E2F1

14,000,000 Leading Edge Experts on the ideXlab platform

Scan Science and Technology

Contact Leading Edge Experts & Companies

Scan Science and Technology

Contact Leading Edge Experts & Companies

The Experts below are selected from a list of 37962219 Experts worldwide ranked by ideXlab platform

Gustavo Leone - One of the best experts on this subject based on the ideXlab platform.

  • division and apoptosis of e2f deficient retinal progenitors
    Nature, 2009
    Co-Authors: Danian Chen, Gustavo Leone, Marek Pacal, Pamela L Wenzel, Paul S Knoepfler, Rod Bremner
    Abstract:

    The activating E2f transcription factors (E2F1, E2f2 and E2f3) induce transcription and are widely viewed as essential positive cell cycle regulators. Indeed, they drive cells out of quiescence, and the 'cancer cell cycle' in Rb1 null cells is E2f-dependent. Absence of activating E2fs in flies or mammalian fibroblasts causes cell cycle arrest, but this block is alleviated by removing repressive E2f or the tumour suppressor p53, respectively. Thus, whether activating E2fs are indispensable for normal division is an area of debate. Activating E2fs are also well known pro-apoptotic factors, providing a defence against oncogenesis, yet E2F1 can limit irradiation-induced apoptosis. In flies this occurs through repression of hid (also called Wrinkled; Smac/Diablo in mammals). However, in mammals the mechanism is unclear because Smac/Diablo is induced, not repressed, by E2F1, and in keratinocytes survival is promoted indirectly through induction of DNA repair targets. Thus, a direct pro-survival function for E2F1-3 and/or its relevance beyond irradiation has not been established. To address E2F1-3 function in normal cells in vivo we focused on the mouse retina, which is a relatively simple central nervous system component that can be manipulated genetically without compromising viability and has provided considerable insight into development and cancer. Here we show that unlike fibroblasts, E2F1-3 null retinal progenitor cells or activated Muller glia can divide. We attribute this effect to functional interchangeability with Mycn. However, loss of activating E2fs caused downregulation of the p53 deacetylase Sirt1, p53 hyperacetylation and elevated apoptosis, establishing a novel E2f-Sirt1-p53 survival axis in vivo. Thus, activating E2fs are not universally required for normal mammalian cell division, but have an unexpected pro-survival role in development.

  • intestinal hyperplasia induced by simian virus 40 large tumor antigen requires e2f2
    Journal of Virology, 2007
    Co-Authors: Teresa M Saenzrobles, Rene Opavsky, Gustavo Leone, Jennifer A Markovics, Jean Leon Chong, Robert H Whitehead, James M Pipas
    Abstract:

    The simian virus 40 large T antigen contributes to neoplastic transformation, in part, by targeting the Rb family of tumor suppressors. There are three known Rb proteins, pRb, p130, and p107, all of which block the cell cycle by preventing the transcription of genes regulated by the E2F family of transcription factors. T antigen interacts directly with Rb proteins and disrupts Rb-E2F complexes both in vitro and in cultured cells. Consequently, T antigen is thought to inhibit transcriptional repression by the Rb family proteins by disrupting their interaction with E2F proteins, thus allowing E2F-dependent transcription and the expression of cellular genes needed for entry into S phase. This model predicts that active E2F-dependent transcription is required for T-antigen-induced transformation. To test this hypothesis, we have examined the status of Rb-E2F complexes in murine enterocytes. Previous studies have shown that T antigen drives enterocytes into S phase, resulting in intestinal hyperplasia, and that the induction of enterocyte proliferation requires T-antigen binding to Rb proteins. In this paper, we show that normal growth-arrested enterocytes contain p130-E2F4 complexes and that T-antigen expression destroys these complexes, most likely by stimulating p130 degradation. Furthermore, unlike their normal counterparts, enterocytes expressing T antigen contain abundant levels of E2F2 and E2F3a. Concomitantly, T-antigen-induced intestinal proliferation is reduced in mice lacking either E2F2 alone or both E2F2 and E2F3a, but not in mice lacking E2F1. These studies support a model in which T antigen eliminates Rb-E2F repressive complexes so that specific activator E2Fs can drive S-phase entry.

