The Cyclin- dependent kinase inhibitor CYC202 (R-roscovitine) inhibits retinoblastoma protein phosphorylation, causes loss of Cyclin D1, and activates the mitogen-activated protein kinase pathway. Cancer Res 64, 262-272.

Wilson-Annan, J., O'Reilly, L. A., Crawford, S. A., Hausmann, G., Beaumont, J. G., Parma, L. P., Chen, L., Lackmann, M., Lithgow, T., Hinds, M. G., et al. (2003). Proapoptotic BH3-only proteins trigger membrane integration of prosurvival Bcl-w and neutralize its activity. J Cell Biol 162, 877-887.

Wilson, T. E., and Lieber, M. R. (1999). Efficient processing of DNA ends during yeast nonhomologous end joining. Evidence for a DNA polymerase beta (Pol4)-dependent pathway. J Biol Chem 274, 23599-23609.

Winston, J. T., Koepp, D. M., Zhu, C., Elledge, S. J., and Harper, J. W. (1999). A family of mammalian F-box proteins. Curr Biol 9, 1180-1182.

Wolter, K. G., Hsu, Y. T., Smith, C. L., Nechushtan, A., Xi, X. G., and Youle, R. J. (1997). Movement of Bax from the cytosol to mitochondria during apoptosis. J Cell Biol 139, 1281-1292.

Won, K. A., and Reed, S. I. (1996). Activation of cyclin E/CDK2 is coupled to site- specific autophosphorylation and ubiquitin-dependent degradation of cyclin E. Embo J 15, 4182-4193.

Woo, R. A., Jack, M. T., Xu, Y., Burma, S., Chen, D. J., and Lee, P. W. (2002). DNA damage-induced apoptosis requires the DNA-dependent protein kinase, and is mediated by the latent population of p53. Embo J 21, 3000-3008. 191

Wu, G., Lyapina, S., Das, I., Li, J., Gurney, M., Pauley, A., Chui, I., Deshaies, R. J., and Kitajewski, J. (2001). SEL-10 is an inhibitor of notch signaling that targets notch for ubiquitin-mediated protein degradation. Mol Cell Biol 21, 7403-7415.

Wyllie, A. H., Kerr, J. F., and Currie, A. R. (1980). Cell death: the significance of apoptosis. Int Rev Cytol 68, 251-306.

Xiong, Y., Connolly, T., Futcher, B., and Beach, D. (1991). Human D-type cyclin. Cell 65, 691-699.

Yada, M., Hatakeyama, S., Kamura, T., Nishiyama, M., Tsunematsu, R., Imaki, H., Ishida, N., Okumura, F., Nakayama, K., and Nakayama, K. I. (2004). Phosphorylation- dependent degradation of c-Myc is mediated by the F-box protein Fbw7. Embo J 23, 2116-2125.

Yamaguchi, R., Maki, M., Hatanaka, M., and Sabe, H. (1994). Unphosphorylated and tyrosine-phosphorylated forms of a focal adhesion protein, paxillin, are substrates for calpain II in vitro: implications for the possible involvement of calpain II in mitosis- specific degradation of paxillin. FEBS Lett 356, 114-116.

Yaneva, M., Kowalewski, T., and Lieber, M. R. (1997). Interaction of DNA-dependent protein kinase with DNA and with Ku: biochemical and atomic-force microscopy studies. Embo J 16, 5098-5112.

Ye, X., Nalepa, G., Welcker, M., Kessler, B. M., Spooner, E., Qin, J., Elledge, S. J., Clurman, B. E., and Harper, J. W. (2004). Recognition of phosphodegron motifs in human cyclin E by the SCF(Fbw7) ubiquitin ligase. J Biol Chem 279, 50110-50119.

Yeh, K. H., Kondo, T., Zheng, J., Tsvetkov, L. M., Blair, J., and Zhang, H. (2001). The F-box protein SKP2 binds to the phosphorylated threonine 380 in cyclin E and regulates ubiquitin-dependent degradation of cyclin E. Biochem Biophys Res Commun 281, 884- 890.

Yoo, S., and Dynan, W. S. (1999). Geometry of a complex formed by double strand break repair proteins at a single DNA end: recruitment of DNA-PKcs induces inward translocation of Ku protein. Nucleic Acids Res 27, 4679-4686.

