Download e2f and survivin – key players in cellular proliferation

Document related concepts
no text concepts found
Transcript
E2F AND SURVIVIN – KEY PLAYERS IN CELLULAR PROLIFERATION AND
TRANSFORMATION
DISSERTATION
Presented in Partial Fulfillment of the Requirements for
the Degree Doctor of Philosophy in the
Graduate School of The Ohio State University
By
Baidehi Maiti, M.B.B.S.
*****
The Ohio State University
2007
Dissertation Committee:
Dr. Rachel A. Altura, Advisor
Approved by
Dr. Arthur H.M. Burghes
Dr. Albert de la Chapelle
Dr. Jiayuh Lin
Advisor
Graduate Program in Molecular Genetics
ABSTRACT
The E2F transcription factor family and survivin play a crucial role in
maintaining homeostasis by coupling the vital processes of cell division and
apoptosis. Deregulation of E2F or survivin signaling, lead to developmental
defects in mice and are hallmarks of virtually every human cancer.
Functionally, the mammalian E2F family consists of activators (E2F1-2
and E2F3a) and repressors (E2F3b and E2F4-8), which regulate cellular
proliferation, apoptosis and differentiation. E2F7 and E2F8, which are the last
two discovered mouse E2F family members, structurally form a sub-class within
the repressor E2Fs. E2F7-8 are distinct in having a duplicated DNA binding
domain, which is highly conserved across all the E2F family members. Both
E2F7 and E2F8 inhibit cellular proliferation when overexpressed in mouse
embryonic fibroblasts (MEFs). Expression of E2F7 and E2F8 are cell cycle
regulated and are detected in the same adult mouse tissues suggesting a
functional overlap or synergy.
Consistent with the expanding complexity and functional diversity of the
large mammalian E2F family, a cell cycle checkpoint is also regulated by the
ii
E2Fs. Simultaneous deletion of E2F1, E2F2 and E2F3, leads to a severe
proliferative defect in the MEFs, correlating with the upregulation of p21 and the
absence of cyclin dependent kinase activity.
A proliferative defect is also observed in the MEFs conditionally deleted
for survivin, a direct transcriptional target of the E2Fs. Survivin performs dual
functions as a regulator of cell division and as an anti-apoptotic protein. During
cell division, survivin ensures high fidelity chromosomal segregation and proper
cytokinesis. Accordingly, survivin deleted MEFs fail to proliferate and become
increasingly polyploid with an abnormal nuclear morphology. However, they do
not undergo apoptosis, indicating a role of survivin in proliferation but not
survival. Additionally, MEFs transformed with oncogenes c-Myc and H-RasV12
fail to proliferate following conditional deletion of survivin, demonstrating a
requirement for survivin in the maintenance of transformation. Preliminary
evidence also suggests that survivin is capable of collaborating with c-Myc or HRasV12 in transforming MEFs, unraveling a novel role for survivin in cancer
initiation.
Collectively, these data uncover important roles of the E2F family and
survivin in cellular proliferation and transformation.
iii
Dedicated to my parents,
Dr. Bibekananda Maiti
and
Dr. Ilorasri Chakraborti
for everything you do
iv
ACKNOWLEDGMENTS
I would like to thank my advisor Dr. Rachel A. Altura for her excellent
guidance, advice, encouragement and support. I am grateful to her for providing
me a scientifically stimulating, enjoyable and productive mentor-student
relationship. She has been instrumental in making this thesis possible.
I wish to extend my gratitude to Dr. Gustavo W. Leone for giving me his
time, supervision, scientific training and advice.
I am thankful to all my current and past graduate advisory committee
members, Dr. Arthur H. M. Burghes, Dr. Albert de la Chapelle, Dr. Jiayuh Lin, Dr.
Lee F. Johnson and Dr. Michael C. Ostrowski for their time and effort towards my
scientific development.
I would like to acknowledge the contributions of all my present and past
colleagues from the Altura and Leone labs. Thanks to Harold I. Saavedra, Rene
Opavsky and Yuying Jiang, not only for their excellent scientific advice but also
for being wonderfully supportive friends. It has also been a great learning
experience to work with Cynthia McAllister, Michael P. Holloway, Lizhao Wu,
Cynthia Timmers and Alain de bruin.
v
Thank you to Dr. Mark A. Seeger, Dr. Berl R. Oakley and Dr. David M.
Bisaro for your kind and active support through some of the most challenging
times of my career. I am also thankful to the staff of Molecular Genetics for their
excellent timely help in the official procedures and paperwork.
Finally, none of my achievements would be possible without the endless
love and support of my family and friends. Thanks so much to my husband Sumit
for remaining ever so loving, caring, understanding and supportive even when
facing my displaced frustrations from failed experiments. Our little daughter
Shivangi continues to be a source of enormous joy, love and energy for me. I am
ever so thankful to my brother Baijayanta for his love, support and unwavering
confidence in my abilities through rain or shine. My parents are a source of great
inspiration for me. Their deepest love and devotion continues to brighten each
day of my life. I can never thank them enough.
vi
VITA
1999……………………………….. M.B.B.S., Calcutta Medical College,
University of Calcutta, Calcutta, India.
1999 - present……………………. Graduate Teaching and Research Associate,
The Ohio State University, Columbus, Ohio.
PUBLICATIONS
Research Publication
1. Timmers C, Sharma N, Opavsky R, Maiti B, Wu L, Wu J, Orringer D,
Trikha P, Saavedra HI, Leone G. E2f1, E2f2, and E2f3 control E2F target
expression and cellular proliferation via a p53-dependent negative
feedback loop. Mol Cell Biol. 2007 Jan;27(1):65-78. PMID: 17167174
2. Logan N, Graham A, Zhao X, Fisher R, Maiti B, Leone G, La Thangue NB.
E2F-8: an E2F family member with a similar organization of DNA-binding
domains to E2F-7. Oncogene. 2005 Jul 21;24(31):5000-4. PMID:
15897886
3. Maiti B, Li J, de Bruin A, Gordon F, Timmers C, Opavsky R, Patil K, Tuttle
J, Cleghorn W, Leone G. Cloning and characterization of mouse E2F8, a
novel mammalian E2F family member capable of blocking cellular
proliferation. J Biol Chem. 2005 May 6;280(18):18211-20. Epub 2005 Feb
18. PMID: 15722552
4. de Bruin A, Maiti B, Jakoi L, Timmers C, Buerki R, Leone G. Identification
and characterization of E2F7, a novel mammalian E2F family member
capable of blocking cellular proliferation. J Biol Chem. 2003 Oct
24;278(43):42041-9. Epub 2003 Jul 31. PMID: 12893818
5. Saavedra HI, Maiti B, Timmers C, Altura R, Tokuyama Y, Fukasawa K,
Leone G. Inactivation of E2F3 results in centrosome amplification. Cancer
Cell. 2003 Apr;3(4):333-46. PMID: 12726860
vii
6. Wu L, Timmers C, Maiti B, Saavedra HI, Sang L, Chong GT, Nuckolls F,
Giangrande P, Wright FA, Field SJ, Greenberg ME, Orkin S, Nevins JR,
Robinson ML, Leone G. The E2F1-3 transcription factors are essential for
cellular proliferation. Nature. 2001 Nov 22;414(6862):457-62. PMID:
11719808
FIELDS OF STUDY
Major Field: Molecular Genetics
viii
TABLE OF CONTENTS
Page
Abstract ........................................................................................................ii
Dedication ....................................................................................................iv
Acknowledgment ..........................................................................................v
Vita ... ...........................................................................................................vii
List of Tables ................................................................................................xi
List of Figures ...............................................................................................xii
List of Abbreviations .....................................................................................xv
Chapter 1: Introduction ................................................................................1
E2F overview .....................................................................................2
The mammalian E2F transcription factor family.................................3
E2Fs regulate cellular proliferation ....................................................4
E2F in apoptosis ................................................................................5
E2Fs and cancer................................................................................6
Survivin overview ...............................................................................7
Survivin as an anti-apoptotic protein ..................................................8
Function of survivin in cell division .....................................................10
Survivin in development.....................................................................12
Survivin and cancer ...........................................................................13
Conclusion .........................................................................................15
Tables and figures .............................................................................16
Chapter 2: Cloning and characterization og mouse E2F8, a novel mammalian
E2F family member capable of blocking cellular proliferation .......................25
ix
Abstract..............................................................................................25
Introduction ........................................................................................26
Materials and Methods.......................................................................29
Results...............................................................................................36
Discussion .........................................................................................47
Tables and Figures ............................................................................50
Chapter 3: Identification and characterization of E2F7, a novel mammalian E2F
family member capable of blocking cellular proliferation ..............................63
Abstract..............................................................................................63
Introduction ........................................................................................64
Materials and methods.......................................................................68
Results...............................................................................................69
Discussion .........................................................................................74
Tables and figures .............................................................................77
Chapter 4: The E2F1-3 transcription factors are essential for cellular proliferation
...........................................................................................................83
Abstract..............................................................................................83
Introduction ........................................................................................84
Results and discussion ......................................................................85
Materials and methods.......................................................................89
Tables and figures .............................................................................92
Chapter 5: Survivin is essential for proliferation and collaborates with c-Myc and
H-Ras (V12) in the transformation of mouse embryonic fibroblasts..............98
Abstract..............................................................................................98
Introduction ........................................................................................99
Materials and methods.......................................................................104
Results and discussion ......................................................................107
Tables and figures .............................................................................112
Chapter 6: Discussion .................................................................................125
E2F7 and E2F8 form unique sub-class in the murine E2F family ......125
Survivin is essential for the proliferation and transformation of MEFs126
List of references ..........................................................................................128
x
LIST OF TABLES
Table
4.1
Page
Genotypic analysis of E13.5 embryos derived from crosses of E2F
mutant animals………………………………………………………………………97
xi
LIST OF FIGURES
Figure
Page
1.1
The cell division cycle...................................................................16
1.2
Rb and E2F regulate gene expression .........................................17
1.3
E2F: the family portrait .................................................................18
1.4
Genomic structure of the survivin locus and splice variants .........19
1.5
Structure-function of survivin splice variants ................................20
1.6
Schematic representation of the human inhibitor of apoptois protein
family ............................................................................................21
1.7
Pathways to apoptosis..................................................................22
1.8
Localization and function of survivin (and its fellow passenger proteins)
during mitosis ...............................................................................23
2.1
Structure of mouse E2F8 gene, mRNA and protein .....................50
2.2
Comparative analysis of E2F8 and other known E2F and DP family
members ......................................................................................52
xii
2.3
Tissue-specific and cell cycle-dependent expression of E2F8 .....54
2.4
Regulation of E2F8 promoter .......................................................56
2.5
Subcellular localization and DNA binding activity of E2F8............58
2.6
E2F8 can homodimerize...............................................................60
2.7
E2F8 overexpression inhibits cellular proliferation .......................61
3.1
Tissue-specific and cell cycle-dependent expression of E2F7 .....77
3.2
Cell cycle regulation of the E2F7 promoter...................................79
3.3
E2F7 has transcriptional repressor function .................................81
4.1
E2F3 conditional knock out strategy.............................................92
4.2
Growth defect in E2F3 single or combinatorial knock out MEFs ..94
4.3
Loss of E2F3 activates checkpoint via p21...................................95
5.1
Conditional knock out strategy of survivin in primary mouse embryonic
fibroblasts .....................................................................................112
5.2
Survivin is essential for proliferation but not for survival of primary
MEFs ............................................................................................114
5.3
Primary MEFs lacking survivin display an abnormal nuclear
morphology...................................................................................116
5.4
Loss of survivin results in polyploidy.............................................120
5.5
Survivin is essential for the maintenance of the myc/ras transformed
phenotype in MEFs.......................................................................122
xiii
5.6
Survivin collaborates with c-Myc and H-RasV12 in transforming wt
primary MEFs ...............................................................................124
xiv
ABBREVIATIONS
APC
Adenomatous polyposis coli
APC/C
Anaphase promoting
complex/cyclosome
ATM
Ataxia-telangiectasia-mutated
BIR
Baculovirus IAP repeat
BIRC
Baculoviral IAP-repeat-containing
BrdU
5-bromo-2_-deoxyuridine
CARDs
Caspase-recruitment domains
Cdk
Cyclin dependent kinase
Chk2
Checkpoint kinase 2
cIAP
Cellular IAP (cIAP)
xv
CMV
Cytomegalovirus
CPC
Chromosomal passenger complex
CPP
Chromosomal Passenger Protein
DAPI
4,6-diamidino-2-phenylindole
DBD
DNA binding domain
DBD
DNA-binding domain(s)
DDR
DNA damage response
DHFR
Dihydrofolate reductase
DMEM
Dulbecco’s modified Eagle’s medium
ERK
Extracellular signal-regulated kinase
EST
Expressed sequence tag(s)
FBS
Fetal bovine serum.
GDP
Guanosine diphosphate
g-o-f
Gain of function
GTP
Guanosine triphosphate
HBXIP
Hepatitis B X-interacting protein
HDAC
Histone Deacetylase
HIAP
Human IAP
xvi
IAP
Inhibitor of apoptosis protein
ILP
IAP-like protein
INCENP
Inner centromere protein.
KIAP
Kidney IAP
l-o-f
Loss of function
LRRs
Leucine-rich repeats
MAPK
Mitogen-activated protein kinase
MEF
Mouse embryonic fibroblast(s)
MIH
Mammalian IAP homologue
ML-IAP
Melanoma IAP
NACHT
Domain found in NAIP, CIITA, HET-E
and TP-1
NAIP
Neuronal apoptosis-inhibitory protein
NES
Nuclear export signal
PBS
Phosphate buffered saline
PCR
Polymerase chain reaction
PI
Propidium Iodide
PI3K
Phosphatidylinositol 3-kinases
xvii
Rb
Retinoblastoma
RING
Really interesting new gene
RNAi
RNA interference
RPE
Retinal pigment epithelium
shRNA
Short hairpin RNA
TK
Thymidine kinase
Ts-IAP
Testicular IAP
UBC
Ubiquitin-conjugation
WT
Wild type
XIAP
X-linked IAP
xviii
CHAPTER 1
INTRODUCTION
Cell division is an essential process in normal development, growth,
reproduction and immunity. It also plays an important role in various disease
processes. Consequently, the cell division cycle is highly regulated with a
multitude of proteins orchestrating the sequence of events in a temporal and
spatial manner. To maintain homeostasis, cell division and apoptosis (or
programmed cell death) must be tightly coupled. A disruption in the coordination
of these two processes can lead to aberrant cellular proliferation with the
accumulation of genetic mutations and ultimately to a transformed phenotype.
The E2F family of transcription factors and the anti-apoptotic protein, survivin are
significant molecules that link cellular proliferation and cell death. Signaling
pathways involving these proteins are deregulated and/or mutated in a vast
majority of human cancers and are the focus of this thesis.
1
E2F overview
In mammals, the cell cycle consists of four stages: G1, S, G2 and M
(Figure 1.1). An important event during the G1-S transition of the cell cycle is the
phosphorylation of tumor suppressor protein, Retinoblastoma (Rb) by the various
cdks (cyclin dependent kinases). Rb when hypophosphorylated binds to the E2F
family of transcription factors thus inactivating them. Phosphorylated Rb no
longer binds E2F. Consequently Rb-mediated repression is lifted and the E2F
target genes can be expressed (Dyson, 1998). The E2F target genes play
important roles in a variety of cellular processes like DNA replication, DNA repair,
mitosis, apoptosis and cell cycle checkpoint functions and differentiation
(DeGregory and Johnson, 2006) (Figure 1.2). Consequently, deregulated E2F
activities result in developmental defects and cancer (DeGregory and Johnson,
2006).
The mammalian E2F transcription factor family
The mammalian E2F transcription factors arise from eight distinct genes,
E2F1 – 8. These loci give rise to at least ten different transcripts with two
isoforms each from the E2F3 and E2F7 genes. The highly conserved DNA
binding domain is shared among all the family members and enables the E2Fs to
2
bind a consensus DNA binding element present on its target gene promoters
(DeGregory and Johnson, 2006; Tsantoulis and Gorgoulis, 2005). E2F7 and
E2F8 are unique in possessing duplicated DNA binding domains. (Christensen et
al., 2005; Logan et al., 2005; Maiti et al., 2005 ∗ ; Logan et al., 2004; DiStefano et
al., 2003; deBruin et al., 2003 ∗ ). A C-terminal transactivation domain, in which is
embedded the Rb-binding region, is present in E2Fs 1-5. The E2Fs 1-6 have a
leucine-zipper dimerization domain through which they bind to their dimerization
partner, DP. This dimerization enables high-affinity DNA binding as well as
regulates the subcellular localization pattern of E2F (Figure 1.3) (DeGregory and
Johnson, 2006; Tsantoulis and Gorgoulis, 2005). E2F activities are also
regulated at the levels of transcription, post translational modifications like
acetylation and phosphorylation, co-factor binding and ubiquitination (Nevins et
al., 1998, Trimarchi and Lees, 2002).
Functionally, E2Fs have been classified as ‘activators’ or ‘repressors’
depending on their action on target promoters and their association with these
promoters during transcriptionally active or repressed states. The ‘activator’ E2Fs
(E2F 1-3) associate with the target gene promoters during active transcription
(G1-S phase of cell cycle), while ‘repressor E2Fs (E2F4-5) associate with the
target genes during early G1 and quiescence. E2F4-5 repress genes at least in
part via recruiting Rb, p130, p107 (the pocket proteins) and their associated co-
∗
These two papers, reporting for the first time, the cloning and characterization of mouse E2F7 and E2F8,
form part of this thesis since they have significant contributions by Maiti B.
3
repressors like HDAC (Histone Deacetylase), BRM/BRG1 (Chromatin remodeling
complex) and SUV39H (Histone methylation machinery), while E2F6 represses
genes by complex formation with either the polycomb group of proteins or with
Mga and Max proteins (Trimarchi et al., 2001; Ogawa et al., 2002). The
mechanism of repression by E2F7 and E2F8 is unclear at this point. Additionally,
the activator E2Fs as well as E2F7 and E2F8 show a cell cycle regulated
oscillation in expression, while no such variation is observed for the repressor
E2Fs 4 and 5 (DeGregory and Johnson, 2006).
E2Fs regulate cellular proliferation
E2Fs have been shown to play an important role in cell cycle progression
as well as cell cycle arrest through gene disruption (knockout) and
overexpression. E2F1 overexpression has been shown to promote S-phase
induction in mouse embryonic fibroblasts (Wang et al., 1998). Accordingly, its
deficiency leads to impaired cell cycle progression in activated T-cells
(DeGregory and Johnson, 2006). Combinatorial knockouts of E2F1 and E2F2
lead to defective proliferation of hematopoietic progenitors and exocrine
pancreas (DeGregory and Johnson, 2006). While E2F1-/-E2F3-/- and E2F2-/E2F3-/- MEFs have severe proliferation defects, E2F1-/-E2F2-/-E2F3-/- MEFs
arrest in every stage of the cell cycle (Wu et al., 2001). On the other hand, MEFs
deficient in the ‘repressor’ E2Fs, E2F4 and E2F5 fail to arrest despite
4
overexpression of p16Ink4a (a cyclin dependent kinase inhibitor leading to
hypophosphorylated Rb) indicating a requirement for ‘repressor’ E2F mediation
of pocket protein functions (DeGregory and Johnson, 2006). Generally speaking,
the ‘activator’ E2Fs, which transactivate the target genes, are ‘pro-proliferative’,
while ‘repressor’ E2Fs, which silence target genes, inhibit proliferation.
Accordingly, E2F7 and E2F8 have also been shown to inhibit cellular proliferation
and repress known E2F targets (Tsantoulis and Gorgoulis, 2005; DeGregory and
Johnson, 2006).
E2F in Apoptosis
E2F1 is the most extensively studied E2F family member in apoptosis.
E2F1 mediates apoptosis via the p53 and p73 pathways (DeGregory and
Johnson, 2006). p53 dependent apoptosis is mediated via the ARF gene, which
is a transcriptional target of E2F1. ARF prevents mdm2 mediated p53
degradation and leads to stabilization of p53. E2F1 also mediates the DNA
damage response (DDR) checkpoint due to DNA damage induced by radiation or
by the intercalating agent adriamycin. Phosphorylation of E2F1 by ATM (Ataxiatelangiectasia-mutated), Rad3-related (ATR) kinases and checkpoint kinase 2
(Chk2) leads to its stabilization, following DNA damage (Lin et al., 2001; Stevens
et al., 2003). E2F1 in turn can lead to ATM dependent phosphorylation of p53,
leading to its stabilization and activation, and hence apoptosis (Lin et al., 2001;
5
Powers et al., 2004; Rogoff et al., 2004). The importance of E2F1 mediated
apoptosis is evident in vivo by the reduced apoptosis of thymocytes leading to
impaired negative selection in E2F1-/- mice. Besides E2F1, E2F3 is also capable
of inducing apoptosis when overexpressed (DeGregory and Johnson, 2006).
E2Fs and cancer
Deregulated E2F activity, as a result of mutations in Rb itself or in
members of its signaling pathway, including p16Ink4a (a cyclin dependent kinase
inhibitor), and Cyclin D is a hallmark of virtually every human cancer.
Additionally, amplification of E2F1, E2F3 or E2F5 has been documented in
retinoblastoma, bladder, lung, ovarian and prostate, gastrointestinal cancers and
metastatic melanomas (Johnson and DeGregory, 2006). Paradoxically,
overexpression or amplification of the same E2F family member could lead to a
favorable or unfavorable prognosis, perhaps depending on the tissue involved
and associated mutations (Johnson and DeGregory, 2006).
All the E2F family members that associate with Rb can lead to oncogenic
transformation in vitro, when overexpressed (Johnson and DeGregory, 2006).