  • E2F1 e2f2 and e2f3 control e2f target expression and cellular proliferation via a p53 dependent negative feedback loop
    Molecular and Cellular Biology, 2007
    Co-Authors: Cynthia Timmers, Baidehi Maiti, Rene Opavsky, Harold I Saavedra, Nidhi Sharma, Daniel A Orringer, Prashant Trikha, Gustavo Leone
    Abstract:

    E2F-mediated control of gene expression is believed to have an essential role in the control of cellular proliferation. Using a conditional gene-targeting approach, we show that the targeted disruption of the entire E2F activator subclass composed of E2F1, E2f2, and E2f3 in mouse embryonic fibroblasts leads to the activation of p53 and the induction of p53 target genes, including p21 CIP1 . Consequently, cyclin-dependent kinase activity and retinoblastoma (Rb) phosphorylation are dramatically inhibited, leading to Rb/E2F-mediated repression of E2F target gene expression and a severe block in cellular proliferation. Inactivation of p53 in E2F1-, E2f2-, and E2f3-deficient cells, either by spontaneous mutation or by conditional gene ablation, prevented the induction of p21 CIP1 and many other p53 target genes. As a result, cyclin-dependent kinase activity, Rb phosphorylation, and E2F target gene expression were restored to nearly normal levels, rendering cells responsive to normal growth signals. These findings suggest that a critical function of the E2F1, E2F2, and E2F3 activators is in the control of a p53-dependent axis that indirectly regulates E2F-mediated transcriptional repression and cellular proliferation.

  • control of the p53 p21cip1 axis by E2F1 e2f2 and e2f3 is essential for g1 s progression and cellular transformation
    Journal of Biological Chemistry, 2006
    Co-Authors: Nidhi Sharma, Cynthia Timmers, Harold I Saavedra, Prashant Trikha, Amanda Obery, Gustavo Leone
    Abstract:

    The E2F family of transcription factors is believed to have an essential role in the control of cellular proliferation by regulating the transcription of genes involved in cell cycle progression. Previous work has demonstrated that the targeted inactivation of E2F1, E2f2, and E2f3 results in elevated p21CIP1 protein levels, loss of E2F target gene expression, and cell cycle arrest at G1/S and G2/M, suggesting a strict requirement for these E2Fs in the control of normal cellular proliferation. We now demonstrate that E2F1, E2f2, and E2f3 are also required for oncogene-mediated transformation of mouse embryonic fibroblasts. Analysis of synchronized populations of mouse embryonic fibroblasts revealed that the inactivation of p21CIP1 restores the ability of E2F1-3-deficient cells to enter and transit through G1/S (but not G2/M). In contrast, loss of p53 restored the ability of these cells to progress through both G1/S and mitosis, leading to their continued proliferation. The inactivation of p53 (but not p21CIP1) rendered E2F1-3-deficient cells sensitive to transformation and tumorigenesis. These results suggest that the negative regulation of the p53-p21CIP1 axis by the E2F1-3 factors is critical for cell cycle progression and cellular transformation.

  • cloning and characterization of mouse e2f8 a novel mammalian e2f family member capable of blocking cellular proliferation
    Journal of Biological Chemistry, 2005
    Co-Authors: Baidehi Maiti, Alain De Bruin, Faye Gordon, Cynthia Timmers, Rene Opavsky, Kaustubha Patil, John Tuttle, Whitney M Cleghorn, Gustavo Leone
    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). We 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 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.

Joseph R Nevins - One of the best experts on this subject based on the ideXlab platform.

  • interaction of e2f7 transcription factor with E2F1 and c terminal binding protein ctbp provides a mechanism for e2f7 dependent transcription repression
    Journal of Biological Chemistry, 2013
    Co-Authors: Beiyu Liu, Igor Shats, Steven P Angus, Michael L Gatza, Joseph R Nevins
    Abstract:

    Previous work has identified distinct functions for E2F proteins during a cellular proliferative response including a role for E2F1–3 in the activation of transcription at G1/S and a role for E2F4–8 in repressing the same group of E2F1–3 target genes as cells progress through S phase. We now find that E2F7 and E2F8, which are induced by E2F1–3 at G1/S, can form a heterodimer with E2F1 through interactions involving the DNA-binding domains of the two proteins. In vitro DNA interaction assays demonstrate the formation of an E2F1-E2F7 complex, as well as an E2F7-E2F7 complex on adjacent E2F-binding sites. We also show that E2F7 recruits the co-repressor C-terminal-binding protein (CtBP) and that CtBP2 is essential for E2F7 to repress E2F1 transcription. Taken together, these findings suggest a mechanism for the repression of transcription by E2F7. Background: E2F7 is a transcription factor that controls cell cycle by repressing the expression of G1/S genes in late S phase. Results: E2F7 forms a heterodimer with E2F1, and it recruits the co-repressor CtBP to repress G1/S transcription. Conclusion: E2F7 represses gene transcription by interacting with E2F1 and co-repressor CtBP. Significance: These findings suggest a mechanism for the repression of transcription by E2F7.