Yoshida, T., Tomioka, I., Nagahara, T., Holyst, T., Sawada, M., Hayes, P., Gama, V., Okuno, M., Chen, Y., Abe, Y., et al. (2004). Bax-inhibiting peptide derived from mouse and rat Ku70. Biochem Biophys Res Commun 321, 961-966.

Yu, C., Rahmani, M., Dai, Y., Conrad, D., Krystal, G., Dent, P., and Grant, S. (2003a). The lethal effects of pharmacological cyclin-dependent kinase inhibitors in human leukemia cells proceed through a phosphatidylinositol 3-kinase/Akt-dependent process. Cancer Res 63, 1822-1833. 192

Yu, Y., Wang, W., Ding, Q., Ye, R., Chen, D., Merkle, D., Schriemer, D., Meek, K., and Lees-Miller, S. P. (2003b). DNA-PK phosphorylation sites in XRCC4 are not required for survival after radiation or for V(D)J recombination. DNA Repair (Amst) 2, 1239- 1252.

Yurchenko, V., Xue, Z., Gama, V., Matsuyama, S., and Sadofsky, M. J. (2008). Ku70 is stabilized by increased cellular SUMO. Biochem Biophys Res Commun 366, 263-268.

Yurchenko, V., Xue, Z., and Sadofsky, M. J. (2006). SUMO modification of human XRCC4 regulates its localization and function in DNA double-strand break repair. Mol Cell Biol 26, 1786-1794.

Zariwala, M., Liu, J., and Xiong, Y. (1998). Cyclin E2, a novel human G1 cyclin and activating partner of CDK2 and CDK3, is induced by viral oncoproteins. Oncogene 17, 2787-2798.

Zhang, H. S., Gavin, M., Dahiya, A., Postigo, A. A., Ma, D., Luo, R. X., Harbour, J. W., and Dean, D. C. (2000). Exit from G1 and S phase of the cell cycle is regulated by repressor complexes containing HDAC-Rb-hSWI/SNF and Rb-hSWI/SNF. Cell 101, 79- 89.

Zhang, W., and Koepp, D. M. (2006). Fbw7 isoform interaction contributes to cyclin E proteolysis. Mol Cancer Res 4, 935-943.

Zhang, X. M., Lin, H., Chen, C., and Chen, B. D. (1999). Inhibition of ubiquitin- proteasome pathway activates a caspase-3-like protease and induces Bcl-2 cleavage in human M-07e leukaemic cells. Biochem J 340, 127-133.

Zhang, Y., Wu, X., Yuan, F., Xie, Z., and Wang, Z. (2001). Highly frequent frameshift DNA synthesis by human DNA polymerase mu. Mol Cell Biol 21, 7995-8006.

Zhao, J., Kennedy, B. K., Lawrence, B. D., Barbie, D. A., Matera, A. G., Fletcher, J. A., and Harlow, E. (2000). NPAT links cyclin E-Cdk2 to the regulation of replication- dependent histone gene transcription. Genes Dev 14, 2283-2297.

Zou, H., Henzel, W. J., Liu, X., Lutschg, A., and Wang, X. (1997). Apaf-1, a human protein homologous to C. elegans CED-4, participates in cytochrome c-dependent activation of caspase-3. Cell 90, 405-413.

Zuzarte-Luis, V., and Hurle, J. M. (2005). Programmed cell death in the embryonic vertebrate limb. Semin Cell Dev Biol 16, 261-269.