Transgenic mice overexpressing E2F1, E2F3 and E2F4 in keratinocytes also
show increased tumorigenicity (Johnson and DeGregory, 2006). E2Fs are
6
relevant targets downstream of mutated Rb in oncogenesis, as shown by their
disruption leading to decreased tumor formation (Johnson and DeGregory,
2006). Some transgenic studies using mice deficient in E2F1, E2F2 or E2F3
reveal a tumor suppressive function for those E2F family members. Current
evidence suggests that the tumor suppressor function of E2Fs have both p53
and apoptosis-dependent and -independent arms (Johnson and DeGregory,
2006). In case of E2F3, this could be related to genomic instability consequent to
the loss of E2F3 (Saavedra et al., 2003). Thus the role of E2Fs in cancer is
context specific and definitely warrants a thorough understanding and careful
approach especially when designing drugs to target E2Fs in cancer therapy.
Survivin overview
Survivin, a direct transcriptional target of E2F, is a 16.5 kDa protein with
dual functions in cellular proliferation and cell death (Altieri, 2003; Lens et al.,
2006). In addition to its essential role as a chromosomal passenger protein
(CPP) during mitosis, survivin prevents cell death by interfering with both
caspase dependent and independent pathways (Wheatley et al., 2005). Analysis
of the spatial and temporal expression of survivin reveals an ‘oncofetal’ pattern
whereby it is ubiquitous during development, undetectable in most terminally
differentiated adult tissues and highly re-expressed in virtually every human
cancer (Altieri, 2006). Preferential expression of survivin in human cancers
7
makes it a potential diagnostic tool, prognostic marker as well as a promising
therapeutic target.
The survivin gene is located on chromosome 17q25 (human) or 11E2
(mouse). The human locus gives rise to at least five alternatively spliced survivin
transcripts (Figure 1.4) Besides the wild type survivin (142 amino acids), survivin2B (165 amino acids) results from an alternative exon 2 insertion; survinin-ΔEx-3
(137 amino acids) arises from the exon 3 skipping resulting in a frameshift; and
survivin-3B (120 amino acids) results from the novel exon 3B insertion (Figure
1.5) (Caldas et al., 2005). These isoforms help fine-tune survivin function and
attribute more functional diversity to this gene.
Survivin as an anti-apoptotic protein
Survivin is the smallest member of the mammalian inhibitor of apoptosis
(IAP) gene family, containing a single Baculovirus IAP repeat (BIR), a ~ 70amino-acid zinc-finger fold but no RING domain (Altieri, 2003). In humans eight
IAPs have been identified so far, namely, cIAP-1, cIAP-2, XIAP, ILP-2, NAIP, MLIAP, apollon and survivin. IAPs are evolutionarily conserved from yeast to
humans and function to protect the cell from the fortuitous activation of the
caspase cascade and subsequent self-destruction (Zangemeister-Wittke et al.,
2004) (Figure 1.6)
8
Mounting evidence in the field has unequivocally proven the anti-apoptotic
function of survivin in both normal tissue and cancer. Inactivation of the survivin
signaling by molecular antagonists like siRNA sequences or dominant negative
mutants result in increased apoptosis in vitro and in vivo (Altieri, 2003).
Mechanistically, Dohi et al., (2004) showed that following the addition of proapoptotic stimuli, a tumor cell specific mitochondrial pool of survivin is mobilized
rapidly to the cytosol, where it blocks cell death at least in part by interfering with
caspase-9 processing. Adenovirus mediated expression of dominant-negative
survivin increases apoptosis in tumor cells and suppresses angiogenesis (BlancBrude et al., 2003). Furthermore, adenovirus mediated mitochondrial expression
of survivin accelerates xenograft tumor growth in SCID mice and abrogates
tumor cell apoptosis (Dohi et al., 2004; Li and Ling 2006).
From a mechanistic viewpoint, survivin inhibits both caspase dependent
and caspase independent apoptosis initiated via the extrinsic or intrinsic cellular
apoptotic pathways. (Figure 1.7) As reviewed by Altieri (2003), cell death caused
by inhibiting survivin signaling, exhibits features of mitochondrial dependent
apoptosis with cytochrome c release, caspase-9 activation and activation of the
caspase cascade. Survivin prevents cell death by binding to caspase-9 in the
cytoplasm, in the presence of the HBXIP (Hepatitis B X-interacting protein) or by
interaction with XIAP or Smac/DIABLO (a pro-apoptotic protein released from the
mitochondria) (Zangemeister-Wittke et al., 2004). Pan caspase inhibitors only
partially rescue apoptosis induced by overexpression of a survivin dominant
9
mutant (T34A), indicating the involvement of a caspase independent pathway by
survivin (Liu et al., 2004).
Function of survivin in cell division
Disruption of survivin activity, using dominant negative mutants or
survivin-specific siRNAs also results in mitotic defects, in a multitude of normal
and tumor cell types studied. These defects are characterized by multipolar
mitotic spindles, misseggregation of chromosomes, polyploidy, multinucleation
and failure of cytokinesis (Altieri, 2003). Consistent with its role in mitosis,
survivin is predominantly expressed during this period in a cell cycle regulated
fashion and has been shown to physically associate with the mitotic apparatus
(Altieri, 2006).
Survivin forms a chromosomal passenger complex (CPC) with INCENP
(inner centromere protein), Borealin/Dasra-B and the kinase subunit Aurora-B
(Vader et al., 2006b). The chromosomal passenger proteins (CPP), survivin,
INCENP and Borealin regulate the proper and timely localization of the active
enzymatic core of the complex, Aurora-B. The CPC shows a very dynamic
localization pattern during mitosis. At prophase it is present diffusely along
chromatin, progressing to the inner kinetochore (site of spindle microtubule
attachment) during prometaphase, to the central spindle during anaphase and
the midbody during cytokinesis (Vader et al, 2006b). This dynamic localization of
10
the CPC correlates with its activity at the different sites, namely, histone
modification, destabilization of improper chromosome-spindle attachments,
maintaining the spindle assembly checkpoint (SAC) in a BubR1 and Mad2
(spindle checkpoint proteins) dependent fashion, thus promoting correct bioriented chromosome-spindle attachment and finally cytokinesis (Lens et al.,
2006; Vader et al., 2006b; Lens et al., 2003) (Figure 1.8). In case of an improper
kinetochore-microtubule interaction (for example kinetochores attached to
microtubules from the same spindle pole or syntelic attachment), survivin and
Aurora-B sense the lack of tension at the kinetochore, and signal directly to the
microtubule, resulting in the activation of a spindle checkpoint by influencing
affinity of checkpoint protein BubR1 to the kinetochore (lens et al., 2003; Lens
and Medema, 2003). Thus, survivin and Aurora-B are crucial for ensuring highfidelity chromosomal segregation during mitosis. Vader et al., (2006a) targeted
the individual members of the CPC in order to further pinpoint their contributions.
Their work revealed that while INCENP stabilized the whole CPP complex and
Borealin promoted survivin binding to INCENP; even in the absence of Borealin,
survivin could localize Aurora-B to the centromere and midbody.
Extensive biochemical work is ongoing to understand the functions of the
different domains or residues of the survivin molecule. Vong et al., (2005)
showed that the centromeric localization of survivin in a precise timely manner
was orchestrated by ubiquitination and deubiquitination of survivin on the Lys(63)
residue by the Ufd1 and hFAM proteins. Recently, Colnaghi et al., (2006) showed
11
that a point mutation (L98A) of the NES (Nuclear export signal) in the survivin
molecule was able to separate the anti-apoptotic and mitotic functions of survivin,
perhaps secondary to its nuclear localization pattern. The NES mutant of
survivin, which is deficient in CRM1/exportin pathway mediated nuclear export, is
still capable of carrying out the mitotic functions but is unable to serve as an antiapoptotic protein (Colnaghi et al., 2006).
Survivin in development
Survivin plays an essential role in normal embryonic development as
evidenced by the early embryonic death of the survivin-/- embryos around day
4.5 post coitum (Uren at al., 2000; Conway et al., 2002). The survivin null
embryos exhibited irregular nuclear morphology, multinucleation, giant cell
formation and microtubule bundling, consistent with a defect in microtubule
organization and cytokinesis (Uren et al., 2000). This was a phenocopy of
INCENP null embryos, suggesting the requirement for survivin as a chromosomal
passenger protein at this stage of development (Uren et al., 2000). The antiapoptotic role of survivin is also relevant in embryogenesis as elucidated by
Jiang et al., (2004), where neuronal precursor cells lacking survivin underwent
massive apoptosis resulting in an immature brain structure at birth. Additionally,
hepatocytes from survivin +/- mice exhibit an increased apoptotic response to
suboptimal activation of Fas ligand mediated cell death signaling (Conway et al.,
12
2002; Altieri, 2003). Thus, survivin’s role in normal development is very likely a
combination of its mitotic and cytoprotective functions.
Survivin expression is undetectable in a vast majority of adult tissues with
a few exceptions of rapidly dividing tissues, as hematopoietic stem/progenitor
cells, T-lymphocytes, vascular endothelium and gastric epithelium (Li And
Brattain, 2006; Fukuda et al., 2006). Survivin plays an important role in
hematopoietic cells, T-lymphocyte proliferation survival and maturation. Okada et
al., (2004) showed that conditional deletion of survivin in T cell lineage resulted in
a differentiation block from CD4-CD8- double negative to CD4+CD8+ double
positive T-lymphocytes, in addition to a proliferation defect in these cells. Survivin
also plays an important role in spermatogenesis, wound healing and liver
regeneration in adults (Li And Brattain, 2006). Thus, survivin plays a crucial role
in the proliferation, survival and maturation of cells during the entire mammalian
lifespan.
Survivin and cancer
Survivin is a ‘cancer gene’ and the top 4th ‘transcriptome’ preferentially
expressed in transformed cells over the normal tissue (Altieri, 2006). Aberrantly
high levels of survivin have been documented in virtually every human
malignancy namely, central nervous system tumors (medulloblastoma,
glioblastoma, astrocyte tumors), laryngeal, esophageal, breast, non-small cell
13
lung, gastric, pancreatic, hepatocellular, renal, endometrial, cervical cancers, as
well as osteosarcoma, soft tissue sarcoma, melanoma, leukemias and
lymphomas (Fukuda et al., 2006). Increased survivin expression corelates with a
more aggressive tumor phenotype in terms of increased proliferation, and higher
radio- and chemotherapy resistance, reflecting of the mitotic and anti-apoptotic
functions of survivin. The nuclear versus cytoplasmic subcellular localization of
survivin in cancer cells has also been associated with clinical outcome, including
patient survival and tumor recurrence (Li et al., 2005; Altura et al., 2003). The
cancer specificity combined with undetectable expression in terminally
differentiated normal adult tissue makes survivin a highly explored molecular
target for cancer therapy.
Targeting survivin with siRNA in cancer cell lines results in increased
apoptosis and heightened chemo- and radiosensitivity (Sommer et al., 2003).
Survivin expression is positively regulated by multiple oncogenic pathways like
E2F-Rb (Jiang et al., 2004), constitutively activated c-H-Ras (Sommer et al.,
2003) and STAT3 (Kanda et al., 2003), and is also negatively regulated by
various tumor suppressor pathways like p53 (Hoffman et al., 2003) and
adenomatous polyposis coli (APC) (Zhang et al., 2001; Altieri, 2006). Thus,
survivin is a fascinating little protein, which is at the crossroads of the multiple
cancer related pathways and arms the cancer cell with pro-proliferative and antiapoptotic properties.
14
Conclusion
In summary, the Rb-E2F and survivin signaling pathways overlap and yet
play very distinct roles in cell division, differentiation, apoptosis and
transformation. The chapters that follow elucidate the cloning and
characterization of a novel mammalian E2F family member E2F8 and the
contribution of survivin in the proliferation and transformation of primary mouse
embryonic fibroblasts (MEFs). This thesis makes an effort towards understanding
the roles played by the E2F family of transcription factors and survivin in the
regulation of cellular proliferation and transformation in mammals.
15
Go (serum starved) cells
M
(Mitosis)
G1
G2
S
(DNA synthesis)
Figure 1.1
The cell division cycle
The cell cycle is comprised of G1, S, G2 and M phases.
16
CDK
P
P
Rb
Rb
P
E2F
G0 quiescent
Proliferating
E2F
Target Gene Expression
DNA replication, cell cycle control, DNA repair, mitosis
Figure 1.2
Rb and E2F regulate Gene expression
Following phosphorylation by cyclin dependent kinases (cdk) Rb can no
longer bind E2Fs ensuring the proper transition from G1 to S phase of
cell cycle.
17
Figure 1.3
E2F: the family portrait.
The highly conserved winged-helix DNA binding domain (DBD) is indicated
in blue, and the hydrophobic heptad repeat domain required for
dimerization is shown in pink. The DBD also appears to contribute to
dimerization. Other domains required for associations with cyclin A/cdk2
and Rb family members are also indicated. E2F1- E2F6 all bind DNA as
heterodimers with DP family proteins, while E2F7 and E2F8 appear to
associate with DNA independent of DP proteins. Note also that mouse
homologs of human DP-2 are usually referred to as DP-3, and there are
several differentially spliced isoforms of DP2/DP3 (not shown). There are
also two known isoforms of E2F7 which differ in their Ctermini (also not
shown).
Adapted from DeGregory and Johnson, 2006
18
Figure 1.4
Genomic structure of the Survivin locus and splice
variants.
The Survivin gene is composed of exons 1–4. All identified
Survivin isoforms contain exons 1 and 2. Survivin-2B has an
extra exon 2B; Survivin-deltaEx3 is missing exon 3;
Survivin-3B contains 5 exons including an extra novel exon
3B derived from a 165 bp long portion of intron 3; and
Survivin-2a consists of 2 exons (exons 1 and 2) as well as a
30 197 bp region of intron 2.
Adapted from Fangusaro et al., 2006
19
Figure 1.5
Structure−function of survivin splice variants.
Common features to all splice forms at the amino terminus include
the dimer interface and the BIR domain. Survivin-2B differs by the
introduction of exon 2B but retains the ORF for the remainder of the
C-terminus. The BH2 domain and mitochondrial localization signal of
survivin-Ex3 are also indicated. A frameshift occurs in exon 3 of
survivin 3B causing a premature stop of the protein with a distinct Cterminus
Adapted from Caldas et al., 2005
20
Figure 1.6
Schematic representation of the human inhibitor of
apoptosis protein family.
In addition to at least one baculoviral IAP repeat (BIR) domain,
most IAPs have other distinct functional domains. The really
interesting new gene (RING) domain found in many IAPs is an
E3 ligase that presumably directs targets to the ubiquitinproteasome degradation system. Caspase-recruitment domains
(CARDs) can mediate homotypic protein–protein interactions,
although the binding partners for the cellular IAP (cIAP) CARDs
have not yet been elucidated. Bruce has a ubiquitin-conjugation
(UBC) domain that is found in many ubiquitin-conjugating
enzymes. The NACHT domain of neuronal apoptosis-inhibitory
protein (NAIP) resembles a nucleotide-oligomerization domain
related to the AAA+ NTPases, whereas the leucine-rich repeats
(LRRs) are similar to those of the Toll-like receptors that function
as pathogen sensors. BIRC, baculoviral IAP-repeat-containing;
HIAP, human IAP; IAP, inhibitor of apoptosis protein; ILP, IAPlike protein; KIAP, kidney IAP; MIH, mammalian IAP homologue;
ML-IAP, melanoma IAP; NACHT, domain found in NAIP, CIITA,
HET-E and TP-1; Ts-IAP, testicular IAP; XIAP, X-linked IAP.
Adapted from Eckelman et al., 2006
21
Figure 1.7
Pathways to Apoptosis.
Activation of cell death pathways can be initiated through different
mechanisms, including through ligand binding (FasL, TNF) to a
death receptor on the cell surface (extrinsic pathway) or via direct
mitochondrial signaling (intrinsic pathway). The mitochondrial
pathway is initiated by activation of the Bax/Bcl-2 pathway leading
to the release of apoptotic factors such as cytochrome c (Cyt C)
and apoptosis-inducing factor (AIF) from the mitochondrial
intermembrane space into the cytoplasm. The release of
cytochrome c from mitochondria results in caspase-3 activation
through formation of the cytochrome c/Apaf-1/caspase-9
apoptosome complex. Caspase-3 cleaves a number of substrates
including cytoskeletal proteins andDNA. Caspase-activated DNase
(CAD) and inhibitor of CAD (ICAD) initiate cleavage and
fragmentation ofDNA. The Inhibitors of Apoptosis (IAPs) inhibit cell
death by physically interacting with caspases. Survivin (SVN) has
been shown to inhibit apoptosis through caspase-dependent and
independent pathways.
Adapted from Fangusaro et al., 2006
22
Figure 1.8
Localization and function of Survivin (and its fellow
passenger proteins) during cell division.
(a) In (pro)metaphase, Survivin localizes on centromeres and
chromosome arms (not visible). During anaphase, Survivin no
longer associates with centromeric DNA but binds to the
overlapping bundles of antiparallel microtubules of the central
spindle that form the midzone. These microtubules bundles
become compact and mature into the midbody during telophase
and cytokinesis, where Survivin eventually localizes. (b) When
localized on centromeres, the CPC, of which Aurora B is the
enzymatic core and Survivin an important targeting subunit,
executes different functions that are important for proper
chromosome alignment and segregation: (i) destabilization of
improper kinetochore–microtubule attachments and thereby
promoting biorientation; (ii) regulation of BubR1 kinetochore
levels; (iii) keeping the spindle checkpoint active in the absence
of tension by recreating unattached kinetochores and/or by
regulating BubR1 kinetochore levels; and (iv) stabilizing
chromosome/kinetochore-induced microtubule formation. (v)
When present on the central spindle and midbody, the complex
is essential for completion of cytokinesis by phosphorylating
several substrates (e.g. MKLP1 and vimentin) involved in
midzone and midbody function. Abbreviation: MT, microtubule.
Adapted from Lens et al., 2006
23
Figure 1.8
Localization and function of Survivin (and its fellow passenger
proteins) during cell division.
24
CHAPTER 2
CLONING AND CHARACTERIZATION OF MOUSE E2F8, A NOVEL
MAMMALIAN E2F FAMILY MEMBER CAPABLE OF BLOCKING CELLULAR
PROLIFERATION 1
ABSTRACT
The E2F transcription factor family plays a crucial and well established
role in cell cycle progression. Deregulation of E2F activities in vivo leads to
developmental defects and cancer. Based on current evidence in the field,
mammalian E2Fs can be functionally categorized into either transcriptional
activators (E2F1, E2F2, and E2F3a) or repressors (E2F3b, E2F4, E2F5, E2F6,
and E2F7). As part of my thesis, I have identified a novel E2F family member,
E2F8, which is conserved in mice and humans and has its counterpart in
Arabidopsis thaliana (E2Ls). Interestingly, E2F7 and E2F8 share unique
structural features that distinguish them from other mammalian E2F repressor
1
Reproduced with permission from J Biol Chem. 2005 May 6;280(18):18211-20. Epub 2005 Feb
18. Maiti B, Li J, de Bruin A, Gordon F, Timmers C, Opavsky R, Patil K, Tuttle J, Cleghorn W,
Leone G. © 2005 by The American Society for Biochemistry and Molecular Biology, Inc. All data
were generated by Maiti B, under the supervision of Leone G, except data pertaining to in silico
structural analysis and E2F8 co-immunoprecipitation.
25
members, including the presence of two distinct DNA-binding domains and the
absence of DP-dimerization, retinoblastoma-binding, and transcriptional
activation domains. Similar to E2F7, overexpression of E2F8 significantly slows
down the proliferation of primary mouse embryonic fibroblasts. These
observations, together with the fact that E2F7 and E2F8 can homodimerize and
are expressed in the same adult tissues, suggest that they may have overlapping
and perhaps synergistic roles in the control of cellular proliferation.
INTRODUCTION
The E2F transcription factor family plays a crucial and well established
role in the regulation of cellular proliferation differentiation and apoptosis.
Although E2Fs can act as transcriptional activators, they can also repress gene
expression when bound to the retinoblastoma (Rb) tumor suppressor protein or
related pocket proteins (p130 and p107). Rb negatively regulates the E2F
transcriptional activity by binding to and masking the transactivation domain of
E2F (Helin et al., 1993; Flemington et al., 1993). When bound to E2F, Rb can
also directly repress E2F-target genes by recruiting chromatin-remodeling
complexes and histone-modifying activities to the promoter (Vandel et al., 2001;
Sellers et al., 1995; Luo et al., 1998; Brehm et al., 1998; Zhang et al., 2000;
Ferreira et al., 1998; Trouche et al., 1997). During the G1-S transition of the cell
cycle, the cyclin-dependent kinases phosphorylate Rb, resulting in the release of
26
E2F from Rb-containing complexes and the expression of E2F-target genes.
Many studies indicate E2F as a key mediator of Rb function in cellular
proliferation, differentiation, and apoptosis (Dyson, 1998; Nevins, 1998). E2Fs
are also regulated at the level of transcription, post-translational modifications,
subcellular localization, association of cofactors, and degradation (Nevins et al.,
1998; Trimarchi and Lees, 2002).
