Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Sci Rep ; 10(1): 17948, 2020 10 21.
Article in English | MEDLINE | ID: mdl-33087787

ABSTRACT

The E2F family, classically known for a central role in cell cycle, has a number of emerging roles in cancer including angiogenesis, metabolic reprogramming, metastasis and DNA repair. E2F1 specifically has been shown to be a critical mediator of DNA repair; however, little is known about DNA repair and other E2F family members. Here we present an integrative bioinformatic and high throughput drug screening study to define the role of E2F2 in maintaining genomic integrity in breast cancer. We utilized in vitro E2F2 ChIP-chip and over expression data to identify transcriptional targets of E2F2. This data was integrated with gene expression from E2F2 knockout tumors in an MMTV-Neu background. Finally, this data was compared to human datasets to identify conserved roles of E2F2 in human breast cancer through the TCGA breast cancer, Cancer Cell Line Encyclopedia, and CancerRx datasets. Through these methods we predict that E2F2 transcriptionally regulates mediators of DNA repair. Our gene expression data supports this hypothesis and low E2F2 activity is associated with a highly unstable tumor. In human breast cancer E2F2, status was also correlated with a patient's response to PARP inhibition therapy. Taken together this manuscript defines a novel role of E2F2 in cancer progression beyond cell cycle and could impact patient treatment.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , E2F2 Transcription Factor/physiology , Genomic Instability/genetics , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Animals , Breast Neoplasms/drug therapy , Cell Cycle/genetics , Cell Line, Tumor , DNA Repair/genetics , Disease Models, Animal , Disease Progression , Drug Resistance, Neoplasm , E2F2 Transcription Factor/genetics , E2F2 Transcription Factor/metabolism , Female , Gene Expression , Humans , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy , Neovascularization, Pathologic/genetics
2.
Sci Rep ; 9(1): 18830, 2019 12 11.
Article in English | MEDLINE | ID: mdl-31827131

ABSTRACT

Adult mammalian cardiomyocytes exit the cell cycle during the neonatal period, commensurate with the loss of regenerative capacity in adult mammalian hearts. We established conditions for long-term culture of adult mouse cardiomyocytes that are genetically labeled with fluorescence. This technique permits reliable analyses of proliferation of pre-existing cardiomyocytes without complications from cardiomyocyte marker expression loss due to dedifferentiation or significant contribution from cardiac progenitor cell expansion and differentiation in culture. Using this system, we took a candidate gene approach to screen for fetal-specific proliferative gene programs that can induce proliferation of adult mouse cardiomyocytes. Using pooled gene delivery and subtractive gene elimination, we identified a novel functional interaction between E2f Transcription Factor 2 (E2f2) and Brain Expressed X-Linked (Bex)/Transcription elongation factor A-like (Tceal) superfamily members Bex1 and Tceal8. Specifically, Bex1 and Tceal8 both preserved cell viability during E2f2-induced cell cycle re-entry. Although Tceal8 inhibited E2f2-induced S-phase re-entry, Bex1 facilitated DNA synthesis while inhibiting cell death. In sum, our study provides a valuable method for adult cardiomyocyte proliferation research and suggests that Bex family proteins may function in modulating cell proliferation and death decisions during cardiomyocyte development and maturation.


Subject(s)
Cell Dedifferentiation , E2F2 Transcription Factor/metabolism , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/metabolism , Animals , Cell Cycle , Cell Proliferation , DNA Replication , E2F2 Transcription Factor/physiology , Mice , Myocytes, Cardiac/physiology , Nerve Tissue Proteins/physiology , Signal Transduction
3.
Sci Rep ; 8(1): 2623, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29422529

ABSTRACT

As a transcription factor, E2F2 participates in regulation of numerous genes. To investigate the role and mechnism of E2F2 in RA, expression of E2F2 in synovial tissue was detected. Proliferation, invasion, and secretion of inflammatory cytokines were measured after E2F2 was knocked-down in RASFs by siRNA transfection. Induction of TNF-α, IL-6, and LPS on expression and nuclear translocation of E2F2, and signal pathways involved in the process were tested. ChIP was used to investigate direct binding of NF-кB to the promoter of E2F2, and E2F2 to the promoter of IL-6. The correlation between mRNA levels of E2F2 and IL-6 or TNF-α in secreted in supernatant of RASFs were also investigated. As a result, silencing E2F2 could inhibit the proliferation and invasion of RASFs. LPS, IL-6 can stimulate the expression of E2F2 in RASFs both via the NF-кB pathway, while TNF-α via the ERK pathway. TNF-α can facilitate the nuclear translocation of E2F2 and TNF-α can bind to promoter of E2F2, and then E2F2 can bind to the promoter of IL-6 directly. Significant correlations was found between levels of E2F2 and IL-6/TNF-α in synoviocytes of RA patients. Our findings indicate that E2F2 may play an important role in pathogenesis of RA.