APPENDIX

Abbreviations

53BP1 p53 binding protein 1

AIF apoptosis inducing factor

Apaf-1 apoptotic protease activating factor 1

APC anaphase promoting complex

ATM ataxia-telangiectasia mutated

ATP adenosine triphosphate

ATR ataxia-telangiectasia and rad3 related

Bad Bcl-2 antagonist of cell death

Bak Bcl-2 antagonist killer

Bax Bcl-2-associated X protein

Bcl-2 B-cell CLL/lymphoma 2

Bcl-xL, Bcl-2 like 1, long form

BH Bcl-2 homology domain

BH3 Bcl-2 homology domain 3

Bik Bcl-2 interacting killer

Bim Bcl-2-interacting protein

BIR baculoviral IAP repeats

193 194

bp base pair

BRCA1 breast cancer 1 gene

BRCT BRCA1 C-termini

BSA bovine serum albumin

CAD caspase-activated DNase

CARD caspase recruitment domain

Caspase cysteine aspartate protease

Caspase-3 apoptosis-related cysteine protease 3

CDK cyclin-dependent kinase

Cdk2 cyclin-dependent kinase 2

cDNA complimentary deoxyribonucleic acid

Cernnunos XLF, XRCC4-like factor

CKI cyclin-dependent kinase inhibitors

CIP CDK inhibitory protein

Cyt C cytochrome c

DED death effector domain

DD death domain

DHFR dihydrofolate reductase

DN dominant negative

DNA deoxyribonucleic acid

DNA-PKcs DNA protein kinase catalytic subunit

DP1/DP2 dimerization protein 1/2 195

DR death receptor

DSB double-strand break

E1A early region 1A (E1A) products of adenovirus 5 (Ad5)

E2F E2 factor, cellular factor required for E2 viral promoter activation

EDTA ethylenediaminotetraacetic acid

EF1α elongation factor 1 α

EGFP enhanced green fluorescence protein

FADD Fas associated protein with death domain

FLIP FLICE-inhibitory protein

G1 phase gap 1 phase of the cell cycle

G2 phase gap 2 phase of the cell cycle

GFP green fluorescence protein

GSK3β glycogen synthase kinase 3β

Gy gray (dose unit of IR)

HDAC histone deacetylase

Hdm2 human homologue of mouse double minute 2

HMG1 high mobility group 1

HR homologous recombination

IAP inhibitor of apoptosis protein

ICAD inhibitor of CAD

ICE interleukin-1beta converting enzyme

IgGλ immunoglobulin G light chain 196

IR ionizing radiation

kDa kilodalton

LMW low molecular weight

LT large T antigen

LV lentivirus

M phase mitosis phase of the cell cycle

Mcl-1 myeloid cell leukemia sequence 1

MCM minichromosome maintenance protein(s)

MEF mouse embryonic fibroblast

MEK mitogen-activated protein/ERK kinase

MMR mismatch repair

MPF maturation promoting factor or M-phase promoting factor

mRNA messenger ribonucleic acid

Nbs1 Nijmegen breakage syndrome, nibrin

NER nucleotide excision repair

NF-κB nuclear factor κB

NHEJ non-homologous end-joining repair

NOXA phorbol-12-myristate-13-acetate-induced protein 1

NP-40 Nonidet P-40, non-ionic detergent p21 cyclin-dependent kinase inhibitor-1A (CDKN1A) p27/Kip1 cyclin-dependent kinase inhibitor

p53 tumor suppressor protein p53 197

p107 retinoblastoma-like 1; RBL1 p130 retinoblastoma-like 2; RBL2

PAGE polyacrylamide gel electrophoresis

PARP-1 poly (ADP-ribose) polymerase-1

PBS phosphate buffered saline

PCR polymerase chain reaction

PMSF phenylmethylsulphonylfluoride

PUMA p53-upregulated modulator of apoptosis

PI propidium iodide

PI3K phosphatidylinositol 3-kinase

PS phosphatidylserine

R point restriction point

Rb retinoblastoma

RNA ribonucleic acid

ROS reactive oxygen species

S phase synthesis phase of the cell cycle

SCF Skp1-Cul1-F box or Skp1-Cul1-Fbw7

SD standard deviation

SDS-PAGE sodium dodecyl sulfate polyacrylamide gel electrophoresis siRNA small interference RNA

SSB single-strand break tBid truncated Bid 198

TK thymidine kinase

TM transmembrane region

TNF tumor necrosis factor

TNFR tumor necrosis factor receptor

TRAIL tumor necrosis factor-related apoptosis-inducing ligand tRNA transfer RNA

TS thymidilate synthase

VEGF vascular endothelial growth factor

XRCC4 X-ray repair complementing defective repair in Chinese hamster cells 4

XLF XRCC4-like factor, Cernnunos