Since the identification of the founder E2F family member, E2F1, six
additional family members have been identified. Structurally, E2F1–6 share a
highly conserved DNA-binding and dimerization domain. E2F1–6 bind DNAtarget sequences as heterodimers with DP1 or DP2. Although heterodimerization
is required for high affinity sequence-specific DNA binding, the specificity of the
dimer is determined by the E2F component (Dyson, 1998; Nevins, 1998;
Trimarchi and Lees, 2002). The most recently identified E2F family member,
E2F7, is unique in having a duplicated conserved E2F-like DNA-binding domain
and in lacking a DP-dimerization domain. E2F1–5 possess conserved
transactivation and pocket protein-binding domains at the C terminus that are
absent in E2F6 and E2F7 (Dyson, 1998; Ren et al, 2002; de Bruin et al, 2003; Di
Stefano et al., 2003; Logan et al., 2003). Based on structural and functional
considerations, E2Fs have been classified as either “activator” (E2F1, E2F2, and
E2F3a) or “repressor” E2Fs (E2F3b, E2F4, E2F5, E2F6, and E2F7). Activator
E2Fs are maximally expressed late in G1 and can be found in association with
E2F-regulated promoters during the G1-S transition, coinciding with the
27
activation of many E2F-target genes. Mouse embryonic fibroblasts ablated for all
of the three activator E2Fs are severely compromised in E2F-target gene
expression as well as the capacity to proliferate, underscoring the importance of
the activator subclass in cell cycle progression (Wu et al., 2001). In contrast to
the activator E2Fs, E2F3b, E2F4, and E2F5 are expressed in quiescent cells and
can be found associated with E2F-binding elements on E2F-target promoters
during G0 phase (Trimarchi and Lees, 2002; Ren et al., 2002; Leone et al., 1998;
Takahashi et al., 2000; Schwede et al., 2003). Among the repressor subclass,
E2F6 mediates repression via recruitment of the Polycomb group of proteins or
complexes containing Mga and Max proteins (Ogawa et al., 2002; Trimarchi et
al., 2001). Although the most recently identified E2F7 protein can function as a
repressor independent of DP interaction, its exact mechanism of mediating
repression is not understood (de Bruin et al, 2003; Di Stefano et al., 2003; Logan
et al., 2003).
E2Fs regulate the transcription of a multitude of genes involved in DNA
replication, cell cycle regulation, chromatin assembly and condensation,
chromosome segregation, DNA repair, and checkpoint control (Dyson, 1998;
Nevins, 1998; Trimarchi and Lees, 2002; Ren et al., 2002). Consistent with its
role in various important cellular processes, E2Fs are evolutionarily conserved
among plants and animals with the exception of yeast. Drosophila melanogaster
has two E2F genes named dE2F1, which acts as an activator, and dE2F2, which
functions as a repressor. Each of these E2Fs heterodimerize with a single DP
28
protein found in Drosophila called dDP (Stevaux et al., 2002). E2F-like activities
have also been found in Xenopus laevis and Caenorhabditis elegans (Ceol et al.,
2001; Suzuki et al., 2000). In the plant, Arabidopsis thaliana, six E2Fs (AtE2Fa-f)
and two DPs (AtDPa-b) have been described. Of these, AtE2Fa-c are
reminiscent of the mammalian activator E2F1–3a subclass and E2Fd-f resemble
the recently discovered E2F7 in that they possess a duplicated DNA-binding
domain and exhibit repressor function. Besides human and mouse, Arabidopsis
is the only other species where E2Fs with duplicated DNA-binding domain have
been described (Mariconti et al., 2002; Kosugi et al., 2002).
As a primary focus if my thesis project, I have identified and characterized
another mouse E2F family member called E2F8, which closely resembles E2F7.
E2F8 is expressed in a cell cycle regulated fashion, has duplicated DNA-binding
domains that are essential for binding to consensus E2F-binding DNA elements,
represses E2F-target genes, and negatively influences cellular proliferation.
MATERIALS AND METHODS
Cloning of the E2F8 cDNA—The mouse testis cDNA library (Origene) was
screened using the primer pairs 5’-AGTACCCCAACCCTGCTGTGAATA- 3’/5’GGACTTGTCTTTGCGGCTGTTTAC-3’ and 5’ CCGGCACAACCTCACCAAAAC3’/5’-TCCCCCGCGTAGAGAAGAGG-3’. Each primer pair resulted in a single
sharp band in three wells that were positive in 96-wells of the master plate.
29
Subsequently, I screened the subplates to obtain the clone with the full-length
E2F8 cDNA. A 5XMyc tag was subcloned in between the BamH1 and EcoR1
sites of the pcDNA3.1/HisB (Invitrogen) vector in-frame with the His6 tag. Next
the 2583-bp open reading frame of E2F8 was subcloned in frame with the His
and Myc tags, thus generating a construct with full-length E2F8 ORF tagged with
His6 and 5X Myc tag at the N terminus.
Generation of Mouse Embryonic Fibroblasts and in Vitro Cell Culture—
Primary mouse embryonic fibroblasts (MEFs) were generated using standard
techniques as described before (Wu et al., 2001). All of the cell lines were grown
in DMEM with 15% FBS. Cells were starved in DMEM containing 0.2% FBS
when they were ~70% confluent. After starving for 48–60 h, they were stimulated
to grow with DMEM containing 15% FBS. Cells were harvested at different time
points after serum stimulation for BrdUrd incorporation assay, Northern blot
assay, and realtime RT-PCR.
BrdUrd Incorporation Assay—BrdUrd was added to the medium, and cells
were incubated with BrdUrd for 2 h being harvested and fixed in
methanol/acetone 1:1. Cells were stained in 35-mm dishes with anti- BrdUrd
primary antibody (NA-61 from Oncogene) and anti-mouse rhodamine secondary
antibody and counterstained with 4’,6-diamidino-2- phenylindole (DAPI). At least
400 cells/35 mm plate were counted.
Northern Blot Assay—For the cell cycle Northern Blot analysis, total RNA
was isolated from MEFs using TRIzol (Invitrogen) and mRNA was subsequently
30
purified using PolyATract mRNA isolation system (Promega). Poly(A) mRNA was
separated on a 1% agarose gel containing 6% formaldehyde and transferred
onto a GeneScreen membrane (PerkinElmer Life Sciences). The mouse tissue
Northern blot was purchased from Origene. The 3’ 1850 bp of the E2F8 cDNA
including the 3_-untranslated region (UTR) was used as a probe for both
Northern blot analyses. The probe was radiolabeled with 50 μCi of [α-32P]dCTP
using Prime-It RmT (Stratagene). Hybridization was carried on overnight under
high stringency conditions (5X saline/ sodium phosphate/EDTA, 50% formamide,
5X Denhardt’s solution, 1% SDS at 42 °C) and washed several times (0.2X SSC,
0.2% SDS at 65 °C) before autoradiography.
Real-time RT-PCR—Approximately 1X106 cells were harvested at the
indicated time point, and total RNA was isolated using the Qiagen RNA Miniprep
column as described by the manufacturer, including a DNase treatment before
elution from the column. Reverse transcription of 2 μg of total RNA was
performed by combining 1 μl of Superscript III reverse transcriptase (Invitrogen),
4 μl of 10X buffer, 0.5 μl of 100 mM oligo(dT) primer, 0.5 μl of 25 mM dNTPs, 1.0
μl of 0.1 M dithiothreitol, 1.0 μl of RNase inhibitor (Roche Applied Science), and
water up to a volume of 20 μl. Reactions were incubated at 50 °C for 60 min and
then diluted 5-fold with 80 μl of water. Real-time RT-PCR was performed using
the Bio-Rad iCycler PCR machine. Each PCR reaction contained 0.5 μl of cDNA
template and primers at a concentration of 100 nM in a final volume of 25 μl of
SYBR Green reaction mixture (Bio-Rad). Each PCR reaction generated only the
31
expected amplicon as shown by the melting-temperature profiles of the final
products and by gel electrophoresis. Standard curves were performed using
cDNA to determine the linear range and PCR efficiency of each primer pair.
Reactions were done in triplicate, and relative amounts of cDNA were normalized
to GAPDH. The sequences of the primer pair used for the E2F8 cDNA were 5’
CCGGCACAACCTCACCAAAAC-3’ and 5’-TCCCCCGCGTAGAGAAGAGG- 3’.
Primer sequences of the E2F-target genes are available upon request.
5’-RACE PCR—Total RNA was isolated from wild-type primary MEFs and
mouse thymus, and the mouse testis cDNA was purchased in the Marathon
Ready cDNA kit (Clontech). cDNA was prepared, and 5’-RACE PCR was
performed using the BD SMART RACE cDNA amplification kit (BD Biosciences)
following the manufacturer’s protocol. The reverse primer used for 5’-RACE PCR
was 5’-TCACGCGTAAGGACTTGTCTTTGC- 3’ mapping to the exon 6 of E2F8
gene, and the nested primer was 5’ CGTCCCGAGGGTTTTGGTGAGGTT-3’
located in the exon 5 of the E2F8 gene. The products of 5’-RACE PCR were
cloned using the TOPO TA cloning kit for sequencing (Invitrogen). At least 20
colonies from each tissue type were sequenced.
Luciferase Reporter Assay—A 3.5-kb promoter fragment was isolated
from the BAC clone RPCI 24-294G9 and subcloned into pBluescript vector. The
primer pair used for amplifying the long promoter (LP) fragment was 5’
GAGAGAGGTACCGTCCTCCAACCCCTCGTTTG-3’ and 5’
AGAGAGAAGCTTGCTGAAGTTTCTCGCCTGACAC-3_ and that for amplifying
32
the short promoter (SP) fragment was 5’
GAGAGAGGTACCAGCTCTGAAGGAGGATTGACAGG- 3’ and 5’AGAGAGAAGCTTGCTGAAGTTTCTCGCCTGACAC- 3’. The amplified
fragments were subcloned into the pGL2Basic vector using the HindIII and KpnI
restriction sites. The two E2F-binding consensus sites in the E2F8 promoter
fragment were mutated using the QuikChange site-directed mutagenesis kit as
described by the manufacturer. All of the constructs were confirmed by
sequencing. Subconfluent REF52 cells grown in triplicates were transfected
using the Superfect reagent (Qiagen). The cells were transfected with the firefly
luciferase expression vectors and thymidine kinase (TK) Renilla luciferase as
internal control. The cells then were serum-starved in DMEM containing 0.2%
FBS for 48 h and stimulated to enter the cell cycle by DMEM containing 15%
FBS. Cells were harvested at the indicated time points, and luciferase was
detected by the Dual-Luciferase reporter assay system (Promega).
Western Blot Analysis—Cell protein lysates were separated in SDS
polyacrylamide gels and transferred to polyvinylidene fluoride membranes. Blots
were first incubated in blocking buffer (10% skim milk in Tris-buffered saline plus
Tween 20) for 1 h and subsequently incubated overnight in blocking buffer
containing the antibody specific for Myc tag 9E10 (Santa Cruz Biotechnology,
SC-40). The primary antibody was then detected using horseradish-peroxidase
conjugated secondary antibody and ECL reagent as described by the
manufacturer (Amersham Biosciences). Immunofluorescence—MEFs were
33
grown in 35-mm dishes and transfected with His-Myc-tagged E2F8 or control
vector. The cells were fixed with 4% paraformaldehyde and methanol/acetone
(1:1). The Myc antibody 9E10 was the primary antibody used, and rhodamineconjugated anti-mouse IgG (Vector Laboratories) was used as the secondary.
The cells were counterstained with DAPI. The procedure for the staining was
same as described before (de Bruin et al., 2003).
Electromobility Shift Assay for DNA Binding—The probe used for the DNA
binding assay was a fragment of the adenoviral E2 promoter containing two E2F
binding sites. The complimentary strands of the probes were biotinylated on the
5’ end and were annealed together to make a double-stranded probe end labeled
with biotin. The sequence of the wild-type probe was 5’
TCGAGACGTAGTTTTCGCGCTTAAATTTGAGAAAGGGCGCGAAACTAGTC
TTAACTCGA- 3’, and that of the mutated probe was 5’
TCGAGACGTAGTTTTAAGGCTTAAATTTGAGAAAGGGCTTGAAACTAGTCC
TAACTCGA- 3’. The binding reaction was carried out in a 20-μl volume using 40
fmol of biotinylated probe and 8 pmol of non-biotinylated wild-type or mutated
probe as and when required. The binding conditions were the same as described
previously (Logan et al., 2004). The supershift analysis was carried out as
previously described using an antibody specific against Myc (Santa Cruz
Biotechnology, SC-40). Proteins were translated using the TNT Quick Coupled
transcription/translation system (Promega). After carrying out the binding reaction
at 30 °C for 30 min, Ficoll was added to it to a final concentration of 4% and it
34
was separated on a 4% polyacrylamide gel. After running the gel, it was
transferred to Hybond N+ membrane (Amersham Biosciences) and UV-crosslinked. It then was probed using the LightShiftTM chemiluminescent EMSA kit
(Pierce) following the manufacturer’s protocol. The chemiluminescence was
detected on Hyperfilm (Amersham Biosciences).
Site-directed Mutagenesis of the DNA-binding Domains—Site-directed
mutagenesis was carried out using the QuikChange site-directed mutagenesis kit
following the manufacturer’s protocol. We used the following primers for DBD1,
5’-CGGAAGGAGAAGAGCGAATTCTTGCTATGCCACAAA- 3’ and 5’
TTTGTGGCATAGCAAGAATTCGCTCTTCTCCTTCCG- 3’, and the following
primers for DBD2, 5’-CGCAAAGACAAGTCCGAATTCGTGATGAGCCAGAAG3’ and 5’ -CTTCTGGCTCATCACGAATTCGGACTTGTCTTTGCG- 3’.
Structural Analysis—Sequence alignments of the E2F8 DNA-binding
domains by the ClustalW method were used to generate models for E2F8 DNA
binding (Schwede et al., 2003). Modeling requests were submitted to the SWISSMODEL protein modeling server using the previously solved E2F4/DP2 crystal
structure Protein Data Bank file (1CF7) as the template.
Co-immunoprecipitation—Transiently transfected 293 cells were
harvested in cold phosphate-buffered saline, and cell pellets were lysed in 10X
volume of lysis buffer (0.05 M sodium phosphate, pH 7.3, 0.3 M NaCl, 0.1%
Nonidet P-40, 10% glycerol with protease inhibitor mixture). Lysates were
incubated with Protein G Plus/protein A-agarose beads (Calbiochem) at 4 °C for
35
1 h to preclear. The precleared lysates were incubated with appropriate antibody
overnight. Protein G Plus/ protein A-agarose beads were added and incubated
for 1 h at 4 °C. Protein binding to the beads were released and resolved by SDSPAGE followed by immunoblotting. Immunoprecipitation and Immunoblotting
were performed using M2 monoclonal anti-FLAG (Sigma), anti-HA (Roche
Applied Science), anti-Myc 9E10 antibodies.
Retroviral Infection—Full-length cDNAs for His-Myc-tagged E2F8 was
subcloned into the pBABE retroviral vector containing a hygromycin- resistance
gene. High-titer retroviruses were produced by transient transfection of retroviral
constructs into the Phoenix-Eco packaging cell line as described previously (Pear
et al., 1993). MEFs were infected with the retrovirus using standard methods and
were selected in the presence of 200 μg ml-1 hygromycin.
Proliferation Assay—MEFs were plated at 4 X 104 cells/60-mm plate and
grown in DMEM with 15% FBS. Duplicate plates were counted daily using a BD
Biosciences Coulter counter and were replated every 72 h at the same density of
the initial plating.
RESULTS
Identification and Cloning of Mouse E2F8— I performed a homology
search across the sequenced mouse genome (Celera and GenBankTM
databases) using the E2F3 DBD amino acid sequence as the reference. The
36
search retrieved the known E2Fs (E2F1–7) and a potentially novel E2F gene,
which we named E2F8. The in silico predicted E2F8 protein (Celera and
GenBankTM databases) possessed two E2F-like DBDs but shared no homology
to any other known domains conserved across the E2F family members. I also
performed a BLASTN (www.ncbi.nlm.nih.gov/BLAST/) search using the predicted
E2F8 transcript as a query. The search recovered several mouse expressed
sequence tags from a variety of mouse tissues and developmental stages
(UniGene Cluster Mm.240566). Using E2F8-specific primers, I screened the
mouse testis Cdna library (Origene Technologies Inc.) and retrieved the fulllength
mouse E2F8 cDNA clone.
Analysis using the University of California Southern California Genome
Browser BLAT tool (Kent, 2002) revealed that the mouse E2F8 gene is located
on chromosome 7 and contains 13exons separated by 12 introns (Figure 2.1A). I
sequenced a BAC clone containing the E2F8 gene to determine the sequences
across the splice junctions. From 5’ to 3’ end of the gene, the 12 splice sites have
the universal consensus splice junction dinucleotides GT/AG.
The first initiation ATG, as predicted by the presence of a Kozak
consensus sequence and an in-frame termination codon, is in exon 2, and the
stop codon TGA are in exon 13, giving rise to a 2583-bp open reading frame
encoding 860 amino acids (Figure 2.1B). E2F8 mRNA has a 300-bp long 5’-UTR
and a 587-bp long 3’-UTR. There are two consensus polyadenylation signals,
AATAAA and ATTAAA, that are 327 and 238 bp upstream of the poly(A) tail,
37
respectively. The polyadenylation signal that is relevant in this particular clone is
the non-canonical polyadenylation signal TATAAA located 18 bases upstream of
the poly(A) tail (Beaudoing et al., 2000).
E2F8 Protein Characteristics and Homology with the Other E2Fs—The
E2F8 protein consists of 860 amino acids with a predicted molecular mass of ~95
kDa. It has two E2F-like DBDs and three putative nuclear localization signals
(Figure 2.1C and Figure 2.1D). The presence of two E2F-like DBDs is
reminiscent of the mammalian E2F7 and Arabidopsis E2Fd-f. The alignment of
the DBDs of mouse E2F1–8 and Arabidopsis E2Fd using the ClustalW program
shows high homology with notable conservation of the RRXYDI DNA recognition
motif (Figure 2.2A). Despite significant homology in the duplicated DBD, E2F8 is
devoid of any other E2F-like domains including the pocket protein binding,
transactivation, and DP dimerization domains, a characteristic shared by
mammalian E2F7 and Arabidopsis E2Fd-f proteins (Figure 2.2C).
Phylogenetically, the DBDs of Arabidopsis E2Fd-f and mouse E2F7 and
E2F8 cluster together (Figure 2.2B). Significantly, when the full-length mouse
E2F proteins were analyzed for evolutionary relationship on the basis of their
primary structure, a segregation pattern that is reflective of their known functional
characteristics was observed. The acquisition of additional E2Fs may stem from
a developmental requirement for E2F activity in multiple different tissues as
organisms evolve to be structurally and functionally more complex down the path
of evolution (Figure 2.2D).
38
E2F8 Expression Pattern in Tissues and over the Cell Cycle— As a first
step toward the understanding of E2F8 function, I investigated its tissue and cell
cycle expression patterns. A 3’~2500-bp fragment of E2F8 cDNA, which lacked
any significant sequence overlap with any of the other known E2Fs, was used to
probe tissue-specific Northern blots. E2F8 was highly expressed in the liver, skin,
thymus, and testis but not in the brain, muscle, and stomach (Figure 2.3A).
Interestingly, this pattern of expression is almost identical to that previously found
for E2F7, giving rise to the possibility that E2F7 and E2F8 may have overlapping
or complementary functions in these organs.
As discussed above, the known E2F family members fall into two distinct
categories with regard to their patterns of expression during the cell cycle.
Consistent with their function, the expression of the activator subclass (E2F1,
E2F2, and E2F3a) is maximal as cells enter the cell cycle, whereas the
expression of the repressor subclass (E2F3b, E2F4, and E2F5) remains
unchanged throughout all of the cell cycle. To determine whether the expression
of E2F8 is cell cycle-dependent, E2F8 mRNA levels were measured by Northern
blot and real-time RT-PCR analysis in synchronized and cycling populations of
primary MEFs. MEFs were synchronized in quiescence (G0) by starvation in
medium containing 0.2% FBS for 48 h and then stimulated by the addition of
medium containing 15% FBS. RNA was isolated from cells harvested at different
time points following serum stimulation. Poly(A) mRNA was purified and
subjected to denaturing electrophoresis as described under “Materials and
39
Methods,” transferred to membrane, and hybridized to an E2F8-specific
radioactive-labeled probe (Figure 2.3B). Both the Northern blot and the real-time
RT-PCR analyses suggest that E2F8 expression is cell growth-dependent with
maximal expression levels found during S phase (Figure 2.3C). The seruminduced activation of E2F8 expression was mild when compared with the
activation of Cdc6 expression, which is a well established cell cycle-regulated
E2F-target gene. This mild induction in E2F8 expression was not simply a
consequence of an inability of primary MEFs to respond to serum, because E2F8
expression was also poorly induced in p53 mutant MEFs, which can be efficiently
induced to enter the cell cycle in response to serum addition.
Regulation of the E2F8 Promoter—To understand the basis of E2F8
tissue-specific and cell growth-dependent expression patterns and to confirm its
transcriptional start site, the genuine 5’ end of the E2F8 geneI used 5’-RACE
PCR with mRNA isolated from the adult mouse thymus, testis, and primary
MEFs. Approximately 20 clones were analyzed from each tissue type, revealing
the presence of three alternative first exons (Figure 2.4A, exons 1a, 1b, and 1c).
As a reference, the first nucleotide of exon 1b was labeled as +1. All of the three
alternative first exons spliced precisely to the common exon 2, which contains
the ATG of the full-length E2F8 open reading frame. The universal splice junction
dinucleotide GT/AG was present in each of the alternative first exons. The usage
of either exon 1a or 1b preserved the full-length open reading frame of E2F8.
The usage of exon 1c introduced two short ORFs preceding the ATG of the full-
40
length E2F8 ORF, which could potentially produce proteins of 98 and 30 amino
acids long. However, none of these ATGs fulfilled the Kozak criteria for efficient
initiation of translational and is most likely not used as initiating codons. Hence,
the ATG located toward the 5’ end of exon 2, which contains a perfect Kozak
sequence, is predicted to represent the translational start site for the full-length
E2F8 ORF. The usage of the three alternative exons, 1a, 1b, or 1c, gives rise to
the same full-length ORF.