Subject(s)
Arthritis, Rheumatoid/pathology , E2F2 Transcription Factor/physiology , Interleukin-6/metabolism , Osteoarthritis/pathology , Synovial Membrane/pathology , Synoviocytes/pathology , Tumor Necrosis Factor-alpha/metabolism , Adult , Aged , Cell Movement , Cell Proliferation , Cells, Cultured , E2F2 Transcription Factor/genetics , Female , Humans , MAP Kinase Signaling System , Male , Middle Aged , NF-kappa B/metabolism
4.
Eur Rev Med Pharmacol Sci ; 21(9): 2150-2156, 2017 05.
Article in English | MEDLINE | ID: mdl-28537669

ABSTRACT

OBJECTIVE: To explore the genes co-upregulated with E2F2 in ovarian cancer and their association with survival outcomes in ovarian cancer patients. MATERIALS AND METHODS: The raw data of GDS3592 was downloaded from GEO datasets for reanalysis. The overlapping subset between the top 150 upregulated genes in ovarian cancer epithelial cells (CEPIs) and the E2F2 positively correlated genes (Pearson's r≥0.5) in ovarian cancer cohort in TCGA was identified. The association between E2F2, MCM4, CCNE2 and WHSC1 and overall survival (OS) and recurrence-free survival (RFS) in ovarian cancer patients were assessed using Kaplan-Meier plotter. RESULTS: E2F2 is a significantly upregulated transcription factor in CEPIs. MCM4, CCNE2, and WHSC1 are co-upregulated with E2F2 among the 308 ovarian cancer samples (Pearson's r=0.5159, 0.3963 and 0.4941 respectively). Enforced E2F2 expression significantly enhanced MCM4, CCNE2 and WHSC1 transcription in SKOV3 and A2780 cells. High E2F2 and CCNE2 expression are associated with worse OS (high E2F2, HR: 1.48, 95%CI: 1.17-1.85, p<0.01; high CCNE2, HR: 1.36, 95%CI: 1.15-1.6, p<0.01). High MCM expression might be associated with worse RFS at the margin of significance (HR: 1.18, 95%CI: 1.00-1.39, p=0.055). CONCLUSIONS: MCM4, CCNE2, and WHSC1 are co-upregulated with E2F2 in ovarian cancer. Enforced E2F2 expression significantly increased MCM4, CCNE2, and WHSC1 expression in ovarian cancer cells. High E2F2 and CCNE2 expression are associated with worse OS among ovarian cancer patients.


Subject(s)
Cyclins/genetics , E2F2 Transcription Factor/physiology , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/genetics , Minichromosome Maintenance Complex Component 4/genetics , Ovarian Neoplasms/mortality , Repressor Proteins/genetics , Cell Line, Tumor , Female , Humans , Ovarian Neoplasms/metabolism , Up-Regulation
5.
Cell Cycle ; 14(23): 3698-712, 2015.
Article in English | MEDLINE | ID: mdl-26505089