  • specificity in the activation and control of transcription factor e2f dependent apoptosis
    Proceedings of the National Academy of Sciences of the United States of America, 2003
    Co-Authors: Timothy C Hallstrom, Joseph R Nevins
    Abstract:

    Previous work has demonstrated a role for the E2F1 gene product in signaling apoptosis, both as a result of the deregulation of the Rb/E2F pathway as well as in response to DNA damage. We now show that the ability of cells to suppress the apoptotic potential of E2F1, as might occur during the course of normal cellular proliferation, requires the action of the Ras–phosphoinositide 3-kinase–Akt signaling pathway. In addition, we also identify a domain within the E2F1 protein, previously termed the marked-box domain, that is essential for the apoptotic activity of E2F1 and that distinguishes the E2F1 protein from E2F3. We also show that the E2F1-marked-box domain is essential for the induction of both p53 and p73 accumulation. Importantly, a role for the marked-box domain in the specificity of E2F1-mediated apoptosis coincides with recent work demonstrating a role for this domain in achieving specificity in the activation of transcription. We conclude that the unique capacity of E2F1 to trigger apoptosis reflects a specificity of transcriptional activation potential, and that this role for E2F1 is regulated through the action of the Akt protein kinase.

  • identification of e box factor tfe3 as a functional partner for the e2f3 transcription factor
    Molecular and Cellular Biology, 2003
    Co-Authors: Paloma H Giangrande, Timothy C Hallstrom, Chainarong Tunyaplin, Kathryn Calame, Joseph R Nevins
    Abstract:

    The ability of the retinoblastoma (Rb) tumor suppressor protein to regulate cell growth is due, at least in part, to its ability to interact with and regulate the E2F family of transcription factors (8, 34). The E2F proteins have been shown to control the expression of a large number of genes involved in DNA replication, cell cycle progression, and cell fate determination. The E2F family is composed of six distinct gene products that form heterodimeric complexes with partners of the DP family. Sequence analysis reveals three distinct subfamilies of E2F genes: the E2F1, E2F2, and E2F3 genes, the E2F4 and E2F5 genes, and the E2F6 gene. This division also coincides with functional distinctions. The E2F1, E2F2, and E2F3 genes are tightly regulated by cell growth and during the cell cycle, whereas E2F4, E2F5 and E2F6 are constitutively expressed. This cell cycle regulation of E2F1, E2F2, and E2F3 transcription is complemented by mechanisms that tightly regulate the accumulation of the proteins. An N-terminal domain unique to E2F1 to E2F3 is responsible for both ubiquitin-mediated degradation of the proteins (29) and targeting by the cyclin A/cdk2 kinase, the latter leading to inhibition of DNA binding capacity (7, 22, 23, 53). The E2F proteins also vary in their role as transcriptional regulatory activities. While E2F1 to E2F3 act as positive regulators of transcription, E2F4 and E2F5 appear to function primarily as transcriptional repressors in concert with Rb family members. E2F6 also appears to function as a transcriptional repressor but in a manner independent of Rb (4, 10, 47, 48). Various experiments have suggested distinct functional roles for the activating E2F proteins E2F1, E2F2, and E2F3. The E2F3 protein appears to be particularly important for cell proliferation, as seen from the inhibition of E2F3 activity by antibody microinjection (25) as well as the results of deletion of the E2F3 gene (18). Moreover, the expression of a number of E2F target genes that encode key cell cycle regulatory proteins, including B-Myb, cyclin A, cdc2, cdc6, and dihydrofolate reductase, are reduced in E2F3 null fibroblasts but not in E2F1 null cells (18). In contrast, E2F1 appears to play a role in triggering an apoptotic response, either when overexpressed in the absence of survival signals (6, 21, 39, 43, 51) or in response to DNA damage (27). In addition, the ability of Myc to induce apoptosis is impaired in the absence of E2F1 function but unaltered by the absence of either E2F2 or E2F3 (26). Given the role of the E2F1, E2F2, and E2F3 proteins as transcriptional activators, the specificity of function might best be explained by an ability of these E2F proteins to activate distinct target genes that then carry out these functions of apoptosis or proliferation. As an example, the p19ARF gene has been shown to be an E2F1 target gene (2, 6), linking the action of the Rb/E2F pathway with the p53 response leading to apoptosis. Similarly, the Apaf1 gene appears to be activated specifically by E2F1 (30). Although one potential mechanism for such specificity could be an ability of these proteins to recognize subtle differences in cis-acting promoter sequences, there is little evidence for distinct DNA sequence recognition among the E2F isoforms. More importantly, analysis of the structure of an E2F-DNA complex did not show a capacity for distinct DNA sequence recognition when the amino acid variation within the E2F family is considered (56). An alternative mechanism for promoter specificity could involve distinct protein-protein interactions. Possibly, sequences within E2F3 allow interaction with a subset of cellular proteins that provide a basis for promoter specificity and that are distinct from the proteins that can interact with E2F1. For example, the interaction of the herpesvirus VP16 transcriptional activator protein with the cellular factor HCF-1 and the cellular Oct-1 transcription factor redirects Oct-1 to herpesvirus immediate-early promoters by virtue of expanded DNA sequence recognition (1). A further example of combinatorial specificity as a mechanism for the specificity of transcription factor function is the pancreatic islet factor STF-1, which interacts with Pbx in a cooperative fashion and targets Pbx to a subset of promoters containing STF-1 binding sites (38). Recently, we identified the YY1-binding protein RYBP as a factor which binds to E2F2 and E2F3 but not E2F1 and recruits these E2Fs to a subset of E2F target promoters containing YY1 binding sites (41). To further explore the mechanistic basis for the specificity of E2F transcription activation, we used yeast two-hybrid screens to identify proteins that specifically interact with E2F3. In so doing, we identified the TFE3 transcription factor as a specific partner for E2F3. Recent experiments identified TFE3 as an activity that can rescue Rb-mediated growth arrest, providing a functional link between TFE3 and the Rb/E2F pathway (M. Nijman, S. Hijmans, and R. Bernards, personal communication). Moreover, previous work identified TFE3 as a fusion partner in chromosomal rearrangements in renal cell carcinomas (14, 45, 50). We further show that TFE3 and E2F3 can act synergistically to activate the p68 subunit gene of DNA polymerase α, dependent on the ability of the two proteins to physically interact, and that the two activities associate with the p68 promoter within intact cells in a mutually dependent manner.