To investigate the transcriptional regulation of E2F8 gene expression, we
analyzed the putative E2F8 promoter region for the presence of consensusbinding sites of different transacting factors. Because E2Fs are known to be
auto-regulated, it was not surprising to find two canonical E2F-binding elements
(a and b) at positions +257 and +385. I cloned a ~2.9-kb genomic fragment
containing the -1993 to +855 region of the E2F8 gene into a firefly-luciferase
transcriptional reporter plasmid and called this construct LP. A second E2F8
genomic fragment extending from +166 to +855 bp, which represents the most
proximal sequence near exon 1c (Figure 2.4A), was also cloned into the fireflyluciferase reporter plasmid, which we called the SP. I tested the promoter activity
of these constructs in REF52 cells using the TK promoter-driven Renilla
luciferase plasmid as an internal control for Transfection efficiencies.
Since both the reporter constructs contained the E2F DNA binding
elements a and b, I determined whether these constructs were responsive to the
overexpression of E2F1, an activator of E2F. As expected, E2F1 overexpression
41
led to a dose-dependent activation of luciferase activity from either the LP or SP
reporter construct (Figure 2.4B). Thus, I hypothesized that the observed growthdependent expression of E2F8 could be the result of an E2F autoregulatory loop
mediated via these putative E2F-binding elements. To test this hypothesis, I
analyzed the transcriptional activity of these reporter constructs in synchronized
REF52 cell populations. The reporter constructs were transfected in REF52 cells
and serum-starved them in medium containing 0.2% FBS for 60 h in order to
synchronize them in G0 and then re-stimulated the cells with medium containing
15% FBS. As expected, the expression from both the SP and LP constructs was
significantly increased in the18-h re-stimulated samples, indicating that the cell
growth regulation of E2F8 is, at least in part, transcriptional in nature (Figure
2.4C). To assess the role of the two consensus E2F-binding elements in
regulating the expression of E2F8, I mutated both the E2F-binding elements, a
and b, in the SP reporter construct (SP*ab) and re-assessed their promoter
activity in E2F1-overexpressing cells. As shown in Figure 2.4E, the mutation of
the two E2F DNA recognition sites reduced but did not completely eliminate the
E2F responsiveness of these reporters, indicating that additional non-consensus
E2Ftarget sites must also mediate their E2F responsiveness. To determine
whether these consensus E2F sites played a role in the growth regulation of
E2F8, I analyzed the activity of both the wild-type (SP) and mutant SP reporter
constructs (SP*b) in quiescent REF52 cells. Mutation of site b (SP*b) led to a
small but reproducible 2-fold increase in reporter expression, indicating that E2F8
42
expression in quiescent cells is likely repressed through an E2F-dependent
mechanism (Figure 2.4D). Although the expression of E2F8 is likely to be
complex, the above data suggest that the E2F-binding sites contribute to the
positive and negative regulation of its expression during the cell cycle.
Subcellular Localization and DNA Binding Activity of E2F8—To gain
insight into the possible function of the E2F8 protein product, I overexpressed a
Myc-tagged version of the murine E2F8 protein (Myc-E2F8) in MEFs and
assessed its effect on cellular proliferation. Western blot analysis of Myc- E2F8transfected cell lysates using Myc epitope-specific antibodies (9E10) revealed a
protein product that migrated in SDSPAGE with a mobility of ~115 kDa. Two
additional Myc-E2F8- specific products of ~110 and 82 kDa were also evident in
these lysates and probably represent degradation cleavage productsof the fulllength protein (Figure 2.5A). Consistent with the presence of three potential
nuclear localization signals, Immunofluorescence microscopy using anti-Myc
epitope antibodies revealed Myc-E2F8 to be completely localized to the nucleus
(Figure 2.5B).
All of the known E2F family members possess a highly conserved E2F
DNA-binding domain that can mediate specific binding to consensus E2F DNAbinding elements. The ability of in vitro translated Myc-E2F8 to bind the
adenoviral E2 promoter fragment containing two intact E2F-binding sites was
tested by EMSA. As shown in Figure 2.5C, in vitro translated Myc-E2F8 bound
specifically to the E2 probe as indicated by the appearance of two distinct bands
43
that were both supershifted with anti-Myc antibodies. In vitro translated MycE2F3/DP1 served as a positive control, and in vitro translated luciferase protein
served as a negative control for DNA binding activity. The binding of E2F8 to the
E2 probe was demonstrated to be specific, because the binding could be
efficiently competed with an excess unlabeled E2 probe but not with an excess of
a mutant E2 probe containing a 2-bp substitution within the E2F-binding
consensus site.
Similar to Arabidopsis E2Fd-f and mammalian E2F7, E2F8 contains two
DBDs (referred to as DBD1 and DBD2 from N to the C terminus). This domain
arrangement is identical to the recently described E2F7 protein, which binds DNA
independently of DP (Logan et al., 2004). Given this similarity and the high
degree of homology between E2F and DP DNA-binding domains, we tested
whether modeling of E2F8 would allow the DNA-binding domains to adopt a
structure homologous to E2F/DP binding as determined from the crystal structure
of the E2F4/DP2 heterodimer (Zheng et al, 1999). Sequence alignments by the
ClustalW method and through the SWISS-MODEL server showed that DBD2 has
a higher sequence homology to the DP DNA-binding domain than DBD1. Based
upon conservation of the residues involved in heterodimerization and the binding
of the E2F DNA consensus sequence, we were able to model E2F8 binding to
DNA with DBD2 adopting the structure of the DP binding partner. Our in silico
analysis and model for DNA binding suggest that E2F8 binds DNA with DBD1 in
44
the position of the E2F binding partner and DBD2 in the position of the DP
binding partner (Figure 2.5E).
To determine whether each domain directly contributes to E2F8 DNA
binding activity, point mutations were introduced in DBD1, DBD2, or both DBD1
and DBD2 (DBD1–2) that are predicted to disrupt DNA binding activity. The
conserved leucine 118 and glycine 119 in DBD1 and leucine 266 and arginine
267 in DBD2 were replaced with glutamate and phenylalanine, respectively
(Figure 2.5E). These leucines contribute to the dimerization interface of the DNAbinding domains and are conserved across all of the E2F family members as
shown in Figure 2.2A. In E2F1, the corresponding conserved leucine at position
132 is thought to make important heterodimerization contacts with DP and its
mutation abrogates DNA binding activity (Zheng et al., 1999; Johnson et al.,
1993). Disruption of these two amino acids in DBD1 or DBD2 of E2F8 completely
abrogated its DNA binding capacity, indicating that the integrity of both of the
DBDs of E2F8 is important for its DNA binding function (Figure 2.5D). These
observations are consistent with our modeling of E2F8, which indicates that DNA
binding is dependent upon dimerization interactions at both interfaces of DBD1
and DBD2 (Figure 2.5E) (Logan et al., 2004; Kosugi et al., 2002).
E2F8 Forms Homodimers—Previous work from LaThangue’s group
demonstrated that the related E2F7 family member can form homodimers (Logan
et al., 2004). To test the possibility that E2F8 could also oligomerize, FLAGtagged E2F8 and HA-tagged E2F8 were exogenously expressed in the 293 cells
45
and their ability to interact with each other by immunoprecipitation and
immunoblotting using anti-FLAG or anti-HA antibodies. To rule out any
nonspecific antibody interactions, singly transfected cells were also
immunoprecipitated with either FLAG-E2F8 or HA-E2F8 as controls (Figure
2.6A). In this analysis, HA-tagged E2F8 could be detected in the anti-FLAG
immunoprecipitates derived from the doubly transfected cells but not from the
singly transfected HA-tagged E2F8 samples. Likewise, FLAG-tagged E2F8 could
be detected in the HA immunoprecipitates from doubly but not singly transfected
samples. These data indicate that E2F8 can indeed form oligomers.
These findings raise the possibility that the inability of the E2F8 DBD
mutants to bind to E2F consensus sites could be due to the disruption of E2F8
oligomerization. Co-immunoprecipitation assays demonstrated that the single
(DBD1 or DBD2) or double (DBD1–2) DBD mutants of E2F8 still retained the
capacity to oligomerize (Figure 2.6B and data not shown). These results
demonstrate that oligomerization of E2F8 is independent of its DNA binding
activity.
E2F8 Overexpression Blocks Cellular Proliferation—E2Fs are thought to
be critical players in orchestrating the control of cellular proliferation. To
determine the potential role of E2F8 in the control of cellular proliferation, E2F8
was overexpressed in MEFs and proliferation was monitored over a period of 7
days. To this end, primary MEFs were infected with retroviruses expressing MycE2F8 and transduced cells were selected for 48 h in hygromycin. Cells were then
46
plated in medium containing 15% FBS, and viable cells were harvested and
counted every 24 h over a period of 7 days. Relative to control-treated cells,
MEFs over-expressing Myc-E2F8 proliferated considerably slower (Figure 2.7A).
Consistent with the observed growth retardation, the expression of E2F-target
genes in synchronized populations of Myc-E2F8-overexpressing cells was
significantly reduced (Figure 2.7B). Whether the inhibition of E2F-target gene
expression is a direct or indirect effect of E2F8 over-expression remains to be
determined.
DISCUSSION
The E2F family members play important roles in cellular proliferation,
apoptosis, and differentiation in both Rb-dependent and independent manners.
E2F activities have been described in the vast majority of eukaryotes studied,
ranging from plants to mammals, with the exception of yeast. Mammals have
seven distinct E2F genes with some family members encoding multiple related
isoforms through differential promoter usage or alternative splicing. The
identification of yet another mammalian E2F family member, E2F8, provides
further complexity to the E2F family of transcription factors. Interestingly, E2F8
has the distinctive feature of possessing two tandem DBDs. However, other than
within these E2F-like DBDs, there is little amino acid sequence conservation
between E2F8 and the other E2Fs.
47
E2F8 is an E2F member based on the presence of conserved DNAbinding domains and its ability to bind to E2F consensus sites found in many
E2F-regulated promoters. Structure modeling predicts that the duplicated DBDs
of E2F8 can interact with each other to form a functional DNA binding unit, thus
alleviating the requirement to interact with DP. Interestingly, this modeling
predicts that the key conserved leucine residues, which are important for the
interaction between the DBDs of the E2F and DP heterodimers, are also
important for the intramolecular interactions between DBD1 and DBD2. This
prediction is supported by our data demonstrating that introduction of point
mutations at this conserved leucine residue in either of the E2F8 DBDs
abrogates DNA binding activity. Co-immunoprecipitation assays demonstrate that
E2F8, like E2F7, can form homodimers, suggesting that these two unique E2Fs
could potentially form contacts with multiple consensus E2F sites at once.
E2F7 and E2F8 share a number of characteristics that could reflect their
unique function. Each is expressed in a cell growth-dependent manner with peak
levels found during S phase and is expressed in the same adult tissues of mice.
Both have the ability to homodimerize and to repress E2F-dependent gene
expression. Importantly, their overexpression can lead to a pronounced decreas
in the proliferative capacity of cells. These observations have led to the
hypothesis that these two E2F family members may have overlapping and/or
synergistic functions in the control of cellular proliferation. Although the
identification of E2F8 adds further complexity to the E2F family of transcription
48
factors, our findings begin to place E2F members into distinct subclasses that
have general structural and functional themes that might be used to differentially
regulate cellular proliferation.
Acknowledgment—We thank Charles Bell for advice in modeling E2F8
structure.
49
Figure 2.1
Structure of mouse E2F8 gene, mRNA, and protein.
A, schematic representation of the E2F8 genomic locus on the
Mus musculus chromosome 7. The 13 exons are represented
by boxes, and the intronic regions are represented by lines. The
lengths of the introns and exons are indicated. B, structure of
the E2F8 mRNA. The ORF extends from 301 to 2883 base
pairs. The 5'- and 3'-untranslated regions are shaded light gray.
The polyadenylation signals are represented by arrows in the 3'UTR. C, schematic diagram of full-length E2F8 protein. The
nuclear localization signals (NLS) and the DBDs as predicted by
in silico analysis are indicated, and the amino acid positions are
indicated within parentheses. D, amino acid sequence of fulllength E2F8 protein. From the N terminus to the C terminus, the
DBDs are named DBD1 and DBD2 and are indicated by
boldface letters in the primary structure of full-length E2F8
protein.
50
Figure 2.1
Structure of mouse E2F8 gene, mRNA, and protein.
51
Figure 2.2
Comparative analysis of E2F8 and other known E2F and DP
family members.
A, sequence alignment of the DBDs of mouse E2F1–8
(represented as mE2F1–8), mouse DP1–2 (represented as mDP1–
2), and A. thaliana E2Fd (AtE2Fd) using the ClustalW program.
The conserved RRXYD motif that makes DNA base contacts is
indicated with a line. The amino acids Leu-Gly in mE2F8 DBD1 and
Leu-Arg in mE2F8 DBD2 are marked with asterisks, indicating that
these amino acids when mutated to Glu and Phe abolished DNA
binding of E2F8 as shown later in Fig. 5d. Conserved amino acids
are shaded yellow. B, phylogenetic relationship of the DBD amino
acid sequences among E2F8, the E2F/DP family in mouse, and
E2Fd-f of A. thaliana (phenogram). The length of each horizontal
line represents the evolutionary distance between branching points,
whereas the units at the bottom of the tree indicate the number of
substitution events. The dotted line on the phenogram indicates a
negative branch length. C, schematic representation of the domain
structure of full-length E2F1–8 proteins. The domains indicated are
N-terminaldomain (NTD), DBD, leucine zipper (LZ), marked
(Marked), Rb binding (Rb bind), and transactivation domains. D,
phylogenetic relationship of the full-length amino acid sequences
between murine E2F1 and E2F8 (phenogram). The length of each
horizontal line represents the evolutionary distance between
branching points, whereas the units at the bottom of the tree
indicate the number of substitution events. The dotted line on the
phenogram indicates a negative branch length.
52
Figure 2.2
Comparative analysis of E2F8 and other known E2F and
DP family members.
53
Figure 2.3
Tissue-specific and cell cycle-dependent expression of
E2F8.
A, mouse tissue blot from Origene was either hybridized with an
E2F8-specific probe or a -actin probe as a loading control. B,
primary MEFs were brought to quiescence by serum starvation
and stimulated to proliferate by the addition of fresh medium with
15% serum. Cells were harvested for RNA at 0, 12, 18, and 24 h
after serum stimulation as indicated, and Northern blot analysis
was performed as described under "Materials and Methods." The
membrane was probed with E2F8-specific probe or with GAPDH
probe as a loading control. C, primary MEFs or p53-/- MEFs were
brought to quiescence by serum starvation and stimulated to
proliferate by the addition of 15% FBS containing medium. Cells
were harvested for RNA at 0, 9, 12, 15, 18, 21, and 24 h after
serum stimulation, and real-time RT-PCR was performed with
primers for E2F8 and Cdc6 as described under "Materials and
Methods." Real-time RT-PCR for GAPDH was done to
standardize the amount of cDNA in each sample. Cells treated
identically were harvested for BrdUrd incorporation. BrdUrd was
added to the medium 3 h before harvesting. Cells were stained
with anti-BrdUrd antibody with DAPI counterstaining, and
BrdUrd-positive cells were counted as described under
"Materials and Methods."
54
Figure 2.3
Tissue-specific and cell cycle-dependent expression of E2F8.
55
Figure 2.4
Regulation of E2F8 promoter.
A, schematic representation of the E2F8 promoter region and
that of the luciferase reporter constructs, SP and LP.
Alternative first exons, 1a, 1b, and 1c, are shown, and their
positions relative to each other are indicated. The two E2Fbinding DNA elements (a and b) are shown between the
exons 1b and 1c and the mutations introduced therein are
indicated. B, induction of luciferase activity of SP and LP
constructs by transient transfection of 50 and 150 ng of E2F1
or the control vector in REF52 cells. Proliferating cells were
transfected and brought to quiescence by serum starvation
before harvesting for measuring luciferase activity as
described under "Materials and Methods." TK promoter
driven Renilla luciferase reporter construct was used as a
control for transfection efficiencies. Each experiment was
done in triplicates, and the results were reproduced at least
three times. C, REF52 cells transiently transfected with SP or
LP constructs were brought to quiescence and stimulated to
grow by adding 15% FBS-containing medium. Cells were
harvested 0 and 18 h after serum stimulation, and luciferase
activity was measured. D, REF52 cells transiently transfected
with SP or SP*b (SP with b elements mutated) construct were
brought to quiescence, and luciferase activity measured. E,
fold induction of luciferase activity by 150 or 1500 ng of E2F1
or the control vector was measured in lysates from REF52
cell transfected with SP or SP*ab (SP construct mutated for
both the E2F-binding elements, a and b).
56
Figure 2.4
Regulation of E2F8 promoter.
57
Figure 2.5
Subcellular localization and DNA binding activity of E2F8.
A, Western blot analysis of lysates from MEFs transiently
transfected with Myc-tagged E2F8 or the control vector. Myc-8specific products are indicated by arrows. B, MEFs transiently
transfected with Myc-tagged E2F8 or the control vector were
stained with anti-Myc antibody and counterstained with DAPI,
showing that Myc-tagged E2F8 is nuclearly localized. C, EMSA
was performed with biotin-labeled adenoviral E2 promoter
fragment and in vitro translated Myc-tagged E2F8 protein (Myc8, indicated by black arrows). In vitro translated luciferase was
used as a negative control, and Myc-tagged E2F3a (Myc-3a)
and DP1 (indicated by white arrows) were used as a positive
control. The binding resulted in two new bands (indicated by a
black or white arrow), which were supershifted using the antiMyc antibody or competed out using non-biotinylated "cold"
competitor (cc). The cold competitor (cc*) point-mutated at the
E2F-binding elements was unable to compete out the binding
indicating sequence-specific interaction. D, in vitro translated
E2F8 point-mutated at DBD1 (Myc-8-DBD1) or DBD2 (Myc-8DBD2) or DBD1 and DBD2 (Myc-8-DBD1–2) was unable to
bind the biotin end-labeled E2 promoter fragment. In vitro
translated Myc-tagged wild-type E2F8 (Myc-8) was used as a
positive control, and luciferase was used as a negative control.
E, structural model for DNA binding by E2F8. Left, the crystal
structure of the E2F4/DP2 heterodimer with E2F4 (pink) and
DP2 (teal) bound to an E2F DNA consensus sequence is
shown (Protein Data Bank code 1CF7) (31). Right, our
structural model based on homology modeling with the solved
structure for the E2F4/DP2 heterodimer illustrating interactions
between DBD1 (gold) and DBD2 (blue) during DNA binding by
E2F8. Below, the boxed dimerization interfaces between DBDs
1 and 2 are enlarged to illustrate the protein regions that make
conserved dimerization contacts. The amino acid side chains
that were mutated for EMSA and co-immunoprecipitation
experiments are highlighted in red and blue for DBD1 and
DBD2, respectively.
58
Figure 2.5
Subcellular localization and DNA binding activity of
E2F8.
59
Figure 2.6
E2F8 can homodimerize.
A, lysates from 293 cells transfected with both HA-tagged
E2F8 (HA-8) and FLAG-tagged E2F8 (Flag-8) were coimmunoprecipitated (IP) using anti-FLAG antibody and
immunoblotted (IB) with anti-HA antibody or vice versa.
Singly transfected 293 cell lysates with either FLAG-E2F8 or
HA-E2F8 were used as negative control to rule out any
nonspecific antibody interactions. B, Myc-8 and Flag-8 were
co-transfected in 293 cells. Lysates were immunoprecipitated
with anti-FLAG antibody and immunoblotted with anti-Myc
antibody. This was repeated with lysates from Myc-E2F8 and
FLAG-E2F8 with DNA-binding domains 1 and 2 mutated
(Myc-8-DBD1–2 and Flag-8-DBD-1–2, respectively).
Immunoprecipitation with normal mouse IgG (Calbiochem)
was used as a negative control to rule out nonspecific
interactions.
60
Figure 2.7
E2F8 overexpression inhibits cellular proliferation.
A, primary MEFs infected with E2F8 overexpressing or control
retrovirus were plated at a density of 4 x 104 cells/60-mm plate.
Cells were grown in 15% FBS containing medium and were
harvested every 24 h for 7 days and counted. B, real-time RTPCR for the E2F-target genes, cyclin A2 (cyc A2), polymerase
(pol ), cdc6, cyclin E (cyc E1), dhfr, and E2F2 was performed on
the cells infected with E2F8-overexpressing retrovirus or the
control virus. Real-time RT-PCR for GAPDH was done as a
control for the amount of cDNA in each sample. The values on
the y axis represent fold induction.
61
Figure 2.7
E2F8 overexpression inhibits cellular
proliferation.
62
CHAPTER 3
IDENTIFICATION AND CHARACTERIZATION OF E2F7, A NOVEL
MAMMALIAN E2F FAMILY MEMBER CAPABLE OF BLOCKING CELLULAR
PROLIFERATION 2
ABSTRACT
The mammalian E2F family of transcription factors plays a crucial role in
the regulation of cellular proliferation, apoptosis, and differentiation. Consistent
with its biological role in a number of important cellular processes, E2F regulates
the expression of genes involved in cell cycle, DNA replication, DNA repair, and
mitosis. It has proven difficult, however, to determine the specific roles played by
the various known family members in these cellular processes. The work
presented here now extends the complexity of this family even further by the
identification of a novel E2F family member, which we now term E2F7. Like the
expression of the known E2F activators, E2F1, E2F2, and E2F3, the expression
2
Reproduced with permission from J Biol Chem. 2003 Oct 24;278(43):42041-9. Epub 2003 Jul
31, de Bruin A, Maiti B, Jakoi L, Timmers C, Buerki R, Leone G. © 2005 by The American Society
for Biochemistry and Molecular Biology, Inc. All data pertaining to the Northern Blot assays
showing E2F7 expression pattern, and in part the data from Luciferase reporter assays showing
E2F7 promoter regulation and E2F7 mediated repression were generated by Maiti B, under the
supervision of Leone G.