ABSTRACT

Traumatic spinal cord injury (SCI) induces cell cycle activation (CCA) that contributes to secondary injury and related functional impairments such as motor deficits and hyperpathia. E2F1 and E2F2 are members of the activator sub-family of E2F transcription factors that play an important role in proliferating cells and in cell cycle-related neuronal death, but no comprehensive study have been performed in SCI to determine the relative importance of these factors. Here we examined the temporal distribution and cell-type specificity of E2F1 and E2F2 expression following mouse SCI, as well as the effects of genetic deletion of E2F1-2 on neuronal cell death, neuroinflammation and associated neurological dysfunction. SCI significantly increased E2F1 and E2F2 expression in active caspase-3(+) neurons/oligodendrocytes as well as in activated microglia/astrocytes. Injury-induced up-regulation of cell cycle-related genes and protein was significantly reduced by intrathecal injection of high specificity E2F decoy oligodeoxynucleotides against the E2F-binding site or in E2F1-2 null mice. Combined E2F1+2 siRNA treatment show greater neuroprotection in vivo than E2F1 or E2F2 single siRNA treatment. Knockout of both E2F1 and E2F2 genes (E2Fdko) significantly reduced neuronal death, neuroinflammation, and tissue damage, as well as limiting motor dysfunction and hyperpathia after SCI. Both CCA reduction and functional improvement in E2Fdko mice were greater than those in E2F2ko model. These studies demonstrate that SCI-induced activation of E2F1-2 mediates CCA, contributing to gliopathy and neuronal/tissue loss associated with motor impairments and post-traumatic hyperesthesia. Thus, E2F1-2 provide a therapeutic target for decreasing secondary tissue damage and promoting recovery of function after SCI.


Subject(s)
E2F1 Transcription Factor/physiology , E2F2 Transcription Factor/physiology , Spinal Cord Injuries/pathology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Cell Death , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , E2F2 Transcription Factor/genetics , E2F2 Transcription Factor/metabolism , Gene Expression , Gene Knockout Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord Injuries/genetics , Spinal Cord Injuries/metabolism
6.
Cell Death Differ ; 22(10): 1577-89, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25656653

ABSTRACT

Tissue homeostasis requires tight regulation of cellular proliferation, differentiation and apoptosis. E2F1 and E2F2 transcription factors share a critical role in tissue homeostasis, since their combined inactivation results in overall organ involution, specially affecting the pancreatic gland, which subsequently triggers diabetes. We have examined the mechanism by which these E2Fs regulate tissue homeostasis. We show that pancreas atrophy in E2F1/E2F2 double-knockout (DKO) mice is associated with mitochondrial apoptosis and activation of the p53 pathway in young animals, before the development of diabetes. A deregulated expression of E2F target genes was detected in pancreatic cells of young DKO animals, along with unscheduled DNA replication and activation of a DNA damage response. Importantly, suppression of DNA replication in vivo with aphidicolin led to a significant inhibition of the p53 pathway in DKO pancreas, implying a causal link between DNA replication stress and p53 activation in this model. We further show that activation of the p53 pathway has a key role in the aberrant phenotype of DKO mice, since targeted inactivation of p53 gene abrogated cellular apoptosis and prevented organ involution and insulin-dependent diabetes in mice lacking E2F1/E2F2. Unexpectedly, p53 inactivation unmasked oncogenic features of E2F1/E2F2-depleted cells, as evidenced by an accelerated tumor development in triple-knockout mice compared with p53(-/-) mice. Collectively, our data reveal a role for E2F1 and E2F2 as suppressors of replicative stress in differentiating cells, and uncover the existence of a robust E2F-p53 regulatory axis to enable tissue homeostasis and prevent tumorigenesis. These findings have implications in the design of approaches targeting E2F for cancer therapy.


Subject(s)
DNA Replication , E2F1 Transcription Factor/physiology , E2F2 Transcription Factor/physiology , Pancreas/metabolism , Pancreas/pathology , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Atrophy , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , E2F1 Transcription Factor/genetics , E2F2 Transcription Factor/genetics , Male , Mice , Mice, Knockout , Stress, Physiological/genetics
7.
Cancer Sci ; 105(10): 1360-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25088905