  • interaction of yy1 with e2fs mediated by rybp provides a mechanism for specificity of e2f function
    The EMBO Journal, 2002
    Co-Authors: Susanne Schlisio, Joseph R Nevins, Terri J Halperin, Miguel Vidal
    Abstract:

    To explore mechanisms for specificity of function within the family of E2F transcription factors, we have identified proteins that interact with individual E2F proteins. A two-hybrid screen identified RYBP (Ring1- and YY1-binding protein) as a protein that interacts specifically with the E2F2 and E2F3 family members, dependent on the marked box domain in these proteins. The Cdc6 promoter contains adjacent E2F- and YY1-binding sites, and both are required for promoter activity. In addition, YY1 and RYBP, in combination with either E2F2 or E2F3, can stimulate Cdc6 promoter activity synergistically, dependent on the marked box domain of E2F3. Using chromatin immunoprecipitation assays, we show that both E2F2 and E2F3, as well as YY1 and RYBP, associate with the Cdc6 promoter at G1/S of the cell cycle. In contrast, we detect no interaction of E2F1 with the Cdc6 promoter. We suggest that the ability of RYBP to mediate an interaction between E2F2 or E2F3 and YY1 is an important component of Cdc6 activation and provides a basis for specificity of E2F function.

  • identification of a novel e2f3 product suggests a mechanism for determining specificity of repression by rb proteins
    Molecular and Cellular Biology, 2000
    Co-Authors: Gustavo Leone, Rosalie C Sears, Faison Nuckolls, Alexander Miron, Laszlo Jakoi, Seiichi Ishida, Monique R Adams, Joseph R Nevins
    Abstract:

    The tumor suppressor function of Rb is intimately related to its ability to interact with E2F and repress the transcription of E2F target genes. Here we describe a novel E2F product that specifically interacts with Rb in quiescent cells. This novel E2F, which we term E2F3b, is encoded by a unique mRNA transcribed from an intronic promoter within the E2F3 locus. The E2F3b RNA differs from the previously characterized E2F3 RNA, which we now term E2F3a, by the utilization of a unique coding exon. In contrast to the E2F3a product that is tightly regulated by cell growth, the E2F3b product is expressed equivalently in quiescent and proliferating cells. But, unlike the E2F4 and E2F5 proteins, which are also expressed in quiescent cells and form complexes with the p130 protein, the E2F3b protein associates with Rb and represents the predominant E2F-Rb complex in quiescent cells. Thus, the previously described specificity of Rb function as a transcriptional repressor in quiescent cells coincides with the association of Rb with this novel E2F product.

Alain De Bruin - One of the best experts on this subject based on the ideXlab platform.