63
of E2F7 is growth-regulated, at least in part, through E2F binding elements on its
promoter, and its protein product is localized to the nucleus and associates with
DNA E2F recognition sites with high affinity. A number of salient features,
however, make this member unique among the E2F family. First, the E2F7 gene
encodes a protein that possesses two distinct DNA-binding domains and that
lacks a dimerization domain as well as a transcriptional activation and a
retinoblastoma-binding domain. In contrast to the E2F activators, E2F7 can block
the E2F-dependent activation of a subset of E2F target genes as well as mitigate
cellular proliferation of mouse embryo fibroblasts. These findings identify E2F7
as a novel member of the mammalian E2F transcription factor family that has
properties of a transcriptional repressor capable of negatively influencing cellular
proliferation.
INTRODUCTION
The retinoblastoma (Rb) gene was the first tumor suppressor identified in
humans some 20 years ago. Studies using tissue culture and in vivo mouse
models have led to the identification of the E2F transcription factor family as an
important effector of Rb function impacting cell proliferation, apoptosis, and
cellular differentiation. From these studies, a paradigm for Rb action in the
control of cellular proliferation has emerged. In this view, cyclin-dependent kinase
activation results in the phosphorylation of Rb and the release of E2F family
64
members from Rb-containing complexes, leading to E2F target activation and
cell cycle progression (Dyson, 1998; Nevins, 1998; Trimarchi and Lees, 2002).
The regulation of E2F activities is not solely due to the action of Rb but is also
subject to control at multiple tiers involving new synthesis, localization,
phosphorylation, acetylation, and degradation (Nevins, 1998; Trimarchi and
Lees, 2002).
The E2F proteins and their heterodimeric DP partners, encoded by eight
distinct genes, contain an evolutionary conserved DNA-binding and dimerization
domain and, at least for E2F1–5, a conserved transactivation and pocket proteinbinding domain. Although heterodimerization with DP proteins is required for E2F
DNA binding activity, the specificity of the heterodimer complex is mediated by
the E2F subunit (Dyson, 1998). Both structural and functional properties have
separated the E2F family members into three subclasses. The expression of
E2F1, E2F2, and E2F3a, which represent the first subclass, oscillates during the
cell cycle, peaks late in G1, and coincides with the activation of G1/S-specific
genes (Leone et al., 1998). During this period, E2F1–3a can be found transiently
bound to many E2F target promoters (Cam et al., 2003). Consistent with an
important role for these proteins in cellular proliferation, the combined disruption
of E2F1, E2F2, and E2F3 in mouse embryonic fibroblasts (MEF) impedes
E2Ftarget expression and cellular proliferation, indicating that these E2Fs may
function as transcriptional activators (Wu et al., 2001).
65
In contrast to the E2F activators, the expression of the second E2F
subclass composed of E2F3b, E2F4, and E2F5 appears relatively constant in
relation to cell growth (Leone et al., 1998). Several studies have implicated the
second E2F subclass in transcriptional repression by recruiting the pocket
proteins Rb, p107, and p130 and their associated histone-modifying enzymes to
E2F target gene promoters (Trimarchi and Lees, 2002). E2F6, the only member
of the third subclass, contains domains for DNA binding and dimerization but
lacks residues for pocket protein binding or transactivation. Instead, E2F6 is
thought to mediate repression either through its direct binding to polycomb group
proteins or through the formation of a large multimeric complex containing Mga
and Max proteins (Ogawa et al., 2002; Trimarchi et al., 2001).
The function of E2Fs in regulating cellular proliferation and differentiation
has been also investigated in non-mammalian species. Of these, Drosophila
melanogaster is the most extensively studied organism, containing at least two
E2F genes (dE2F1–2) and one DP gene (dDP). In many ways, dE2F1 appears to
be a homolog of the mammalian E2F activator subclass, whereas dE2F2
resembles members of the mammalian repressor subclass (Stevaux et al.,
2002). E2F related activities have also been described in Caenorhabditis elegans
and Xenopus laevis but have not been found in yeast (Ceol et al., 2001; Suzuki
et al., 2000).
It is noteworthy that the Arabidopsis thaliana genome contains six E2F
genes (AtE2Fa–e) and two DP genes (AtDPa and AtDPb). Similar to their
66
mammalian E2F1–3 counterparts, AtE2Fa–c proteins can transactivate E2F
targets and are equipped with conserved residues for DNA binding, dimerization,
transactivation, and pocket protein binding (Mariconti et al., 2002; Kosugi et al.,
2002). On the other hand, AtE2Fd–f diverge from the mammalian E2F3b-6
repressors, because they possess two DNA-binding domains (DBD) and lack any
of the other conserved regions necessary for E2F-dependent repression
(Mariconti et al., 2002; Kosugi et al., 2002).
Comparative analysis of the protein sequences between the human,
mouse, fly, nematode, and A. thaliana genomes revealed that the DBD of the
E2F proteins is highly conserved. We therefore used the mouse E2F3 DNAbinding amino acid sequence as bait to identify additional putative E2F family
members within the recently completed mouse and human genome. We
identified a new putative mammalian E2F, which we now designate as E2F7. The
salient feature of this member is that it contains two tandem DBDs that are highly
related to the DBDs of all other E2Fs and thus has a similar structural
organization as AtE2Fd–f from plants. In contrast to most known mammalian
E2Fs, this novel member lacks the residues necessary for dimerization,
transactivation, and pocket protein binding. We show that this protein can bind to
E2F DNAbinding consensus sites, can act as a transcriptional repressor, and can
inhibit cellular proliferation.
67
MATERIALS AND METHODS
Serum Starvation and Serum Stimulation—Subconfluent MEFs were
synchronized by incubation in Dulbecco’s modified Eagle’s medium with 0.2%
FBS for 48 h and then stimulated to proliferate by the addition of Dulbecco’s
modified Eagle’s medium supplemented with 12.5% FBS. The cells were
collected at different time points after serum stimulation and were processed for
BrdU incorporation assays, flow cytometry, and RNA isolation as previously
described (Leone et al., 1998). For the BrdU incorporation assays, we counted at
least twice 300 4,6-diamidino-2- phenylindole (DAPI) counter-stained nuclei for
each time point.
Northern Blot Analysis—Total RNA for Northern blot analysis was isolated
using TRIzol (Invitrogen), and mRNA was subsequently purified using PolyATract
mRNA isolation system as described by the manufacturer (Promega). Purified
mRNA was separated on a 1% agarose gel containing 6% formaldehyde and
transferred onto a Gene Screen membrane (PerkinElmer Life Sciences). The
cDNA probe corresponding to the first 1200 bp of the E2F7 open reading frame
was labeled using Prime-It RmT (Stratagene) with 50 μCi of [α-32P]dCTP. The
cell cycle blot as well as a commercially purchased mouse tissue Northern blot
(Origene) were hybridized overnight under high stringency conditions (5XSSPE,
50% formamide, 5X Denhardt’s solution, 1% SDS at 42 °C) and washed several
times (0.2XSSC, 0.2% SDS at 65 °C) before autoradiography.
68
Reporter Assays—MEFs were grown in triplicate and transfected with the
firefly luciferase expression vectors, together with either thymidine kinase (TK)
Renilla luciferase or cytomegalovirus (CMV)- β- galactosidase as internal
controls, as described previously (Sears et al., 1997). After transfection, the cells
were brought to quiescence by serum starvation and then stimulated to grow by
the addition of fresh medium with serum. The cells were harvested at various
time points, and luciferase was detected by a dual luciferase reporter assay
system (Promega). β -Galactosidase assays were performed as described
previously (Sears et al., 1997).
RESULTS
Tissue-specific and Cell Cycle-dependent Expression of E2F7—To
determine the patterns of E2F7 expression in adult mice, mRNA levels were
measured by Northern blot analysis using an E2F7-specific probe containing the
N-terminal 1200 bp of coding sequence. Two specific sized transcripts hybridized
to this probe; however, at higher stringencies the slower 5.5-kb migrating
transcript was the predominant species that could be detected (data not shown).
As shown in Figure 3.1A, the highest levels of E2F7 expression were observed in
the skin and thymus, with little or no expression in the brain, muscle, and
stomach.
69
Previous studies have detailed the distinct pattern of expression of the
various E2F family genes following mitogenic stimulation of cells. To determine
whether E2F7 expression was responsive to growth stimuli, E2F7 expression
was measured by semi-quantitative reverse transcriptase-PCR and Northern blot
analysis in synchronized cell populations. To this end, mouse embryonic
fibroblasts were starved in 0.2% FBS for 48 h and subsequently stimulated with
12.5% FBS, and the cells were harvested at different time points for RNA
isolation. Cells similarly treated in parallel cultures were incubated with BrdU,
fixed at the various time points post-stimulation, and processed for indirect
immunofluorescence. This analysis indicated that serum stimulation of quiescent
cells results in an increase of E2F7 expression similar to that of E2F1, E2F2, and
E2F3 (18), with levels peaking during the S phase, suggesting that the regulation
of E2F7 expression is cell growth-dependent (Figure 3.1B).
Analysis of Promoter Sequence Controlling the Expression of the E2F7
Locus—The cell growth-dependent accumulation of E2F7 mRNA suggests that it
may be transcriptionally regulated in response to growth stimuli, albeit that other
transcriptional- independent mechanisms are also possible. Examination of
sequences upstream of the predicted ATG start codon revealed the presence of
a number of potential binding sites for a variety of transcription factors (Figure
3.2A). Notably, the putative E2F7 promoter contained three potential E2F-binding
elements near the 5’ end of the predicted first exon, with Sp1 recognition sites
very close to each of the E2F binding elements.
70
To explore the role of transcriptional regulation in the control of E2F7
expression, we isolated and cloned a 2-kb genomic DNA fragment containing the
5’ sequence flanking the first exon into a luciferase reporter construct (pr-E2F7).
To first determine whether the growth-regulated accumulation of E2F7 mRNA
could be accounted for a transcriptional mechanism, the activity of the prE2F7
reporter construct was measured in synchronized MEF populations. To this end,
MEFs were transfected with the prE2F7 reporter construct along with a TK-driven
Renilla luciferase plasmid as an internal control; the cells were then growtharrested by serum deprivation and subsequently stimulated to reenter the cell
cycle by the addition of 12.5% serum. The cells were then harvested at various
times after serum addition, and cell extracts were assayed for firefly and Renilla
luciferase activity. As shown in Figure 3.2B, E2F7 promoter activity was low in
quiescent cells and in early G1 cells but increased 8-fold by 24 h poststimulation, corresponding with the serum-dependent induction of E2F7 mRNA
observed previously (Figure 3.1B).
To investigate the role of the putative E2F recognition sites in regulating
the growth-dependent activation of E2F7, the three E2F binding elements on its
promoter were eliminated in combination (Figure 3.2A). MEFs were transfected
with either the wild-type or each of the mutated E2F7 promoter constructs, as
well as with a TK-Renilla luciferase plasmid as an internal control. Transfected
cells were growth-arrested in low serum medium and then stimulated to reenter
the cell cycle by the addition of 12.5% serum. The promoter with the three E2F
71
sites mutated exhibited a 5-fold increase in activity in quiescent cells (Figure 3.2B
and Figure 3.2C), suggesting that these E2F-binding elements play a negative
regulatory role on the regulation of its expression. In addition, mutation of the
E2F-binding sites also resulted in higher E2F7 promoter activity when cells were
restimulated with serum, as early as 6 h post-stimulation, indicating that E2F7
expression can also be regulated positively through E2F-binding siteindependent transcriptional mechanisms. The fact that wild-type and mutant
E2F7 promoter constructs could be activated equally well by the overexpression
of E2F1 (Figure 3.2D and Figure 3.2E), suggests that E2F activators can
regulate the E2F7 promoter independently of these three E2F-binding elements.
Together, these data suggest that E2F7 expression is subject to E2F-mediated
negative and positive regulation by different mechanisms.
E2F7 has Transcriptional Repression Function—We next examined the
possible role of E2F7 in the transcriptional control of E2F-responsive genes. To
this end, MEFs were transfected with luciferase reporter constructs carrying a
variety of E2F-responsive promoters, including TK, E2F2, E2F3a, polymerase α,
and Rb along with increasing amounts of a Myc-E2F7 expressing vector.
Standardization with β-galactosidase activity as an internal control revealed that
E2F7 expression in cycling population of cells led to a consistent decrease in
some of the E2F target genes, with little or no effect in others (Figure 3.3A). The
inability to activate transcription is consistent with the fact that E2F7 does not
contain a transactivation domain.
72
The inhibition of E2F target genes was accentuated when synchronized
population of cells was assayed instead. The prE2F2 reporter gene is activated
in an E2F-dependent manner during the G1/S transition in cells induced to enter
the cell cycle. To determine whether E2F7 can inhibit this G1/S-specific
induction, MEFs were transfected with the Myc-E2F7 expression vector and an
E2F-reporter construct (prE2F2) as before, but where subsequently arrested in
G0 by serum deprivation, and then induced to enter the cell cycle by the addition
of serum for 15 h. Expression of Myc-E2F7 inhibited the serum-induced
activation of the prE2F2 reporter in a dose-dependent manner (Figure 3.3B),
suggesting that E2F7 can block the ability of endogenous E2F complexes to
activate this reporter.
To determine whether the ability of E2F7 to repress E2F target gene
expression is E2F-dependent, we asked whether Myc-E2F7 could inhibit the
ability of E2Fs to transactivate E2F-responsive reporter constructs. In these
experiments, MEFs were transiently transfected with E2F1 and the prE2F2
reporter construct and increasing amounts of E2F7. As previously shown, E2F1
expression resulted in the activation of reporter gene activity by ~6-fold (Figure
3.3C). This E2F1-mediated activation was effectively inhibited by Myc-E2F7 in a
dose-dependent manner. These results suggest that E2F7 can act to inhibit E2Fmediated activation of target genes.
73
DISCUSSION
E2F plays an important role in the control of cell proliferation in many
different species, including mammals, flies, nematodes, amphibians, and plants.
Comparative evaluation of amino acid sequences reveal that the DBD of E2Fs
are evolutionary conserved and therefore represent an essential feature that
serves to identify E2F family members. Herein we describe the cloning and
characterization of a novel murine gene that contains this most conserved
domain common to all known E2Fs. Functional studies confirm that this novel
protein can bind to consensus E2F DNA recognition sequences, supporting its
classification as a genuine member of the E2F family, and we have thus named it
E2F7. Despite this designation, structural and functional data indicate that E2F7
functions in a different manner from the other E2F family members.
The most notable feature of E2F7 is that it contains two DBDs and lacks
the heterodimerization domain, suggesting that this protein recognizes E2F
consensus sites independently of a DP partner. This novel E2F member most
closely resembles the recently identified AtE2Fd–f subfamily members in A.
thaliana. Kosugi and Ohashi (2002) speculate that the tandem repeat of the
DBDs within the plant E2Fs might structurally mimic the DBD formed in E2F/DP
heterodimers. This speculation was supported by the fact that deletion of either
one of the two DBDs in AtE2Fd–f resulted in a complete loss of its ability to bind
DNA.
74
Like E2F6, E2F7 also lacks the C-terminal domains necessary for
transactivation and pocket protein binding, consistent with its inability to activate
known E2F-responsive genes. Instead, E2F7 is able to block the endogenous
E2F-dependent transcriptional activity as well as the transactivating activity of
overexpressed E2F1. The mechanism of transcriptional repression is unclear but
might involve competition between E2F7 and other E2Fs for the same DNA
target sites. E2F6 has been shown to actively repress gene expression by
interacting directly with polycomb group proteins through their association via its
marked box domain (Trimarchi et al., 2001). Although E2F7 does not possess a
similar marked box domain, it might contain a functionally related domain(s) that
could potentially recruit co-repressors to E2F-regulated promoters. Interestingly,
E2F7-mediated transcriptional repression appears to be target gene-dependent
because only a subset of known E2F-regulated promoters can be efficiently
repressed by E2F7 overexpression, indicating functional specificity for distinct
E2F targets. Whether this specificity is dictated by the selectivity of binding to
different E2F DNA recognition sites, by the recruitment of co-factors, or through
other mechanisms is not known.
The identification of E2F7 as a novel E2F family member adds a further
level of complexity to the control of E2F-dependent gene expression. It is
possible that by inhibiting the DNA binding capacity of the other known E2Fs
during the cell cycle, it might modulate cell cycle progression or might provide
cells with the ability to respond to additional external cues. In this respect, our
75
data show that the regulation of E2F7 expression is cell growth-dependent,
suggesting that E2F7 may play an important role in counter balancing E2Finduced gene expression, as has been demonstrated for the antagonistic
relationship between the Drosophila E2Fs (Stevaux et al., 2002). Although
measurement of E2F7 expression by either Northern, reverse transcriptasePCR, or reporter assays indicate that its regulation is cell growth-dependent,
analysis of the E2F7 gene product has yet to be carried out and must await the
generation of E2F7- specific antibodies. In contrast to other E2Fs, E2F7
overexpression led to an inhibition of cellular growth in primary MEFs,
accompanied by a moderate but reproducible accumulation of cells in the G2
phase. The E2F7-mediated accumulation of cells in the G2 phase may arise from
a decrease in G2 progression, a delay in the exit of cells from G2, or an inability
of cells to efficiently enter mitosis. Further investigation will be required to identify
the exact mechanism of cell growth inhibition mediated by E2F7 and to identify
the specific targets regulated by E2F7 that might mediate this effect.
Because E2F7 overexpression has the capacity to slow down cellular
growth, one could envision that E2F7 may function as a tumor suppressor gene
and that mutations within this gene might result in cancer formation. Although we
have yet to establish the true physiological function of E2F7, its broad expression
pattern suggests that it may provide an important contribution to the regulation of
E2F activities in multiple tissues during development and/or tumorigenesis.
76
Figure 3.1
Tissue-specific and cell cycle-dependent
expression of E2F7.
A, a mouse tissue blot from ORIGENE was either
hybridized with an E2F7 specific probe or a -actin
probe as a loading control. B and C, expression of E2F7
following serum stimulation of quiescent MEFs. The
cells were brought to quiescent by serum starvation and
stimulated to proliferate by the addition of fresh medium
with 12.5% serum. The samples were taken at the
indicated times post-stimulation, and Northern blot
analysis (B) as described under "Experimental
Procedures“. Glyceraldehyde-3-phosphate
dehydrogenase (GAPDH) (C) was measured and used
as internal expression control.
77
B
Figure 3.1
Tissue-specific and cell cycle-dependent
expression of E2F7.
78
Figure 3.2
Cell cycle regulation of the E2F7 promoter.
A, genomic organization of the E2F7 promoter. The genomic
region including exons 1 and 2 is shown together with the DNA
sequences immediately upstream of each exon. The E2F
recognition sites within the E2F7 upstream regulatory
sequences and the site where the 2-kb promoter fragment was
fused to the firefly luciferase reporter is illustrated. The asterisks
indicate the positions of the E2F-binding elements where
mutagenesis was carried as outlined under "Experimental
Procedures." B, analysis of the E2F7 wild-type promoter (wt prE2F7) and the E2F7 promoter containing mutated E2F binding
elements (mut pr-E2F7) following mitogenic stimulation. MEFs
were transfected with wt pr-E2F7-luciferase or mut pr-E2F7luciferase, together with a TK-Renilla luciferase vector as an
internal control as described in the text. Asynchronous
proliferating (P), density-arrested (D), and synchronized cell
populations were analyzed. Firefly luciferase activity was
normalized to Renilla luciferase activity. C, analysis of wt prE2F7 and mut pr-E2F7 during quiescence. The data represent
the average and standard deviation from three independent
experiments. D, the E2F7 promoter is activated by E2F1 and
E2F3. MEFs were transfected with 2 µg of the wt pr-E2F7luciferase construct, together with 0.5 µg of either pCDNA3E2F1 (E2F1), pCDNA3-E2F3 (E2F3), or pCDNA3 control vector
along with 2 µg of CMV- β-galactosidase and treated as
described under "Experimental Procedures." The luciferase
values were first normalized to β-galactosidase activity and
reported as fold induction relative to the values obtained from
transfections with a control expression vector. E, activation of
the E2F7 promoter by E2F1 is independent of the three E2Fbinding elements. MEFS were transfected with either 2 µg of wt
pr-E2F7 or mut pr-E2F7 and with either pCDNA3-E2F1 (E2F1)
or control pCDNA3 vector and with 2 µg of CMV- galactosidase. The luciferase values were normalized to galactosidase activity and shown as fold induction relative to the
values obtained with control vector.
79
Figure 3.2
Cell cycle regulation of the E2F7 promoter.
80
Figure 3.3
E2F7 has transcriptional repressor function.
A, E2F7 can block the activation of multiple E2F-responsive
promoters. MEFs were transiently transfected with 2 µg of the
indicated E2F-responsive reporter constructs along with
increasing amounts of pCDNA-MycE2F7 (myc-7) and 2 µg of
CMV- -galactosidase. Extracts from transfected cells were
prepared as already described, and the luciferase and galactosidase activities were measured. The normalized
luciferase activities are presented as the average fold activation
relative to the activity in extracts from cells transfected with the
reporter construct alone. B, E2F7 overexpression inhibits the
serum-induced activation of the E2F2 promoter. MEFs were
transiently transfected with 2 µg of pr-E2F2 along with increasing
amounts of pCDNA3-Myc-E2F7 or corresponding doses of the
pcDNA3 control vector (con) and 2 µgofthe -galactosidase
plasmid as an internal control. The cells were treated as
described under "Results," and the extracts were prepared and
assayed for luciferase and -galactosidase activities; the
luciferase activity was normalized to the corresponding galactosidase activity. C, E2F7 inhibits the E2F1-induced
activation of the E2F2 promoter. MEFs were transiently
transfected with the 2 µg of the prE2F2 reporter construct and
0.2 µg of the pCDNA3-E2F1, along with either increasing
amounts of pCDNA3-MycE2F7 or pcDNA3 control vector and 2
µg of the CMV- -galactosidase. The cells were brought to
quiescence and then assayed for luciferase and -galactosidase
activity. Luciferase activity was normalized to -galactosidase
activity.
81
Figure 3.3
E2F7 has transcriptional repressor function.