ABSTRACT

Mutant mouse models are indispensable tools for clarifying gene functions and elucidating the pathogenic mechanisms of human diseases. Here, we describe novel cancer models bearing point mutations in the retinoblastoma gene (Rb1) generated by N-ethyl-N-nitrosourea mutagenesis. Two mutations in splice sites reduced Rb1 expression and led to a tumor spectrum and incidence similar to those observed in the conventional Rb1 knockout mice. The missense mutant, Rb1(D326V/+) , developed pituitary tumors, but thyroid tumors were completely suppressed. Immunohistochemical analyses of thyroid tissue revealed that E2F1, but not E2F2/3, was selectively inactivated, indicating that the mutant Rb protein (pRb) suppressed thyroid tumors by inactivating E2F1. Interestingly, Rb1(D326V/+) mice developed pituitary tumors that originated from the intermediate lobe of the pituitary, despite selective inactivation of E2F1. Furthermore, in the anterior lobe of the pituitary, other E2F were also inactivated. These observations show that pRb mediates the inactivation of E2F function and its contribution to tumorigenesis is highly dependent on the cell type. Last, by using a reconstitution assay of synthesized proteins, we showed that the D326V missense pRb bound to E2F1 but failed to interact with E2F2/3. These results reveal the effect of the pRb N-terminal domain on E2F function and the impact of the protein on tumorigenesis. Thus, this mutant mouse model can be used to investigate human Rb family-bearing mutations at the N-terminal region.


Subject(s)
E2F1 Transcription Factor/physiology , E2F2 Transcription Factor/physiology , E2F3 Transcription Factor/physiology , Mutation , Retinoblastoma Protein/genetics , Thyroid Neoplasms/genetics , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Thyroid Neoplasms/etiology
8.
Bioinformatics ; 30(21): 3036-43, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25028721

ABSTRACT

MOTIVATION: Oncogenes are known drivers of cancer phenotypes and targets of molecular therapies; however, the complex and diverse signaling mechanisms regulated by oncogenes and potential routes to targeted therapy resistance remain to be fully understood. To this end, we present an approach to infer regulatory mechanisms downstream of the HER2 driver oncogene in SUM-225 metastatic breast cancer cells from dynamic gene expression patterns using a succession of analytical techniques, including a novel MP grammars method to mathematically model putative regulatory interactions among sets of clustered genes. RESULTS: Our method highlighted regulatory interactions previously identified in the cell line and a novel finding that the HER2 oncogene, as opposed to the proto-oncogene, upregulates expression of the E2F2 transcription factor. By targeted gene knockdown we show the significance of this, demonstrating that cancer cell-matrix adhesion and outgrowth were markedly inhibited when E2F2 levels were reduced. Thus, validating in this context that upregulation of E2F2 represents a key intermediate event in a HER2 oncogene-directed gene expression-based signaling circuit. This work demonstrates how predictive modeling of longitudinal gene expression data combined with multiple systems-level analyses can be used to accurately predict downstream signaling pathways. Here, our integrated method was applied to reveal insights as to how the HER2 oncogene drives a specific cancer cell phenotype, but it is adaptable to investigate other oncogenes and model systems. AVAILABILITY AND IMPLEMENTATION: Accessibility of various tools is listed in methods; the Log-Gain Stoichiometric Stepwise algorithm is accessible at http://www.cbmc.it/software/Software.php.


Subject(s)
Breast Neoplasms/genetics , E2F2 Transcription Factor/physiology , Gene Expression Regulation, Neoplastic , Genes, erbB-2 , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Adhesion , Cell Line, Tumor , Cell-Matrix Junctions/metabolism , E2F2 Transcription Factor/genetics , E2F2 Transcription Factor/metabolism , Female , Gene Knockdown Techniques , Humans , Models, Genetic , Proto-Oncogene Mas , Signal Transduction/genetics , Transcription, Genetic , Transcriptome , Up-Regulation
9.
Mol Cell Biol ; 34(17): 3229-43, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24934442

ABSTRACT

While the E2F transcription factors (E2Fs) have a clearly defined role in cell cycle control, recent work has uncovered new functions. Using genomic signature methods, we predicted a role for the activator E2F transcription factors in the mouse mammary tumor virus (MMTV)-polyomavirus middle T oncoprotein (PyMT) mouse model of metastatic breast cancer. To genetically test the hypothesis that the E2Fs function to regulate tumor development and metastasis, we interbred MMTV-PyMT mice with E2F1, E2F2, or E2F3 knockout mice. With the ablation of individual E2Fs, we noted alterations of tumor latency, histology, and vasculature. Interestingly, we noted striking reductions in metastatic capacity and in the number of circulating tumor cells in both the E2F1 and E2F2 knockout backgrounds. Investigating E2F target genes that mediate metastasis, we found that E2F loss led to decreased levels of vascular endothelial growth factor (Vegfa), Bmp4, Cyr61, Nupr1, Plod 2, P4ha1, Adamts1, Lgals3, and Angpt2. These gene expression changes indicate that the E2Fs control the expression of genes critical to angiogenesis, the remodeling of the extracellular matrix, tumor cell survival, and tumor cell interactions with vascular endothelial cells that facilitate metastasis to the lungs. Taken together, these results reveal that the E2F transcription factors play key roles in mediating tumor development and metastasis in addition to their well-characterized roles in cell cycle control.