  • cyclin f dependent degradation of e2f7 is critical for dna repair and g2 phase progression
    The EMBO Journal, 2019
    Co-Authors: Ruixue Yuan, Alain De Bruin, Bart Westendorp, Qingwu Liu, Hendrika A Segeren, Laurensia Yuniati, Daniele Guardavaccaro, Robert Jan Lebbink
    Abstract:

    E2F7 and E2F8 act as tumor suppressors via transcriptional repression of genes involved in S-phase entry and progression. Previously, we demonstrated that these atypical E2Fs are degraded by APC/CCdh1 during G1 phase of the cell cycle. However, the mechanism driving the downregulation of atypical E2Fs during G2 phase is unknown. Here, we show that E2F7 is targeted for degradation by the E3 ubiquitin ligase SCFcyclin F during G2. Cyclin F binds via its cyclin domain to a conserved C-terminal CY motif on E2F7. An E2F7 mutant unable to interact with SCFcyclin F remains stable during G2. Furthermore, SCFcyclin F can also interact and induce degradation of E2F8. However, this does not require the cyclin domain of SCFcyclin F nor the CY motifs in the C-terminus of E2F8, implying a different regulatory mechanism than for E2F7. Importantly, depletion of cyclin F causes an atypical-E2F-dependent delay of the G2/M transition, accompanied by reduced expression of E2F target genes involved in DNA repair. Live cell imaging of DNA damage revealed that cyclin F-dependent regulation of atypical E2Fs is critical for efficient DNA repair and cell cycle progression.

  • chk1 and 14 3 3 proteins inhibit atypical e2fs to prevent a permanent cell cycle arrest
    The EMBO Journal, 2018
    Co-Authors: Ruixue Yuan, Alain De Bruin, Harmjan R. Vos, Robert M. Van Es, Jing Chen, Boudewijn M.t. Burgering, Bart Westendorp
    Abstract:

    The atypical E2Fs, E2F7 and E2F8, act as potent transcriptional repressors of DNA replication genes providing them with the ability to induce a permanent S-phase arrest and suppress tumorigenesis. Surprisingly in human cancer, transcript levels of atypical E2Fs are frequently elevated in proliferating cancer cells, suggesting that the tumor suppressor functions of atypical E2Fs might be inhibited through unknown post-translational mechanisms. Here, we show that atypical E2Fs can be directly phosphorylated by checkpoint kinase 1 (Chk1) to prevent a permanent cell cycle arrest. We found that 14-3-3 protein isoforms interact with both E2Fs in a Chk1-dependent manner. Strikingly, Chk1 phosphorylation and 14-3-3-binding did not relocate or degrade atypical E2Fs, but instead, 14-3-3 is recruited to E2F7/8 target gene promoters to possibly interfere with transcription. We observed that high levels of 14-3-3 strongly correlate with upregulated transcription of atypical E2F target genes in human cancer. Thus, we reveal that Chk1 and 14-3-3 proteins cooperate to inactivate the transcriptional repressor functions of atypical E2Fs. This mechanism might be of particular importance to cancer cells, since they are exposed frequently to DNA-damaging therapeutic reagents.

  • atypical e2fs control lymphangiogenesis through transcriptional regulation of ccbe1 and flt4
    PLOS ONE, 2013
    Co-Authors: Bart Weijts, Andreas Van Impel, Stefan Schultemerker, Alain De Bruin
    Abstract:

    Lymphatic vessels are derived from venous endothelial cells and their formation is governed by the Vascular endothelial growth factor C (VegfC)/Vegf receptor 3 (Vegfr3; Flt4) signaling pathway. Recent studies show that Collagen and Calcium Binding EGF domains 1 protein (Ccbe1) enhances VegfC-dependent lymphangiogenesis. Both Ccbe1 and Flt4 have been shown to be indispensable for lymphangiogenesis. However, how these essential players are transcriptionally regulated remains poorly understood. In the case of angiogenesis, atypical E2fs (E2f7 and E2f8) however have been recently shown to function as transcriptional activators for VegfA. Using a genome-wide approach we here identified both CCBE1 and FLT4 as direct targets of atypical E2Fs. E2F7/8 directly bind and stimulate the CCBE1 promoter, while recruitment of E2F7/8 inhibits the FLT4 promoter. Importantly, inactivation of e2f7/8 in zebrafish impaired venous sprouting and lymphangiogenesis with reduced ccbe1 expression and increased flt4 expression. Remarkably, over-expression of e2f7/8 rescued Ccbe1- and Flt4-dependent lymphangiogenesis phenotypes. Together these results identified E2f7/8 as novel in vivo transcriptional regulators of Ccbe1 and Flt4, both essential genes for venous sprouting and lymphangiogenesis.