82
CHAPTER 4
THE E2F1-3 TRANSCRIPTION FACTORS ARE ESSENTIAL FOR CELLULAR
PROLIFERATION 3
ABSTRACT
The retinoblastoma tumour suppressor (Rb) pathway is believed to have a
critical role in the control of cellular proliferation by regulating E2F activities
(Dyson, 1998; Nevins, 1998). E2F1, E2F2 and E2F3 belong to a subclass of E2F
factors thought to act as transcriptional activators important for progression
through the G1/S transition (DeGregory et al., 1997). Here we show, by taking a
conditional gene targeting approach, that the combined loss of these three E2F
factors severely affects E2F target expression and completely abolishes the
ability of mouse embryonic fibroblasts to enter S phase, progress through mitosis
and proliferate. Loss of E2F function results in an elevation of p21Cip1 protein,
leading to a decrease in cyclin-dependent kinase activity and Rb
Reproduced with permission from Nature. 2001 Nov 22; 414(6862): 457-62. Wu L, Timmers C, Maiti
B, Saavedra HI, Sang L, Chong GT, Nuckolls F, Giangrande P, Wright FA, Field SJ, Greenberg
ME, Orkin S, Nevins JR, Robinson ML, Leone G. © 2001 Macmillan Magazines Ltd. The data
pertaining to the growth defect (using Flow cytometry), p21 upregulation (kinase assays and
Western Blots) were generated by Maiti B, under the supervision of Leone G.
3
83
phosphorylation. These findings suggest a function for this subclass of E2F
transcriptional activators in a positive feedback loop, through down-modulation of
p21Cip1, that leads to the inactivation of Rb-dependent repression and S phase
entry. By targeting the entire subclass of E2F transcriptional activators we
provide direct genetic evidence for their essential role in cell cycle progression,
proliferation and development.
INTRODUCTION
The delineation of a pathway controlling the progression of cells out of
quiescence, through G1 and into S phase, has been established (Dyson, 1998;
Nevins, 1998). Principal events in this pathway include the activation of cyclindependent kinases (CDKs), the coordinated phosphorylation of Rb and p130 by
cyclin-CDK complexes, and the subsequent release and accumulation of E2F
activities (Dyson, 1998; Nevins, 1998). Although E2F has an essential role in
control of cell growth during Drosophila development (Duronio et al., 1995;
Royzman et al., 1997), current knockout mouse models have failed to
demonstrate a similar requirement for any E2F family member in mammals (Field
et al., 1996; Humbert et al., 2000a; Humbert et al., 2000b; Leone et al., 2001;
Lindeman et al., 1998; Rempel et al., 2000; Yamasaki et al., 1996). One
interpretation of these observations is that under normal circumstances, loss of a
84
single E2F member can be functionally compensated by other related E2F
activities.
RESULTS AND DISCUSSION
Of the six known E2F family members, E2F1, E2F2 and E2F3 can
specifically interact with Rb, and their expression is cell-cycle regulated (Lees et
al., 1993; Leone et al., 1998). To test for functional redundancy among this
subclass of E2F family members, we generated and interbred E2F1, E2F2 and
E2F3 mutant mice (Field et al., 1996; Leone et al., 2001) (Figure 4.1), and found
that although E2F1-/-E2F2-/- mice were viable and developed to adulthood,
E2F1-/-E2F3-/- and E2F2-/-E2F3-/- animals died early during embryonic
development (Table 4.1), at or just before 9.5 embryonic days (E9.5), pointing to
a central role for E2F3 in mouse development.
To explore directly the potential role for this subclass of E2F transcription
activators in cellular proliferation, we measured the ability of cells deficient for
these E2F family members to proliferate. We introduced a conditional or floxed
E2F3 allele (E2F3f/f; Figure 4.1) into E2F mutant backgrounds and obtained
E2F1-/-E2F3f/f, E2F2-/-E2F3f /f andE2F1-/-E2F2-/-E2F3f /f embryos at the
predicted frequencies (Table 4.1 and data not shown), confirming that the floxed
85
E2F3 allele did not have any adverse effect on the development of animals.
Infection of mouse embryonic fibroblasts (MEFs), which were derived from these
embryos, with a retrovirus expressing the Cre recombinase resulted in the
specific deletion of the floxed exon 3 (Figure 4.1D).
To determine whether the loss of E2F function would lead to an
accumulation of cells in a specific phase of the cell cycle, TKO cells were
analysed for DNA content by flow cytometry. As shown in Figure 4.2B, control
virus-treated E2F1-/-E2F2-/-E2F3f/f cells, rendered quiescent by serum
deprivation, accumulated a G1 content of DNA, and could be efficiently induced
to enter the cell cycle after serum addition, as suggested by the timely
accumulation of an S phase content of DNA. The similar profiles of control- and
Cre-treated wild-type cells indicated that expression of Cre itself had little or no
effect on the ability of cells to proliferate or progress through the cell cycle
(Figure 4.2B). Unsynchronized TKO cell populations had a similar profile of DNA
content as control cells, except that most of these cells were also negative for
BrdU incorporation (Figure 4.2C, compare top and bottom samples; and data not
shown). Moreover, TKO cells failed to accumulate aG1 content of DNA on serum
deprivation and failed to respond to serum addition (Figure 4.2C). Together,
these findings show that ablation of all three E2F family members arrests cell
growth irrespective of cell cycle position, indicating a role for these E2Fs
throughout the cell cycle. Recent work also suggests a continuing role for E2F
during S phase that seems to be important for the regulation of mitotic regulators
86
and progression of cells through G2/M (Lukas et al., 1999). This view is
consistent with recent genome-wide analyses of cell-cycle-regulated genes,
where a large fraction of E2Fregulated gene targets were found to encode
proteins known to be involved in the progression of cells into G2 and through
mitosis (Ishida et al., 2001; Muller et al., 2001).
To identify relevant downstream activities that might be responsible for
mediating the growth arrest observed upon loss of E2F3, we assessed the status
of various inhibitors of the cell cycle. Whereas p53 or p27Kip1 remained
unchanged, p21Cip1 protein levels were markedly elevated in TKO cells (Figure
4.3A). This increase in p21Cip1 protein could be accounted for by a concomitant
increase in its levels of messenger RNA (Figure 4.3B). Consistent with these
findings, Cdk2-associated activities were markedly reduced and Rb was found to
be predominantly in its hypophosphorylated state (Figure 4.3A, Figure 4.3C).
Furthermore, cyclin-B-associated kinase activity was also significantly reduced in
TKO cells, even though a large population of the cells possessed either an S
phase or G2 content of DNA. These results might, at least in part, explain the
apparent growth arrest that occurs through various phases of the cell cycle,
namely in G1, S and G2/M of TKO cells. The fact that overexpression of cyclin
E/Cdk2 could drive TKO cells into S phase (Figure 4.3D) suggests that the
upregulation of p21Cip1 protein is probably a relevant consequence resulting
from the ablation of E2F1, E2F2 and E2F3.
87
The status of Rb-E2F-associated DNA binding activities were also
assessed in these cells. Consistent with previous studies, electrophoretic mobility
shift assays (EMSA) of lysates from control- or Cre-treated E2F1-/-E2F2-/E2F3f/f cells synchronized by serum deprivation, revealed the characteristic Rbrelated p130-E2F4/5 protein complexes (Leone et al., 1998). These G0-specific
complexes are thought to recruit histone deacetylase and remodelling activities,
and are presumed to mediate transcriptional repression of E2F target (Brehm et
al., 1998; Harbour et al., 2000; Luo et al., 1998; Magnaghi-Jaulin et al., 1998;
Weintraub et al., 1992; Weintraub et al., 1995; Zhang et al., 1999). In contrast to
control-treated MEFs, serum stimulation of TKO cells did not lead to the
dissociation and disappearance of p130-E2F4/5 complexes nor to the
appearance of the S-phase specific p107-E2F4 complex (Figure 4.3E). These
findings are consistent with the observed increase in p21Cip1 protein and
decrease in Cdk activity and Rb phosphorylation (Figure 4.3A, Figure 4.3C). We
propose that E2F activators mediate cell cycle progression by two mechanisms:
first, they can directly activate cell-cycle-dependent gene expression, and
second, they can activate a positive feedback loop, through inhibition of p21Cip1
protein accumulation, that would promote Rb phosphorylation and complete
derepression of Rb-E2F target genes. The decrease in E2F target gene
expression observed in TKO cells could be viewed to result from both the
continued Rb-mediated transcriptional repression and the absence of E2Fmediated transcription activation. Although our results establish the critical nature
88
of E2F1, E2F2 and E2F3 in the control of cellular proliferation, the relative
importance of E2F-mediated transcriptional repression versus activation remains
to be determined. The mechanism by which E2F family members regulate
p21Cip1 expression and its in vivo consequences will be an important topic of
future experimentation.
Although a role for E2F in the regulation of the cell cycle and proliferation
has been speculated for over a decade (Dyson, 1998; Nevins, 1998; Tsai et al.,
1998; Yamasaki et al., 1998; Ziebold et al., 2001), the complexity of the E2F
network has precluded previous loss-of-function mouse models from
demonstrating the essentiality for individual E2F family members in mammalian
cell proliferation. The ‘conditional' gene knockout strategy described here allowed
us to circumvent detrimental consequences that arise from the inactivation of
multiple E2F family members in mice. By making genetic modifications to the
entire E2F activator subclass, as opposed to individual members, we provide
direct genetic evidence suggesting that E2F activators are essential for cell cycle
progression, proliferation and development.
MATERIALS AND METHODS
Retroviral infections
Full-length complementary DNAs for Cre recombinase, Myc-tagged
versions of mouse E2F3a and E2F3b and HA-tagged versions of human E2F1,
89
E2F2 and E2F3a were subcloned into the pBabe retroviral vector containing
either a puromycin-(pBpuro), hygromycin-(pBhygro), or phleomycin-resistant
gene (pBbleo). High-titre viruses were produced by transient transfection of
retroviral constructs into the Phoenix-Eco packaging cell line as described
previously (Pear et al., 1993). We infected MEFs with the retrovirus using
standard methods. Infected cells were then selected for a total of five days in the
presence of one or more antibiotics using the following concentrations: 2.5 mgml1
for puromycin, 400 mgml-1 for hygromycin and 25 mgml-1 for phleomycin.
Serum starvation and serum stimulation
Subconfluent MEFs were synchronized by incubation in DMEM with 0.2%
FBS for either 60 h (for primary MEFs) or 72 h (for immortalized MEFs).
Synchronized cells were then stimulated to proliferate by the addition of DMEM
supplemented with 15% FBS. Cells were collected at different time points after
serum stimulation and were processed for BrdU incorporation assays (Leone G
et al., 1998), flow Cytometry (Leone G et al., 1998), histone H1 kinase assays
(Leone G et al., 1998), EMSA (Nevins et al., 1997), western blots, or northern
blots. For BrdU incorporation assays, we counted at least 300 4,6-diamidino-2phenylindole (DAPI) counter-stained nuclei for each time point.
Western blot analyses
90
Cell protein lysates were separated in SDS acrylamide gels and blotted
into polyvinylidene fluoride membranes. Blots were incubated in blotto buffer (5%
skim milk in Tris-buffered saline) with antibodies specific for E2F3 (SC-878,
Santa Cruz), phospho-Akt (Cell Signalling, no. 9271L), phospho-Erk (Cell
Signalling, no. 9101S), p21Cip1 (M-19 and C-19, Santa Cruz), p27Kip1
(Transduction Laboratory), p53 (14461C, PharMingen and Ab-1, Oncogene), Rb
(G3-245, PharMingen), c-Myc-epitope (C-33, Santa Cruz), HA epitope (Y-11,
Santa Cruz), cyclin B1 (GNS1, Santa Cruz) and Cdk2 (M2, Santa Cruz). The
primary antibodies were then detected using horseradish-peroxidase-conjugated
secondary antibodies and ECL reagent as described by the manufacture
(Amersham).
91
Figure 4.1
E2F3 conditional knock out strategy
A, Two alternative splicing forms (E2F3a and E2F3b) with
separate promoters (bent arrows) are encoded by the E2F3
locus15. The solid bar represents a SpeI–EcoRV fragment used
as a Southern probe. loxP sites are indicated as solid triangles;
thin arrows represent PCR primers. DT, diphtheria toxin; TK,
thymidine kinase; RV, EcoRV. B, PCR analysis of mice with the
indicated genotypes. M, DNA marker; C, negative control.
92
A
B
Figure 4.1
E2F3 conditional knock out strategy
93
A
B
C
Figure 4.2
Growth defect in E2F3 single or combinatorial knock out
MEFs
A, BrdU incorporation assays of MEFs that were infected with a
control (open boxes) or a Cre retrovirus. B, C, Cell cycle analysis
by FACS for wild-type MEFs (C) or E2F1-/-E2F2-/-E2F3f/f MEFs
(C) that were infected with a control (top panels (-)) or a Cre
retrovirus (bottom panels (+)). In C, a fraction of cells had
become tetraploid by the time the experiment was performed, as
indicated by the appearance of two distinct peaks in serumstarved samples (0 h). Hatched areas indicate the percentage of
cells (S) containing an S-phase content of DNA.
94
Figure 4.3
Loss of E2F3 activates checkpoint via p21
A, Western blots of control- or Cre-retrovirus-infected E2F1-/E2F2-/-E2F3f/f MEFs using antibodies against the indicated cell
cycle regulators. Rb-PP, phosphorylated form of Rb. B, p21
Northern blot of E2F1-/-E2F2-/-E2F3f/f MEFs that were treated as
in A. P, proliferating conditions. C, Kinase assays of E2F1-/E2F2-/-E2F3f/f MEFs treated as in A. Ig, rabbit immunoglobulinas controls. The graph represents the relative kinase activity. D,
BrdU incorporation assays of E2F1-/-E2F2-/-E2F3f/f MEFs that
were infected with a control (-) or a Cre retrovirus (+). Serumstarved cells were infected with adenoviruses expressing cyclin
E and Cdk2 (E) or with a control adenovirus (C). Adenovirusinfected cells were incubated with BrdU in either 0.2% serum (-)
or 15% serum (+) for 24 h, and processed for
immunohistochemistry. E, E2F1-/-E2F2-/-E2F3f/f MEFs were
treated as in A, and were assayed for E2F DNA-binding activity
by EMSA using an E2F-specific 32P-labelled DNA probe.
95
D
A
B
E
C
Figure 4.3
Loss of E2F3 activates checkpoint via p21
96
Table 4.1
Genotypic Analysis of E13.5 embryos derived
from crosses of E2F mutant animals
97
CHAPTER 5
SURVIVIN IS ESSENTIAL FOR PROLIFERATION AND COLLABORATES
WITH C-MYC AND H-RAS (V12) IN THE TRANSFORMATION OF MOUSE
EMBRYONIC FIBROBLASTS. 4
ABSTRACT
Survivin is an important molecular target in cancer, in part due to its
predominant expression in cancer cells in comparison with normal differentiated
cells. Current evidence suggests that survivin’s dual function as an anti-apoptotic
protein and a chromosomal passenger protein makes it a factor favoring cancer
progression. The role of survivin in cellular proliferation and transformation is not
clearly understood. To this end, we made use of primary mouse embryonic
fibroblasts (MEFs), conditionally deleted for survivin. Immunofluorescence
studies revealed localization of endogenous survivin in the wild-type MEFs during
mitosis, similar to previous observations in other cell types, in agreement with the
role of survivin as a chromosomal passenger protein in these cells. When
survivin was deleted by virally mediated Cre recombinase expression, we
4
All data were generated by Maiti B under the supervision of Altura RA, except where indicated
otherwise.
98
observed severe proliferation defects, manifest as an increase in the number of
polyploid cells and a massive increase in nuclear diameter likely secondary to a
mitotic defect. In contrast to the cell death phenotype reported in other cell types
following Survivin deletion, we did not detect any significant increase in cellular
apoptosis or caspase activity. To gain an understanding of survivin’s role in
transformation, wild-type primary MEFs were engineered to express known
oncogenes (c-myc or H-rasV12) in combination with survivin. Our preliminary
data suggest that survivin collaborates with c-myc and H-rasV12 to transform
wild-type primary MEFs. This is the first report of a collaboration of survivin with
another oncogene in transformation. The fact that survivin can potentiate the
activity of other oncogenes is a novel finding and suggests a potential
mechanism for this gene in tumor cell initiation and growth. In conclusion, these
data largely support targeting a disruption of survivin in cancer therapy and
uncover a novel role of survivin in the context of transformation.
INTRODUCTION
Survivin is an unusual protein with dual functions in mitosis and apoptosis
(Wheatley et al., 2005). During mitosis, survivin acts as a chromosomal
passenger protein forming a complex with Aurora-B kinase, inner centromere
protein (INCENP) and Borealin/Dasra-B (Vader et al., 2005). Survivin is a crucial
mediator that targets the chromosomal passenger complex in a spatial and
99
temporal manner, an essential process to maintain high fidelity chromosomal
segregation during mitosis (Vader et al., 2005). The spindle assembly checkpoint
is dependent on survivin, as evident by the significant reduction of BubR1 (which
regulates kinetochore-microtubule attachments and is a part of the mitotic
checkpoint complex, inhibiting anaphase promoting complex/cyclosome, APC/C)
at the kinetochores in prometaphase (Lens et al., 2006). Targeting survivin by
RNA interference in euploid human lung fibroblast cells (IMR-90) or a retinal
pigment epithelial (RPE) cell line results in defects of both chromosomal
segregation and cytokinesis but not enhancement of cell death (Yang et al.,
2004). Targeting of embryonic stem cells by disrupting the survivin gene locus
also results in a similar phenotype (Uren at al., 2000; Conway et al., 2002). In
contrast, conditional targeting of survivin in normal neural stem cells results in
massive apoptosis indicating a cell-type specific role for survivin as a mitotic
regulator or anti-apoptotic protein in untransformed cells (Jiang et al., 2005).
The expression pattern of survivin is under tight temporal and spatial
control. It is abundantly expressed during embryogenesis, whereas it is
undetectable in most terminally differentiated adult tissues (Altieri, 2006).
Proliferating adult tissues that do express survivin include early hematopoietic
cells, T-lymphocytes, gastric epithelium, vascular endothelium and
spermatogonia (Li and Brattain, 2006). In addition, survivin is highly expressed in
virtually every human cancer making it a highly explored molecular target for anticancer therapy (Altieri, 2006).
100
Targeting of survivin by RNA interference in a multitude of cancer cell
lines leads to an increase in susceptibility to DNA-damage induced programmed
cell death, such as by chemicals or radiation. This indicates that high levels of
survivin mediate resistance to chemotherapy and radiation (Li and Ling, 2006).
This is clinically significant in that patients harboring tumors expressing high
survivin have poorer clinical outcomes (Li and Ling, 2006). Current evidence
suggests that survivin plays an essential role in tumor maintenance by facilitating
proliferation, angiogenesis, multi-drug resistance and by preventing apoptosis
(Altieri, 2006). However, whether survivin plays a role in the initiation or
establishment of tumors still remains unclear. Lines of evidence that support a
role for survivin in this process include the following. High levels of expression of
chromosomal passenger proteins (CPPs) such as Aurora-A, Aurora-B, survivin
and INCENP in human tumors correlate with the degree of aneuploidy and
genomic instability (Nguyen and Ravid, 2006). According to the ‘genomic
instability theory of cancer’, chromosomal instability early in tumor formation
leads to loss of tumor suppressor genes and/or gain of oncogenes, thus driving
malignant transformation (Nguyen and Ravid, 2006). Experimentally, Tatsuka et
al., (2005) showed that exogenous expression of Aurora-A (mammals have three
aurora kinases – A, B and C) potentiated transformation induced by G12V
mutated H-Ras expression in mouse 3T3 cells. The anti-apoptotic functions of
the chromosomal passenger protein, survivin, combined with its potential to
cause genomic instability when aberrantly expressed, led us to hypothesize a
101
role for survivin in tumor initiation. To enable the efficient targeting of survivin in
cancer it is imperative to understand the interplay of survivin with other signaling
pathways that co-exist in cancer cells. Typically, genetic analysis of a cancer cell
reveals a gain of function (g-o-f) by mutation/amplification of oncogenic pathways
and/or a loss of function (l-o-f) by mutation/silencing of tumor suppressor
pathways. In the current study, I made use of two well-studied oncogenes c-Myc
and H-Ras V12 to explore survivin’s potential collaborative role in cancer
initiation.
c-Myc is a nuclear transcription factor which is aberrantly expressed, via
genetic or epigenetic means including amplification and translocation, in up to
50% of all human cancers (Arvanitis and Felsher, 2006). Physiologically, c-Myc,
following dimerization with its partner Max, binds DNA and activates genes
involved in proliferation, differentiation and apoptosis (Arvanitis and Felsher,
2006). c-Myc plays an indispensable role in cellular proliferation, as evidenced by
a profound and stable growth defect of Rat1A fibroblasts, deficient in c-Myc
(Berns et al., 2000). Members of the Ras gene family comprise the most
frequently mutated genes found in any human cancer (Giehl, 2005). Point
mutations in the Ras gene itself resulting in a constitutively active form (for
example H-RasV12) of the Ras proteins, protein overexpression or dysregulation
of growth factor receptor tyrosine kinase (RTK) pathways (upstream of Ras)
result in activation of the Ras signaling in a vast majority of human cancers
(Giehl, 2005; Cox and Der, 2002). Ras proteins, which are membrane bound
102
GTP-ases, cycle between an active GTP-bound and inactive GDP-bound state,
acting as a molecular switch to modulate cellular proliferation, differentiation,
apoptosis, motility and phagocytosis via a wide range of effector pathways that
include Raf kinase, PI3K (phosphatidylinositol 3-kinases) and mitogen-activated
protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) kinase (Giehl,
2005).