Subject(s)
E2F Transcription Factors/physiology , Mammary Neoplasms, Experimental/etiology , Animals , Antigens, Polyomavirus Transforming , E2F Transcription Factors/deficiency , E2F Transcription Factors/genetics , E2F1 Transcription Factor/deficiency , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/physiology , E2F2 Transcription Factor/deficiency , E2F2 Transcription Factor/genetics , E2F2 Transcription Factor/physiology , E2F3 Transcription Factor/deficiency , E2F3 Transcription Factor/genetics , E2F3 Transcription Factor/physiology , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/secondary , Mammary Tumor Virus, Mouse , Mice , Mice, Knockout , Neoplastic Cells, Circulating/pathology , Neovascularization, Pathologic/genetics , Retroviridae Infections/etiology , Retroviridae Infections/pathology , Signal Transduction , Tumor Microenvironment , Tumor Virus Infections/etiology , Tumor Virus Infections/pathology
10.
PLoS One ; 8(6): e65755, 2013.
Article in English | MEDLINE | ID: mdl-23799044

ABSTRACT

Insufficient neovascularization, characterized by poor endothelial cell (EC) growth, contributes to the pathogenesis of ischemic heart disease and limits cardiac tissue preservation and regeneration. The E2F family of transcription factors are critical regulators of the genes responsible for cell-cycle progression and growth; however, the specific roles of individual E2Fs in ECs are not well understood. Here we investigated the roles of E2F2 and E2F3 in EC growth, angiogenesis, and their functional impact on myocardial infarction (MI). An endothelial-specific E2F3-deficient mouse strain VE-Cre; E2F3(fl/fl) was generated, and MI was surgically induced in VE-Cre; E2F3(fl/fl) and E2F2-null (E2F2 KO) mice and their wild-type (WT) littermates, VE-Cre; E2F3(+/+) and E2F2 WT, respectively. The cardiac function, infarct size, and vascular density were significantly better in E2F2 KO mice and significantly worse in VE-Cre; E2F3(fl/fl) mice than in their WT littermates. The loss of E2F2 expression was associated with an increase in the proliferation of ECs both in vivo and in vitro, while the loss of E2F3 expression led to declines in EC proliferation. Thus, E2F3 promotes while E2F2 suppresses ischemic cardiac repair through corresponding changes in EC proliferation; and differential targeting of specific E2F members may provide a novel strategy for therapeutic angiogenesis of ischemic heart disease.


Subject(s)
Cell Proliferation , Coronary Vessels/physiopathology , E2F2 Transcription Factor/physiology , E2F3 Transcription Factor/physiology , Neovascularization, Physiologic , Animals , Cells, Cultured , Coronary Vessels/pathology , Endothelial Cells/physiology , Female , Gene Expression , Male , Mice , Mice, Knockout , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Ventricular Function, Left
11.
Cell Death Differ ; 20(7): 931-40, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23558950

ABSTRACT

The 'activating' E2fs (E2f1-3) are transcription factors that potently induce quiescent cells to divide. Work on cultured fibroblasts suggested they were essential for division, but in vivo analysis in the developing retina and other tissues disproved this notion. The retina, therefore, is an ideal location to assess other in vivo adenovirus E2 promoter binding factor (E2f) functions. It is thought that E2f1 directly induces apoptosis, whereas other activating E2fs only induce death indirectly by upregulating E2f1 expression. Indeed, mouse retinoblastoma (Rb)-null retinal neuron death requires E2f1, but not E2f2 or E2f3. However, we report an entirely distinct mechanism in dying cone photoreceptors. These neurons survive Rb loss, but undergo apoptosis in the cancer-prone retina lacking both Rb and its relative p107. We show that while E2f1 killed Rb/p107 null rod, bipolar and ganglion neurons, E2f2 was required and sufficient for cone death, independent of E2f1 and E2f3. Moreover, whereas E2f1-dependent apoptosis was p53 and p73-independent, E2f2 caused p53-dependent cone death. Our in vivo analysis of cone photoreceptors provides unequivocal proof that E2f-induces apoptosis independent of E2f1, and reveals distinct E2f1- and E2f2-activated death pathways in response to a single tumorigenic insult.