  • cloning and characterization of mouse e2f8 a novel mammalian e2f family member capable of blocking cellular proliferation
    Journal of Biological Chemistry, 2005
    Co-Authors: Baidehi Maiti, Alain De Bruin, Faye Gordon, Cynthia Timmers, Rene Opavsky, Kaustubha Patil, John Tuttle, Whitney M Cleghorn, Gustavo Leone
    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). We 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 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.

  • specificity of E2F1 e2f2 and e2f3 in mediating phenotypes induced by loss of rb
    Cell Growth & Differentiation, 2002
    Co-Authors: Harold I Saavedra, Alain De Bruin, Cynthia Timmers, Thomas J Rosol, Michael Weinstein, Michael L Robinson, Gustavo Leone
    Abstract:

    The Rb/E2F pathway plays a critical role in the control of cellular proliferation. Here, we report that E2F1, E2F2, and E2F3 make major individual contributions toward the in vivo phenotypic consequences of Rb deficiency. In the developing lens of Rb / embryos, loss of E2F1, E2F2, or E2F3 reduces the unscheduled proliferation of fiber cells, with the loss of E2F3 having the most pronounced effect. In Rb-deficient retinas, all three E2Fs contribute equally to the ectopic proliferation of postmitotic neuronal cells. In contrast, E2F1 is unique in mediating apoptosis in both Rb / lenses and retinas. In the central nervous system, loss of E2F1 or E2F3 can almost completely eliminate the ectopic DNA replication and apoptosis observed in Rb / embryos, and loss of E2F2 partially reduces the unscheduled DNA replication and has no effect on apoptosis. These results provide clear evidence for functional specificity among E2Fs in the control of Rbdependent proliferation and apoptosis in a tissuespecific manner.

James Degregori - One of the best experts on this subject based on the ideXlab platform.

  • distinct and overlapping roles for e2f family members in transcription proliferation and apoptosis
    Current Molecular Medicine, 2006
    Co-Authors: James Degregori, David G Johnson
    Abstract:

    Since the discovery almost fifteen years ago that E2F transcription factors are key targets of the retinoblastoma protein (RB), studies of the E2F family have uncovered critical roles in the control of transcription , cell cycle and apoptosis. E2F proteins are encoded by at least eight genes, E2F1 through E2F8. While specific roles for individual E2Fs in mediating the effects of RB loss are emerging, it is also becoming clear that there are no simple divisions of labor among the E2F family. Instead, an individual E2F can function to activate or repress transcription, promote or impede cell cycle progression and enhance or inhibit cell death, dependent on the cellular context. While functional redundancy among E2Fs and the striking influences of cellular context on the effects of E2F loss or gain of function have prevented a simple delineation of unique functions within the E2F family, these complexities undoubtedly reflect the extensive regulation and importance of this transcription factor family.

  • E2F1 and e2f2 are differentially required for homeostasis driven and antigen induced t cell proliferation in vivo
    Journal of Immunology, 2005
    Co-Authors: Deborah Deryckere, James Degregori
    Abstract:

    Homeostasis-driven T cell proliferation occurs in response to a lymphopenic environment and is mediated by TCR and IL-7 signaling. In this report, we demonstrate a defect in the proliferation of murine naive and memory T cells lacking both E2F1 and E2F2 in response to lymphopenic conditions, suggesting that E2F1 and E2F2 function redundantly downstream of TCR and/or IL-7 signaling during homeostasis-driven proliferation. In contrast, T cell proliferation in response to antigenic stimulation is either unaffected (in vivo) or potentiated (ex vivo) by loss of E2F1 and E2F2, indicating divergent requirements for these E2F factors in T cell proliferation mediated by distinct stimuli. E2F1/E2F2 double knockout (DKO) T cells enter S phase in response to homeostatic signaling, but fail to divide, suggesting that S phase progression is either incomplete or defective. In addition, E2F1/E2F2 DKO mice do not recover normal T cell numbers following exposure to a sublethal dose of radiation, indicating that this defect in homeostasis-driven proliferation is physiologically relevant. Consistent with their failure in cell cycle progression, the differentiation of DKO T cells into memory T cells in response to homeostatic signals is significantly reduced. These observations support the idea that proliferation is required for memory T cell formation and also have implications for the development of clinical strategies to minimize the occurrence of lymphopenia-induced autoimmunity.