Although expression of Ras or Myc proteins by themselves lead to p53dependent premature senescence or apoptosis of wild type primary mouse
embryonic fibroblasts respectively; when co-expressed, Ras and Myc collaborate
to transform these cells (Land et al., 1983; Weinberg et al., 1997). Recent
evidence suggests that survivin is upregulated by H-Ras G12V and survivin
facilitates H-Ras G12V induced tranformation and foci formation in rat embryonic
3T3 cells (Sommer et al., 2003). To gain a better understanding of the role of
survivin in tumor initiation and the interplay of co-existing oncogenic pathways
with survivin in cancer, I used transformation assays using primary MEFs that
concomitantly express survivin and another oncogene (H-RasV12 or c-Myc).
Since it had not been characterized previously, I initially investigated the role of
survivin in the proliferation and survival of primary MEFs, using MEFs
conditionally deleted for survivin by a Cre-lox-P system. These data show that
survivin is essential for the proliferation but not for the survival of primary MEFs.
In addition, evidence is provided that survivin is essential for the maintenance of
the Myc/Ras transformed phenotype. These preliminary data supports
103
collaboration between survivin and the oncogenes c-Myc and H-RasV12 in the
initiation of transformation.
MATERIALS AND METHODS
Construction of the plasmids
Survivin cDNA with myc-His tag at the 3’end was PCR amplified from the
pcDNA4TM/TO/myc-His B (Invitrogen)- survivin construct described previously
(Caldas et al., 2005) and subcloned in the SnaB1 site of retroviral pBabe vector
containing a Hygromycin resistance cassette. c-Myc and H-RasV12 constructs in
pBabe vectors with Hygromycin or puromycin resistance cassettes were
generous gifts from Leone G.
Generation of the Mouse embryonic fibroblasts and in vitro cell culture
Mice containing a conditional floxed survivin (survivinlox/lox) allele have
been described elsewhere (Xing et al., 2004). These mice were interbred to
generate homozygous survivin f/f embryos.
Primary mouse embryonic fibroblasts were isolated from E18.5 survivin f/f
embryos using standard methods. For the experiments, the MEFs were grown in
DMEM containing 15% FBS. If indicated, the MEFs were starved in 0.2% FBS
104
containing DMEM for 48 hours and restimulated using DMEM containing 15%
FBS. MEFs were grown at 370C and 5.5% CO2.
Viral infections
High titer retrovirus was produced from the transient transfection of pBabe
constructs in to Phoenix-Eco packaging cell lines as described elsewhere (Pear
et al., 1993). Following infection, the MEFs were selected using the appropriate
antibiotic(s) at the following concentrations - 2.5 μgml-1 for puromycin, 400 μgml-1
Hygromycin for a period of 48-72 hours.
Adenovirus expressing cre recombinase (Ad-CMV-Cre) was purchased
from Vector Biolabs. The adenoviral GFP (rAD.eGFP) was made by the viral
vector core laboratory at the Columbus Children’s Research Institute and was a
generous gift from Kaspar B. Both the virus were used at 40 MOI.
Immunohistochemistry and confocal microscopy
MEFs were grown on glass coverslips until harvested. Subsequently, they
were fixed in 4% paraformaldehyde for 10 min at room temperature. The cells
were then permeabilized in 0.5% Triron X-100 for 10 min at room temperature.
Following permeabilization, the cells were washed with Phosphate buffered
saline (PBS) and blocked with 5% normal goat serum for 1 hour at room
temperature. Then the cells were incubated with primary antibody overnight at
40C. The primary antibodies used include polyclonal FL142 anti survivin antibody
105
from Santa Cruz Biotech and monoclonal alpha- tubulin from Sigma at the
concentrations of 1μg/ml. Next, the cells were incubated with 1μg/ml of
secondary antibodies goat anti-mouse-FITC (Santa Cruz Biotech) and goat antirabbit-Biotin (Santa Cruz Biotech) for one hour at room temperature. Then the
cells were washed and further incubated with tertiary NeutrAvidin Texas Red
(Molecular Probes) and Hoechst 33258 for nuclear staining. Finally, the cover
slips were mounted using Vectashield (Vector Labs) and analyzed by confocal
microscopy on Zeiss LSM 510 META using the Plan Apochromat 63/1.4 Oil/DIC
objective.
Cell proliferation assay
The MEFs were plated in 6 well dishes at starting concentration of 15X103
cells/well, in triplicates. The cells were harvested and counted every 24 hours for
7 days (Jiang, Y).
In vitro colony formation assay
Following retroviral transduction and selection, cells were plated at a
density of 30X103 cells/ 100mm tissue culture dish and allowed to grow in
15%FBS/DMEM for 7-10 days to let the colonies grow. After this period, they
were harvested, fixed with 70% ethanol and stained with 5mg/ml crystal violet in
20% methanol.
106
Caspase assay
The caspase-9 activity was measured from 50μg of protein extract of each
cell type, in triplicates, using Caspase-Glo Assay kits (Promega, Madison, WI).
BrdU/PI Flow cytometry
The cells were pulsed for one hour with 10μM BrdU following which they
were harvested, fixed in 70% ethanol and stained with 10μg/ml PI (Propidium
Iodide)/RNAse (BD Biosciences) and 1μg/ml Anti-BrdU-FITC antibody using the
manufacturer’s protocol
(http://www.bdbiosciences.com/pharmingen/protocols/BrdU_Incorporation.shtml).
Subsequently, the data was collected using BD FACSDiva and analyzed using
BD FACSDiva software.
RESULTS AND DISCUSSION
Previous studies have shown that survivin has cell-type specific roles as
an anti-apoptotic protein and/or mitotic regulator in vitro and in vivo ((Jiang et al.,
2005; Yang et al., 2004). To determine the predominant function of survivin in
primary MEFs, we used MEFs isolated from E18.5 embryos containing the
homozygous floxed allele of survivin (Svnf/f). The knock out strategy and the
position of the lox-P sites is outlined in Figure 5.1A. Using Cre recombinase that
was delivered via retroviral or adenoviral infection, the survivin gene was deleted
107
between the lox-P sites, in the majority of the cell population, as evident by PCR
using the primer set P1, P2 and P4 (Figure 5.1B).
To determine if the deletion of survivin would have any effect on cellular
proliferation, we performed growth curves over a seven-day period on MEFs
transduced by cre or control virus. Results showed that MEFs infected with
control virus grew exponentially under the experimental conditions, however
MEFs lacking survivin following expression of cre-recombinase, failed to
proliferate (Figure 5.2A, Jiang Y). To determine if the failure of cell growth (Figure
5.2A) was a consequence of increased cell death we performed caspase activity
assays on the MEF cells. As shown in Figure 5.2B (Jiang Y), MEFs lacking
survivin had a minimal increase in caspase activity and no increase in TUNEL
positive cells when compared with the control MEFs (Jiang Y, data not shown).
This data strongly argues against an anti-apoptotic role for survivin in the MEF
cells.
To understand the mechanism of the growth defect in the MEFs lacking
survivin, we evaluated the subcellular localization pattern of survivin during
mitosis, in the primary MEFs. Survivin localized to the kinetochore during
prophase (Figure 5.4A) to the spindle midzone during anaphase (Figure 5.4B)
and to the midbody during cytokinesis (Figure 5.4C) – a pattern consistent with
its function as a chromosomal passenger protein in the survivin f/f cells. MEFs
lacking survivin exhibited enlarged, irregular and multiple nuclei (Figure 5.4D,
Figure 5.4E). Quantitative analysis revealed the maximum nuclear diameter of
108
the control survivin f/f MEFs to be 10 – 20μm with up to a four fold increase in the
nuclear diameter of survivin -/- MEFs. Flow cytometric analysis performed
following serum starvation and restimulation, showed that survivin -/- cells
entered S-phase normally, as evident by >10 fold increase in BrdU incorporation
at 20 hours post stimulation, similar to the control cells (Figure 5.5B and Figure
5.5C). However, in the survivin -/- cells, a polyploid (> 4N nuclear content)
population of cells (Figure 5.5C and Figure 5.5D) accumulated at a high rate. The
accumulation of large and multinucleated cells, in combination with polyploidy,
suggests a proliferation defect, secondary to a failure in cytokinesis in the
survivin deficient MEFs.
Survivin is important in the maintenance of the human cancer cell
transformed phenotype, as demonstrated by human tumor studies in vitro and in
vivo. (Altieri, 2006; Li and Ling, 2006). We hypothesized that survivin would also
prove to be essential for the maintenance of transformation in MEFs transformed
by c-Myc and H-RasV12. Towards this end, the human oncogenes c-Myc and HRasV12 were co-expressed by retroviral transduction which resulted in the
transformation of the survivin f/f primary MEFs (Land et al., 1983; Weinberg et
al., 1997). Subsequently, I deleted the mouse survivin gene using adenoviral
expression of Cre recombinase (Ad-Cre). Twenty-four hours following infection of
Ad-cre or Ad-GFP as a control, the cells were plated for colony formation in 100
mm tissue culture dishes at a concentration of 30X103 cells/plate. 7-10 days after
plating, the colonies were harvested and stained for visualization (see materials
109
and methods). While the myc/Ras transformed cells infected with Ad-GFP gave
rise to abundant colonies with a transformed morphology, this colony formation
was about eight-fold reduced in the cells treated with Ad-cre (Figure 5.5 A and
Figure 5.5 B). These data suggests an absolute requirement of survivin for the
maintenance of the myc/ras transformed phenotype.
To determine if survivin plays a role in the initiation of transformation, I coexpressed (using retroviral transduction method) human survivin with either cMyc or H-Ras V12 human oncogenes in primary MEFs and subjected them to
similar in vitro colony formation assays as described above. While cells
transduced with control virus or survivin alone did not give rise to transformed
colonies, those cells co-expressing survivin and c-Myc, or survivin and H-RasV12
(used as positive control) gave abundant transformed looking colonies implying
an important role of survivin in cancer initiation (Figure 5.6).
In summary, from these data we can conclude that survivin is essential for
the proliferation of the primary MEFs but does not play a role in their survival.
Survivin is also required for the maintenance of a transformed phenotype in
MEFs transformed with c-Myc and H-RasV12. Furthermore, these preliminary
results suggest that survivin plays a role in the initiation of tumor formation.
Confirmatory experiments, including soft agar colony formation assay and tumor
assays in nude mice are currently underway to further characterize the
transformed phenotype of MEFs co-expressing survivin and c-Myc or survivin
and H-RasV12.
110
These data, demonstrating the oncogenic potential of survivin is novel and
has far reaching implications with respect to therapeutic targeting of survivin in
cancer cells. It also supports a cautious approach to the proposed use of survivin
as an anti-apoptotic therapeutic agent in the treatment of human degenerative
diseases.
111
Figure 5.1
Conditional knockout strategy of Survivin in
primary mouse embryonic fibroblasts
A, The genomic locus of mouse survivin is shown
with the exons represented by blue boxes. The
positions of the two lox-P sites are indicated as well
as the primers used to amplify the floxed or deleted
allelle. B, A representative PCR illustrating the
products of predicted sizes obtained after conditional
deletion of survivin. The floxed allele gives a 560 bp
product while the deleted allele gives a 425 bp
product.
112
A
B
Svn f/f MEFs
Virus: none
Control
Cre
Figure 5.1
Conditional knockout strategy of Survivin in
primary mouse embryonic fibroblasts
113
Figure 5.2
Survivin is essential for proliferation but not for
survival of primary MEFs
A, Growth curve of Svn f/f primary MEFs infected with
control (blue) or cre (pink) retrovirus. The y-axis
represents the number of cells while the x-axis
represents days. B, Caspase 9 activity assay in the
same MEFs described in A. The y-axis represents
luminescence detected in a spectrophotometer using
the Caspase-glo assay kit (Promega).The cells lacking
survivin exhibit a minimal increase in caspase 9
activity.
114
A
Growth Curves
200000
Cell number
control
150000
Cre
100000
50000
0
0
1
2
3
4
5
6
7
Days
B
Luminescence
Caspase Activity
14000
12000
10000
8000
6000
4000
2000
0
Control virus
(survivin f/f)
Cre virus
(survivin -/-)
Figure 5.2
Survivin is essential for proliferation but not for
survival of primary MEFs
115
Figure 5.3
Primary MEFs lacking survivin display an abnormal
nuclear morphology
A,B and C, Survivin (Texas Red) localization relative to DNA
(Hoechst 33258, the blue color has been changed to white for
better visualization) α-tubulin (FITC) in control cells at
prophase (survivin localized to kinetochore), anaphase
(survivin at spindle midzone) and cytokinesis (survivin at
midbody). The survivin localization is marked by white
arrow(s) and 5 μm scale is shown. D and E, Aberrantly
enlarged nuclear morphology survivin deficient primary
MEFs. F, a control cell in interphase shown as a reference.
G, Quantification of the maximum nuclear diameter in control
and cre infected MEFs measured using Zeiss LSM image
browser. The y-axis represents nuclear diameter in μm.
116
A
B
C
Figure 5.3
Primary MEFs lacking survivin display an abnormal
nuclear morphology
117
Figure 5.3 (contd.)
D
E
F
118
Figure 5.3 (contd.)
G
Maximum nuclear diameter in μ m
90
80
70
60
50
control Svn f/f
Cre Svn f/f
40
30
20
10
0
119
Figure 5.4
Loss of Survivin results in polyploidy
BrdU (FITC)/ PI flow cytometric analysis in control
viral (A) or cre viral infected cells (C) serum starved
for 48 hours and control virus (B) or cre virus
infected cells (D) 20 hours after restimulation (B).
P2 indicates BrdU positive cells in S-phase, P3
includes cells with 2N ploidy, P4 shows cells with
4N DNA content and P5 has cells with ploidy of 8N
and above . The percentages of cells in P5 is
indicated.
120
survivin f/f
A
survivin f/f
0 hours post stimulation
20 hours post stimulation
4.8%
8.6%
P5
P5
survivin -/C
B
survivin -/-
D
0 hours post stimulation
20 hours post stimulation
13.1%
20.6%
P5
P5
Figure 5.4
Loss of Survivin results in polyploidy
121
Figure 5.5
Survivin is essential for the maintenance of
the myc/ras transformed phenotype in MEFs
A, In vitro colony formation assay (see materials
and methods) on Ad-GFP and Ad-cre infected
MEFs which have been transformed by c-Myc and
H-RasV12. The colony formation is severely
compromised when survivin is deleted by Ad-cre,
B, Quantification of colonies showing about seven
fold decrease in survivin deleted cells
122
c-Myc+H-Ras(V12) transformed MEFs
Ad-GFP infected
Ad-Cre infected
Figure 5.5
Survivin is essential for the maintenance of
the myc/ras transformed phenotype in MEFs
123
c-Myc +
H-Ras(V12)
Survivin
Control
Survivin +
Survivin +
c-Myc
H-Ras(V12)
Figure 5.6
Survivin collaborates with c-Myc and H-RasV12 in
transforming wt primary MEFs
In vitro colony formation assay (see materials and
methods) on primary MEFs transduced by retrovirus as
indicated. No transformed colonies grew from cells
infected with control virus or survivin alone. Abundant
transformed colonies grew from cells co-expressing
Survivin+c-Myc, survivin+H-RasV12 or c-Myc+HRasV12
indicating survivin can collaborate with other oncogenes
in initiating transformation
124
CHAPTER 6
DISCUSSION
E2F7 and E2F8 form unique sub-class in the murine E2F family
The E2F family of transcription factors is conserved from plants to
mammals with the exception of yeast. The E2F family increases in complexity
along the evolutionary ladder as exemplified by the fact that Drosophila has only
two E2F family members – one activator and one repressor, whereas the
mammalian E2Fs to date have at least ten E2F transcripts arising from eight
distinct genomic loci. E2F7 and E2F8 are two unique family members based on
the presence of duplicated highly conserved DNA binding domains and an
absence of any other domain conserved across the E2Fs 1-6. Structural analysis
suggests that the presence of two DNA binding domains in tandem abrogates the
requirement for dimerization with DP for DNA binding. This is also supported by
the fact that the integrity of both DNA binding domains is required for DNA
125
binding by E2F7 and E2F8. The expression of E2F7 and E2F8 are cell cycle
regulated at least in part via E2F mediated auto-regulation. The tissue
expression patterns of E2F7 and E2F8 overlap in adult mouse indicating a
common functional theme for these last two identified murine E2F family
members. When overexpressed in mouse embryonic fibroblasts (MEFs), E2F7
and E2F8 inhibit cellular proliferation, with concomitant downregulation of various
E2F target genes, suggesting a potential tumor suppressor function of these
proteins. Future studies will give mechanistic insights in to E2F7-8 mediated
repression as well as their roles in development, differentiation and
tumorigenesis.
Survivin is essential for the proliferation and transformation of MEFs
Survivin is an E2F transcriptional target and an essential gene in murine
development. During cell division, survivin ensures high fidelity chromosomal
segregation and cytokinesis. MEFs conditionally deleted for survivin exhibit a
severe proliferation defect with increased polyploidy suggesting a defect in
cytokinesis. Survivin also plays an anti-apoptotic role in various normal and
malignant tissues, but this function is not apparent in the MEFs since there is no
increased caspase activity or apoptosis evident after conditional deletion of
survivin. Survivin is abundantly and ubiquitously expressed during
embryogenesis, but is absent in the terminally differentiated adult tissue.
126
However, survivin is highly expressed in virtually every human cancer making it
an important molecular target in cancer therapy. Conditional deletion of survivin
in MEFs transformed by overexpression of c-Myc and H-RasV12, reveal a
requirement for this gene in the maintenance of a transformed phenotype in
these cells. This is consistent with what has been observed in several different
human cancer cells after disruption of survivin with RNA interference. The
requirement for proliferation and transformation of MEFs support current efforts
and clinical trials through targeting of survivin in human tumors. In addition,
preliminary evidence presented here, suggests a novel role for survivin in
initiating transformation in MEFs. Soft agar colony formation assay and in vivo
tumor formation in nude mice are currently ongoing to rigorously test the
oncogenic potential of survivin. These data strongly advocate a cautious
approach in the proposed use of survivin gene therapy for human degenerative
diseases.
127
LIST OF REFERENCES
Altieri DC. (2003) Survivin, versatile modulation of cell division and apoptosis in
cancer. Oncogene. Nov 24;22(53):8581-9.
Altieri DC. (2004) Molecular circuits of apoptosis regulation and cell division
control: the survivin paradigm.J Cell Biochem. Jul 1;92(4):656-63.
Altieri DC. (2006) Targeted therapy by disabling crossroad signaling networks:
the survivin paradigm. Mol Cancer Ther. Mar;5(3):478-82. Review.
Altura RA, Olshefski RS, Jiang Y, Boue DR. (2003) Nuclear expression of
Survivin in paediatric ependymomas and choroid plexus tumours
correlates with morphologic tumour grade. Br J Cancer. Nov 3;89(9):17439
Arvanitis C, Felsher DW. (2006 ) Conditional transgenic models define how MYC
initiates and maintains tumorigenesis. Semin Cancer Biol. Aug;16(4):3137.
Beaudoing, E., Freier, S., Wyatt, J. R., Claverie, J.-M., and Gautheret, D. (2000)
Genome Res. 10, 1001–1010
Berns K, Hijmans EM, Koh E, Daley GQ, Bernards R. (2000) A genetic screen to
identify genes that rescue the slow growth phenotype of c-myc null
fibroblasts.Oncogene. Jul 6;19(29):3330-4.
Blanc-Brude OP, Mesri M, Wall NR, Plescia J, Dohi T, Altieri DC. Therapeutic
targeting of the survivin pathway in cancer: initiation of mitochondrial
128
apoptosis and suppression of tumor-associated angiogenesis. Clin Cancer
Res. 2003 Jul;9(7):2683-92.
Brehm, A. et al. (1998) Retinoblastoma protein recruits histone deacetylase to
repress transcription. Nature 391, 597-601.
Brehm, A., Miska, E. A., McCance, D. J., Reid, J. L., Bannister, A. J., and
Kouzarides, T. (1998) Retinoblastoma protein recruits histone deacetylase
to repress transcription. Nature 391, 597–601
Caldas H, Jiang Y, Holloway MP, Fangusaro J, Mahotka C, Conway EM, Altura
RA.( 2005) Survivin splice variants regulate the balance between
proliferation and cell death.Oncogene. Mar 17;24 (12):1994-2007
Caldas H, Jiang Y, Holloway MP, Fangusaro J, Mahotka C, Conway EM, Altura
RA. (2005) Survivin splice variants regulate the balance between
proliferation and cell death.Oncogene. Mar 17;24(12):1994-2007.
Cam, H., and Dynlacht, B. D. (2003) Emerging roles for E2F: Beyond the G1/S
transition and DNA replication Cancer Cell 3, 311–316
Ceol, C. J., and Horvitz, H. R. (2001) dpl-1 DP and efl-1 E2F Act with lin-35 Rb to
Antagonize Ras Signaling in C. elegans Vulval Development Mol. Cell 7,
461–473
Christensen J, Cloos P, Toftegaard U, Klinkenberg D, Bracken AP, Trinh E,
Heeran M, Di Stefano L, Helin K. (2005) Characterization of E2F8, a novel
E2F-like cell-cycle regulated repressor of E2F-activated transcription.
Nucleic Acids Res Sep 22;33(17):5458-70.
Colnaghi R, Connell CM, Barrett RM, Wheatley SP. (2006) Separating the antiapoptotic and mitotic roles of survivin. J Biol Chem. Nov 3;281(44):334506.
Conway EM, Pollefeyt S, Steiner-Mosonyi M, Luo W, Devriese A, Lupu F, Bono
F, Leducq N, Dol F, Schaeffer P, Collen D, Herbert JM. (2002) Deficiency
of survivin in transgenic mice exacerbates Fas-induced apoptosis via
mitochondrial pathways. Gastroenterology. Aug;123(2):619-31.