  • the development of diabetes in E2F1 e2f2 mutant mice reveals important roles for bone marrow derived cells in preventing islet cell loss
    Proceedings of the National Academy of Sciences of the United States of America, 2003
    Co-Authors: Jing W Zhu, Christopher J Hogan, Jeffery S Tessem, Joshua Beilke, Marileila Varellagarcia, Jan Jensen, James Degregori
    Abstract:

    Our studies of mice deficient for the E2F1 and E2F2 transcription factors have revealed essential roles for these proteins in the cell cycle control of pancreatic exocrine cells and the regulation of pancreatic beta cell maintenance. Pancreatic exocrine cells in E2F1/E2f2 mutant mice become increasingly polyploid with age, coinciding with severe exocrine atrophy. Furthermore, mice deficient for both E2F1 and E2F2 develop nonautoimmune, insulin-dependent diabetes with high penetrance. Surprisingly, transplantation of wild-type bone marrow can prevent or rescue diabetes in E2F1-/-E2f2-/- mice. We hypothesize that exocrine degeneration results in a destructive environment for beta cells, which can be alleviated by restoration of the hematopoietic system that is also defective in E2F1-/-E2f2-/- mice. The demonstration that beta cell maintenance under conditions of stress is influenced by bone marrow-derived cells may provide important insight into the design of therapies to boost islet mass and function in diabetic patients.

  • defective gene expression s phase progression and maturation during hematopoiesis in E2F1 e2f2 mutant mice
    Molecular and Cellular Biology, 2003
    Co-Authors: Jing W Zhu, Christopher J Hogan, James Degregori
    Abstract:

    E2F plays critical roles in cell cycle progression by regulating the expression of genes involved in nucleotide synthesis, DNA replication, and cell cycle control. We show that the combined loss of E2F1 and E2F2 in mice leads to profound cell-autonomous defects in the hematopoietic development of multiple cell lineages. E2F2 mutant mice show erythroid maturation defects that are comparable with those observed in patients with megaloblastic anemia. Importantly, hematopoietic defects observed in E2F1/E2F2 double-knockout (DKO) mice appear to result from impeded S phase progression in hematopoietic progenitor cells. During DKO B-cell maturation, differentiation beyond the large pre-BII-cell stage is defective, presumably due to failed cell cycle exit, and the cells undergo apoptosis. However, apoptosis appears to be the consequence of failed maturation, not the cause. Despite the accumulation of hematopoietic progenitor cells in S phase, the combined loss of E2F1 and E2F2 results in significantly decreased expression and activities of several E2F target genes including cyclin A2. Our results indicate specific roles for E2F1 and E2F2 in the induction of E2F target genes, which contribute to efficient expansion and maturation of hematopoietic progenitor cells. Thus, E2F1 and E2F2 play essential and redundant roles in the proper coordination of cell cycle progression with differentiation which is necessary for efficient hematopoiesis.

  • distinct roles for e2f proteins in cell growth control and apoptosis
    Proceedings of the National Academy of Sciences of the United States of America, 1997
    Co-Authors: James Degregori, Gustavo Leone, Alexander Miron, Laszlo Jakoi, Joseph R Nevins
    Abstract:

    E2F transcription activity is composed of a family of heterodimers encoded by distinct genes. Through the overproduction of each of the five known E2F proteins in mammalian cells, we demonstrate that a large number of genes encoding proteins important for cell cycle regulation and DNA replication can be activated by the E2F proteins and that there are distinct specificities in the activation of these genes by individual E2F family members. Coexpression of each E2F protein with the DP1 heterodimeric partner does not significantly alter this specificity. We also find that only E2F1 overexpression induces cells to undergo apoptosis, despite the fact that at least two other E2F family members, E2F2 and E2F3, are equally capable of inducing S phase. The ability of E2F1 to induce apoptosis appears to result from the specific induction of an apoptosis-promoting activity rather than the lack of induction of a survival activity, because co-expression of E2F2 and E2F3 does not rescue cells from E2F1-mediated apoptosis. We conclude that E2F family members play distinct roles in cell cycle control and that E2F1 may function as a specific signal for the initiation of an apoptosis pathway that must normally be blocked for a productive proliferation event.

Cynthia Timmers - One of the best experts on this subject based on the ideXlab platform.

  • E2F1 e2f2 and e2f3 control e2f target expression and cellular proliferation via a p53 dependent negative feedback loop
    Molecular and Cellular Biology, 2007
    Co-Authors: Cynthia Timmers, Baidehi Maiti, Rene Opavsky, Harold I Saavedra, Nidhi Sharma, Daniel A Orringer, Prashant Trikha, Gustavo Leone
    Abstract:

    E2F-mediated control of gene expression is believed to have an essential role in the control of cellular proliferation. Using a conditional gene-targeting approach, we show that the targeted disruption of the entire E2F activator subclass composed of E2F1, E2f2, and E2f3 in mouse embryonic fibroblasts leads to the activation of p53 and the induction of p53 target genes, including p21 CIP1 . Consequently, cyclin-dependent kinase activity and retinoblastoma (Rb) phosphorylation are dramatically inhibited, leading to Rb/E2F-mediated repression of E2F target gene expression and a severe block in cellular proliferation. Inactivation of p53 in E2F1-, E2f2-, and E2f3-deficient cells, either by spontaneous mutation or by conditional gene ablation, prevented the induction of p21 CIP1 and many other p53 target genes. As a result, cyclin-dependent kinase activity, Rb phosphorylation, and E2F target gene expression were restored to nearly normal levels, rendering cells responsive to normal growth signals. These findings suggest that a critical function of the E2F1, E2F2, and E2F3 activators is in the control of a p53-dependent axis that indirectly regulates E2F-mediated transcriptional repression and cellular proliferation.

  • control of the p53 p21cip1 axis by E2F1 e2f2 and e2f3 is essential for g1 s progression and cellular transformation
    Journal of Biological Chemistry, 2006
    Co-Authors: Nidhi Sharma, Cynthia Timmers, Harold I Saavedra, Prashant Trikha, Amanda Obery, Gustavo Leone
    Abstract:

    The E2F family of transcription factors is believed to have an essential role in the control of cellular proliferation by regulating the transcription of genes involved in cell cycle progression. Previous work has demonstrated that the targeted inactivation of E2F1, E2f2, and E2f3 results in elevated p21CIP1 protein levels, loss of E2F target gene expression, and cell cycle arrest at G1/S and G2/M, suggesting a strict requirement for these E2Fs in the control of normal cellular proliferation. We now demonstrate that E2F1, E2f2, and E2f3 are also required for oncogene-mediated transformation of mouse embryonic fibroblasts. Analysis of synchronized populations of mouse embryonic fibroblasts revealed that the inactivation of p21CIP1 restores the ability of E2F1-3-deficient cells to enter and transit through G1/S (but not G2/M). In contrast, loss of p53 restored the ability of these cells to progress through both G1/S and mitosis, leading to their continued proliferation. The inactivation of p53 (but not p21CIP1) rendered E2F1-3-deficient cells sensitive to transformation and tumorigenesis. These results suggest that the negative regulation of the p53-p21CIP1 axis by the E2F1-3 factors is critical for cell cycle progression and cellular transformation.

  • cloning and characterization of mouse e2f8 a novel mammalian e2f family member capable of blocking cellular proliferation
    Journal of Biological Chemistry, 2005
    Co-Authors: Baidehi Maiti, Alain De Bruin, Faye Gordon, Cynthia Timmers, Rene Opavsky, Kaustubha Patil, John Tuttle, Whitney M Cleghorn, Gustavo Leone
    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). We 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 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.

  • specificity of E2F1 e2f2 and e2f3 in mediating phenotypes induced by loss of rb
    Cell Growth & Differentiation, 2002
    Co-Authors: Harold I Saavedra, Alain De Bruin, Cynthia Timmers, Thomas J Rosol, Michael Weinstein, Michael L Robinson, Gustavo Leone
    Abstract:

    The Rb/E2F pathway plays a critical role in the control of cellular proliferation. Here, we report that E2F1, E2F2, and E2F3 make major individual contributions toward the in vivo phenotypic consequences of Rb deficiency. In the developing lens of Rb / embryos, loss of E2F1, E2F2, or E2F3 reduces the unscheduled proliferation of fiber cells, with the loss of E2F3 having the most pronounced effect. In Rb-deficient retinas, all three E2Fs contribute equally to the ectopic proliferation of postmitotic neuronal cells. In contrast, E2F1 is unique in mediating apoptosis in both Rb / lenses and retinas. In the central nervous system, loss of E2F1 or E2F3 can almost completely eliminate the ectopic DNA replication and apoptosis observed in Rb / embryos, and loss of E2F2 partially reduces the unscheduled DNA replication and has no effect on apoptosis. These results provide clear evidence for functional specificity among E2Fs in the control of Rbdependent proliferation and apoptosis in a tissuespecific manner.

  • the E2F1 3 transcription factors are essential for cellular proliferation
    Nature, 2001
    Co-Authors: Cynthia Timmers, Baidehi Maiti, Faison Nuckolls, Paloma H Giangrande, Harold I Saavedra, Seth J Field, Ling Sang, Gabriel T Chong, Fred A Wright, Michael E Greenberg
    Abstract:

    The retinoblastoma tumour suppressor (Rb) pathway is believed to have a critical role in the control of cellular proliferation by regulating E2F activities1,2. E2F1, E2F2 and E2F3 belong to a subclass of E2F factors thought to act as transcriptional activators important for progression through the G1/S transition3. 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 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.