Cox AD, Der CJ. (2002) Ras family signaling: therapeutic targeting. Cancer Biol
Ther. Nov-Dec;1(6):599-606.
de Bruin A, Maiti B, Jakoi L, Timmers C, Buerki R, Leone G. Identification and
characterization of E2F7, a novel mammalian E2F family member capable
129
of blocking cellular proliferation. J Biol Chem. 2003 Oct 24;278(43):420419. Epub 2003 Jul 31.
Deb-Basu D, Aleem E, Kaldis P, Felsher DW. (2006) CDK2 is required by MYC
to induce apoptosis. Cell Cycle. Jun;5(12):1342-7.
DeGregori J, Johnson DG. (2006) Distinct and Overlapping Roles for E2F Family
Members in Transcription, Proliferation and Apoptosis. Curr Mol Med.
Nov;6(7):739-48.
DeGregori, J., Leone, G., Miron, A., Jakoi, L.and Nevins, J. R. (1997) Distinct
roles for E2F proteins in cell growth control and apoptosis. Proc. Natl
Acad. Sci. USA 94, 7245-7250.
Di Stefano L, Jensen MR, Helin K. (2003) E2F7, a novel E2F featuring DPindependent repression of a subset of E2F-regulated genes. EMBO J. Dec
1;22(23):6289-98.
Dohi T, Beltrami E, Wall NR, Plescia J, Altieri DC. (2004) Mitochondrial survivin
inhibits apoptosis and promotes tumorigenesis. J Clin Invest;114: 1117–
27.
Duronio,R. J., O'Farrell, P. H., Xie, J E., Brook, A.and Dyson, N. (1995). The
transcription factor E2F is required for S phase during Drosophila
embryogenesis. Genes Dev. 9, 1445-1455
Dyson, N. (1998) The regulation of E2F by pRB-family proteins. Genes Dev. 12,
2245–2262
Eckelman BP, Salvesen GS, Scott FL. (2006) Human inhibitor of apoptosis
proteins: why XIAP is the black sheep of the family. EMBO Rep.
Oct;7(10):988-94.
Fangusaro JR, Caldas H, Jiang Y, Altura RA. (2006) Survivin: an inhibitor of
apoptosis in pediatric cancer.Pediatr Blood Cancer. Jul;47(1):4-13.
Ferreira, R., Magnaghi-Jaulin, L., Robin, P., Harel-Bellan, A., and Trouche, D.
(1998) The three members of the pocket proteins family share the ability
to repress E2F activity through recruitment of a histone deacetylase. Proc.
Natl. Acad. Sci. U. S. A. 95, 10493–10498
Field, S. J. et al. (1996). E2F-1 functions in mice to promote apoptosis and
suppress proliferation. Cell 85, 549-561
130
Flemington, E. K., Speck, S. H., and Kaelin, W. G., Jr. (1993) E2F-1-mediated
transactivation is inhibited by complex formation with the retinoblastoma
susceptibility gene product. Proc. Natl. Acad. Sci. U. S. A. 90, 6914–6918
Fukuda S, Pelus LM. (2006) Survivin, a cancer target with an emerging role in
normal adult tissues. Mol Cancer Ther. May;5(5):1087-98.
Giehl K. (2005) Oncogenic Ras in tumour progression and metastasis. Biol
Chem. Mar;386(3):193-205.
Harbour, J. W. & Dean, D. C. (2000) Chromatin remodeling and Rb activity. Curr.
Opin. Cell Biol. 12, 685-689.
Helin, K., Harlow, E., and Fattaey, A. (1993) Inhibition of E2F-1 transactivation by
direct binding of the retinoblastoma protein. Mol. Cell. Biol. 13, 6501–6508
Hoffman WH, Biade S, Zilfou JT, Chen J, Murphy M. (2002) Transcriptional
repression of the anti-apoptotic survivin gene by wild type p53. J Biol
Chem. Feb 1;277(5):3247-57.
Humbert, P. O. et al. (2000a).E2F4 is essential for normal erythrocyte maturation
and neonatal viability. Mol. Cell 6, 281-291
Humbert, P. O. et al. (2000b). E2f3 is critical for normal cellular proliferation.
Genes Dev. 14, 690-703
Ishida, S. et al. (2001) Role for E2F in control of both DNAreplication and mitotic
functions as revealed from DNA microarray analysis. Mol. Cell. Biol. 21,
4684-4699.
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
Jiang Y, de Bruin A, Caldas H, Fangusaro J, Hayes J, Conway EM, Robinson
ML,Altura RA. (2005) Essential role for survivin in early brain
development.J Neurosci. Jul 27;25(30):6962-70.
Jiang Y, Saavedra HI, Holloway MP, Leone G, Altura RA. (2004) Aberrant
regulation of survivin by the RB/E2F family of proteins. J Biol Chem. Sep
24;279(39):40511-20.
Johnson DG, Degregori J. (2006) Putting the Oncogenic and Tumor Suppressive
Activities of E2F into Context. Curr Mol Med. Nov;6(7):731-8.
131
Johnson, D. G., Schwarz, J. K., Cress, W. D., and Nevins, J. R. (1993)
Expression of transcription factor E2F1 induces quiescent cells to enter S
phase. Nature 365, 349–352
Kanda N, Seno H, Konda Y, et al. (2004) STAT3 is constitutively activated and
supports cell survival in association with survivin expression in gastric
cancer cells. Oncogene;23:4921–9.
Kent, W. J. (2002) BLAT
656–664
The BLAST-Like Alignment Tool. Genome Res. 12,
Kosugi, S., and Ohashi, Y. (2002) E2Ls, E2F-like Repressors of Arabidopsis That
Bind to E2F Sites in a Monomeric Form. J. Biol. Chem. 277, 16553–16558
Kovesdi, I., Reichel, R., and Nevins, J. R. (1987) Role of an Adenovirus E2
Promoter Binding Factor in E1A-Mediated Coordinate Gene Control Proc.
Natl. Acad. Sci. U. S. A. 84, 2180–2184
Land, H., L. F. Parada, R. A. Weinberg. (1983). Tumorigenic conversion of
primary embryo fibroblasts requires at least two cooperating oncogenes.
Nature 304:596
Lees, J. A. et al. (1993) The retinoblastoma protein binds to a family of E2F
transcription factors. Mol. Cell. Biol. 13, 7813-7825.
Lens SM, Medema RH. (2003) The survivin/Aurora B complex: its role in
coordinating tension and attachment. Cell CycleNov-Dec;2(6):507-10.
Lens SM, Vader G, Medema RH. (2006) The case for Survivin as mitotic
regulator. Curr Opin Cell Biol. Dec;18(6):616-22.
Lens SM, Wolthuis RM, Klompmaker R, Kauw J, Agami R, Brummelkamp T,
Kops G, Medema RH. (2003) Survivin is required for a sustained spindle
checkpoint arrest in response to lack of tension. EMBO J. Jun
16;22(12):2934-47
Leone, G. et al. (1998) E2F3 activity is regulated during the cell cycle and is
required for the induction of S phase. Genes Dev. 12, 2120-2130.
Leone, G. et al. (2000) Identification of a novel E2F3 product suggests a
mechanism for determining specificity of repression by Rb proteins. Mol.
Cell. Biol. 20, 3626-3632.
Leone, G. et al. (2001) Myc requires distinct E2F activities to induce S phase and
apoptosis. Mol. Cell 8, 105-113
132
Leone, G., DeGregori, J., Yan, Z., Jakoi, L., Ishida, S., Williams, R. S., and
Nevins, J. R. (1998) E2F3 activity is regulated during the cell cycle and is
required for the induction of S phase Genes Dev. 12, 2120–2130
Leone, G., Nuckolls, F., Ishida, S., Adams, M., Sears, R., Jakoi, L., Miron, A.,
and Nevins, J. R. (2000) Identification of a Novel E2F3 Product Suggests
a Mechanism for Determining Specificity of Repression by Rb Proteins.
Mol. Cell. Biol. 20, 3626–3632
Li F, Brattain MG. (2006) Role of the Survivin gene in pathophysiology. Am J
Pathol. Jul;169(1):1-11.
Li F, Ling X. (2006) Survivin study: an update of "what is the next wave"? J Cell
Physiol. Sep;208(3):476-86.
Li F, Yang J, Ramnath N, Javle MM, Tan D. (2005) Nuclear or cytoplasmic
expression of survivin: what is the significance? Int J Cancer. Apr
20;114(4):509-12.
Lin, W.C., Lin, F.T. and Nevins, J.R. (2001) Selective induction of E2F1 in
response to DNA damage, mediated by ATM-dependent phosphorylation.
Genes Dev., 15, 1833-1844.
Lindeman, G. J. et al. (1998) A specific, nonproliferative role for E2F-5 in choroid
plexus function revealed by gene targeting. Genes Dev. 12, 1092-1098.
Liu T, Brouha B, Grossman D. (2004). Rapid induction of mitochondrial events
and caspase-independent apoptosis in Survivin-targeted melanoma cells.
Oncogene 23(1):39–48.
Logan N, Graham A, Zhao X, Fisher R, Maiti B, Leone G, La Thangue NB. E2F8: an E2F family member with a similar organization of DNA-binding
domains to E2F-7. Oncogene. 2005 Jul 21;24(31):5000-4.
Logan, N., Delavaine, L., Graham, A., Reilly, C., Wilson, J., Brummelkamp,T. R.,
Hijmans, E. M., Bernards, R., and La Thangue, N. B. (2004) E2F-7: a
distinctive E2F family member with an unusual organization of DNAbinding domains. Oncogene 23, 5138–5150
Lukas, C. et al. (1999) Accumulation of cyclin B1 requires E2F and cyclin-Adependent rearrangement of the anaphase-promoting complex. Nature
401, 815-881.
Luo, R. X., Postigo, A. A. & Dean, D. C. (1998) Rb interacts with histone
deacetylase to repress transcription. Cell 92, 463-473.
133
Luo, R. X., Postigo, A. A., and Dean, D. C. (1998) Rb interacts with histone
deacetylase to repress transcription. Cell 92, 463–473
Magnaghi-Jaulin, L. et al. (1998) Retinoblastoma protein represses transcription
by recruiting a histone deacetylase. Nature 391, 601-605.
Maiti B, Li J, de Bruin A, Gordon F, Timmers C, Opavsky R, Patil K, Tuttle J,
Cleghorn W, Leone G.( 2005) Cloning and characterization of mouse
E2F8, a novel mammalian E2F family member capable of blocking cellular
proliferation. J Biol Chem. May 6;280 (18):18211-20.
Mariconti, L., Pellegrini, B., Cantoni, R., Stevens, R., Bergounioux, C., Cella, R.,
and Albani, D. (2002) The E2F Family of Transcription Factors from
Arabidopsis thaliana J. Biol. Chem. 277, 9911–9919
Muller, H. et al. (2001) E2Fs regulate the expression of genes involved in
differentiation, development, proliferation, and apoptosis. Genes Dev. 15,
267-285.
Nevins, J. R. (1998) Toward an Understanding of the Functional Complexity of
the E2F and Retinoblastoma Families. Cell Growth & Differ. 9, 585–593
Nevins, J. R., DeGregori, J., Jakoi, L. & Leone, G. (Academic, San Diego, 1997)
in Methods in Enzymology (ed. Dunphy,W. G.) 678.
Nevins, J. R., DeGregori, J., Jakoi, L., and Leone, G. (1997) E2F3 activity is
regulated during the cell cycle and is required for the induction of S phase.
Methods Enzymol. 283, 205–219
Nguyen HG, Ravid K. (2006) Tetraploidy/aneuploidy and stem cells in cancer
promotion: The role of chromosome passenger proteins. J Cell Physiol.
Jul;208(1):12-22.
Ogawa, H., Ishiguro, K., Gaubatz, S., Livingston, D. M., and Nakatani, Y. (2002)
A Complex with Chromatin Modifiers That Occupies E2F- and MycResponsive Genes in G0 Cells. Science 296, 1132–1136
Okada H, Bakal C, Shahinian A, Elia A, Wakeham A, Suh WK, Duncan GS,
Ciofani M, Rottapel R, Zuniga-Pflucker JC, Mak TW. (2004) Survivin loss
in thymocytes triggers p53-mediated growth arrest and p53-independent
cell death.J Exp Med. Feb 2;199(3):399-410.
Pear, W. S., Nolan, G. P., Scott, M. L., and Baltimore, D. (1993) Production of
High-Titer Helper-Free Retroviruses by Transient Transfection. Proc. Natl.
Acad. Sci. U. S. A. 90, 8392–8396
134
Pear,W. S., Nolan, G. P., Scott, M. L. and Baltimore, D. (1993) Production of
high-titer helper-free retroviruses by transient transfection. Proc. Natl
Acad. Sci. USA 90, 8392-8396.
Powers, J.T., Hong, S., Mayhew, C.N., Rogers, P.M., Knudsen, E.S. and
Johnson, D.G. (2004) E2F1 uses the ATM signaling pathway to induce
p53 and Chk2 phosphorylation and apoptosis.Mol. Cancer Res., 2, 20314.
Rempel, R. E. et al. (2000) Loss of E2F4 activity leads to abnormal development
of multiple cellular lineages. Mol. Cell. 6, 293-306.
Ren, B., Cam, H., Takahashi, Y., Volkert T., Terragni, J., Young, R. A., and
Dynlacht, B. D. (2002) E2F integrates cell cycle progression with DNA
repair, replication, and G2/M checkpoints Genes Dev. 16, 245–256
Rogoff, H.A., Pickering, M.T., Frame, F.M., Debatis, M.E., Sanchez, Y., Jones, S.
and Kowalik, T.F. (2004) Apoptosis associated with deregulated E2F
activity is dependent on E2F1 and Atm/Nbs1/Chk2. Mol. Cell. Biol. , 24,
2968-77.
Royzman, I., Whittaker, A. J. and Orr-Weaver, T. L. (1997). Mutations in
Drosophila DP and E2F distinguish G1-S progression from an associated
transcriptional program. Genes Dev. 11, 1999-2011
Saavedra HI, Maiti B, Timmers C, Altura R, Tokuyama Y, Fukasawa K, Leone G.
(2003) Inactivation of E2F3 results in centrosome amplification. Cancer
Cell. Apr;3(4):333-46.
Schwede, T., Kopp, J., Guex, N., and Peitsch, M. C. (2003) SWISS-MODEL: an
automated protein homology-modeling server Nucleic Acids Res. 31,
3381–3385
Sears, R., Ohtani, K., and Nevins, J. R. (1997) Identification of positively and
negatively acting elements regulating expression of the E2F2 gene in
response to cell growth signals. Mol. Cell. Biol. 17, 5227–5235
Sellers, W. R., Rodgers, J. W., Kaelin, and W. G., Jr. (1995) A potent
transrepression domain in the retinoblastoma protein induces a cell cycle
arrest when bound to E2F sites. Proc. Natl. Acad. Sci. U. S. A. 92, 11544–
11548
Sommer KW, Schamberger CJ, Schmidt GE, Sasgary S, Cerni C. (2003)
Inhibitor of apoptosis protein (IAP) survivin is upregulated by oncogenic cH-Ras. Oncogene. Jul 3;22(27):4266-80
135
Stevaux, O., Dimova, D., Frolov, M. V., Taylor-Harding, B., Morris, E., and
Dyson, N. (2002) Distinct mechanisms of E2F regulation by Drosophila
RBF1 and RBF2. EMBO J. 21, 4927–4937
Stevens, C., Smith, L. and La Thangue, N.B. (2003) Chk2 activates E2F-1 in
response to DNA damage. Nat. Cell Biol., 5, 401-9
Suzuki, A., and Hemmati-Brivanlou, A. (2000) Xenopus Embryonic E2F Is
Required for the Formation of Ventral and Posterior Cell Fates during
Early Embryogenesis. Mol. Cell 5, 217–229
Takahashi, Y., Rayman, J. B., and Dynlacht, B. D. (2000) Analysis of promoter
binding by the E2F and pRB families in vivo: distinct E2F proteins mediate
activation and repression Genes Dev. 14, 804–816
Tatsuka M, Sato S, Kitajima S, Suto S, Kawai H, Miyauchi M, Ogawa I, Maeda
M, Ota T, Takata T. (2005) Overexpression of Aurora-A potentiates
HRAS-mediated oncogenic transformation and is implicated in oral
carcinogenesis. Oncogene. Feb 3;24(6):1122-7.
Timmers C, Sharma N, Opavsky R, Maiti B, Wu L, Wu J, Orringer D, Trikha P,
Saavedra HI, Leone G. (2007) E2f1, E2f2, and E2f3 control E2F target
expression and cellular proliferation via a p53-dependent negative
feedback loop. Mol Cell Biol. Jan;27(1):65-78.
Trimarchi, J. M., and Lees, J. A. (2002) Sibling rivalry in the E2F family. Nat. Rev.
Mol. Cell. Biol. 3, 11–20
Trimarchi, J. M., Fairchild, B., Wen, J., and Lees, J. A. (2001) The E2F6
transcription factor is a component of the mammalian Bmi1-containing
polycomb complex Proc. Natl. Acad. Sci. U. S. A. 98, 1519–1524
Trouche, D., Le Chalony, C., Muchardt, C., Yaniv, M., and Kouzarides, T.(1997)
RB and hbrm cooperate to repress the activation functions of E2F1. Proc.
Natl. Acad. Sci. U. S. A. 94, 11268–11273
Tsai, K. Y. et al. (1998) Mutation of E2f-1 suppresses apoptosis and
inappropriate S phase entry and extends survival of Rb-deficient mouse
embryos. Mol. Cell 2, 293-304.
Tsantoulis PK, Gorgoulis VG. (2005) Involvement of E2F transcription factor
family in cancer. Eur J Cancer.Nov;41(16):2403-14.
Uren AG, Wong L, Pakusch M, Fowler KJ, Burrows FJ, Vaux DL, Choo KH.
(2000) Survivin and the inner centromere protein INCENP show similar
136
cell-cycle localization and gene knockout phenotype. Curr Biol. Nov
2;10(21):1319-28.
Vader G, Kauw JJ, Medema RH, Lens SM. (2006a) Survivin mediates targeting
of the chromosomal passenger complex to the centromere and midbody.
EMBO Rep. Jan;7(1):85-92.
Vader G, Medema RH, Lens SM. (2006b) The chromosomal passenger complex:
guiding Aurora-B through mitosis. J Cell Biol. Jun 19;173(6):833-7.
Vandel, L., Nicolas, E., Vaute, O., Ferreira, R., Ait-Si-Ali, S., and Trouche, D.
(2001) Transcriptional repression by the retinoblastoma protein through
the recruitment of a histone methyltransferase. Mol. Cell. Biol. 21, 6484–
6494
Vong QP, Cao K, Li HY, Iglesias PA, Zheng Y. (2005) Chromosome alignment
and segregation regulated by ubiquitination of survivin. Science. Dec
2;310(5753):1499-504.
Wang, Z.M., Yang, H. and Livingston, D.M. (1998) Endogenous E2F-1 promotes
timely G0 exit of resting mouse embryo fibroblasts. Proc. Natl. Acad. Sci.
USA, 95, 15583-6.
Weinberg, R. A. (1997). The cat and mouse games that genes, viruses, and cells
play. Cell 88:573
Weintraub, S. J. et al. (1995) Mechanism of active transcriptional repression by
the retinoblastoma protein. Nature 375, 812-815.
Weintraub, S. J., Prater, C. A.& Dean,D. C. (1992) Retinoblastoma protein
switches the E2F site from positive to negative element. Nature 358, 259261.
Wheatley SP, McNeish IA. (2005) Survivin: a protein with dual roles in mitosis
and apoptosis. Int Rev Cytol.;247:35-88.
Wu, L., Timmers, C., Maiti, B., Saavedra, H. I., Sang, L., Chong, G. T., Nuckolls,
F., Giangrande, P., Wright, F. A., Field, S. J., Greenberg, M. E., Orkin, S.,
Nevins, J. R., Robinson, M. L., and Leone, G. (2001) The E2F1-3
transcription factors are essential for cellular proliferation. Nature 414,
457–462
Xing Z, Conway EM, Kang C, Winoto A. (2004) Essential role of survivin, an
inhibitor of apoptosis protein, in T cell development, maturation, and
homeostasis. J Exp Med. Jan 5;199(1):69-80.
137
Yamasaki, L. et al. (1996) Tumor induction and tissue atrophy in mice lacking
E2F-1. Cell 85, 537-548.
Yamasaki, L. et al. (1998) Loss of E2F-1 reduces tumorigenesis and extends the
lifespan of Rb1(+/-) mice. Nature Genet. 18, 360-364.
Yang D, Welm A, Bishop JM. (2004) Cell division and cell survival in the absence
of survivin.Proc Natl Acad Sci U S A. Oct 19;101(42):15100-5.
Zangemeister-Wittke U, Simon HU. (2004) An IAP in action: the multiple roles of
survivin in differentiation, immunity and malignancy. Cell Cycle.
Sep;3(9):1121-3.
Zhang T, Otevrel T, Gao Z, Ehrlich SM, Fields JZ, Boman BM. (2001) Evidence
that APC regulates survivin expression: a possible mechanism
contributing to the stem cell origin of colon cancer. Cancer Res ;61:8664–
7.
Zhang, H. S., Gavin, M., Dahiya, A., Postigo, A. A., Ma, D., Luo, R. X., Harbour,
JW, Dean DC. (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. Mar 31;101(1):79-89.
Zhang, H. S., Postigo, A. A. & Dean, D. C. (1999) Active transcriptional
repression by the Rb-E2F complex mediates G1 arrest triggered by
p16INK4a, TGFb, and contact inhibition. Cell 97, 53-61.
Zheng, N., Fraenkel, E., Pabo, C. O., and Pavletich, N. P. (1999) Genes Dev.
13,666–674
Ziebold,U., Reza, T., Caron, A. and Lees, J. A. (2001) E2F3 contributes both the
inappropriate proliferation and to the apoptosis arising in Rb mutant
embryos. Genes Dev. 15, 386-391.
138