Subject(s)
Apoptosis/physiology , E2F1 Transcription Factor/physiology , E2F2 Transcription Factor/physiology , E2F3 Transcription Factor/physiology , Retinal Cone Photoreceptor Cells/pathology , Animals , Apoptosis/genetics , Cell Division/genetics , Cell Division/physiology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , E2F1 Transcription Factor/deficiency , E2F1 Transcription Factor/genetics , E2F2 Transcription Factor/deficiency , E2F2 Transcription Factor/genetics , E2F3 Transcription Factor/deficiency , E2F3 Transcription Factor/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Retina/pathology , Retina/physiopathology , Retinal Cone Photoreceptor Cells/physiology , Retinoblastoma Protein/deficiency , Retinoblastoma Protein/genetics , Retinoblastoma Protein/physiology , Retinoblastoma-Like Protein p107/deficiency , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p107/physiology , Tumor Protein p73 , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology
12.
Am J Physiol Gastrointest Liver Physiol ; 301(1): G20-31, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21527726

ABSTRACT

E2F transcription factors are key regulators of the cell cycle although the relative contribution of each E2F member in regulating cellular proliferation is still poorly defined. Present evidence suggests that E2F2 may act both as a suppressor and promoter of proliferation, depending on the cellular context. We used a loss-of-function mutant mouse model to investigate the function of E2F2 in liver regeneration after partial hepatectomy, a paradigm of cell-cycle progression. Liver mass recovery and histology were examined over 9 days in 70% hepatectomized E2F2(-/-) and wild-type animals. Transcriptome analysis was performed in quiescent and 48-h regenerating liver samples. TIGR MultiExperiment Viewer was used for the statistical analysis of microarray data, significance was determined by Fischer, and P values were adjusted applying Benjamini-Hochberg multiple-testing correction. We show that E2F2 is required for adult hepatocyte proliferation and for timely liver regeneration, as disruption of the E2F2 gene in hepatocytes leads to a reduced rate of S-phase entry and to delayed liver regeneration. Transcriptome analysis followed by ontological classification of differentially expressed genes and gene-interaction network analysis indicated that the majority of genes involved in normal liver regeneration were related to biosynthetic and catabolic processes of all major biomolecules as well as cellular location and intracellular transport, confirming the complex nature of the regeneration process. Remarkably, transcripts of genes included in functional categories that are crucial for cell cycle, apoptosis and wound-healing response, and fibrosis were absent in the transcriptome of posthepatectomized E2F2(-/-) mice. Our results indicate that the transcriptional activity of E2F2 contributes to promote adult hepatocyte proliferation and liver regeneration.


Subject(s)
Cell Proliferation , E2F2 Transcription Factor/physiology , Hepatocytes/physiology , Liver Regeneration/genetics , Animals , E2F2 Transcription Factor/genetics , Female , Gene Expression Profiling , Hepatocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
13.
Dev Biol ; 351(1): 35-45, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21185283

ABSTRACT

E2F transcription factors regulate the progression of the cell cycle by repression or transactivation of genes that encode cyclins, cyclin dependent kinases, checkpoint regulators, and replication proteins. Although some E2F functions are independent of the Retinoblastoma tumor suppressor (Rb) and related family members, p107 and p130, much of E2F-mediated repression of S phase entry is dependent upon Rb. We previously showed in cultured mouse embryonic fibroblasts that concomitant loss of three E2F activators with overlapping functions (E2F1, E2F2, and E2F3) triggered the p53-p21(Cip1) response and caused cell cycle arrest. Here we report on a dramatic difference in the requirement for E2F during development and in cultured cells by showing that cell cycle entry occurs normally in E2f1-3 triply-deficient epithelial stem cells and progenitors of the developing lens. Sixteen days after birth, however, massive apoptosis in differentiating epithelium leads to a collapse of the entire eye. Prior to this collapse, we find that expression of cell cycle-regulated genes in E2F-deficient lenses is aberrantly high. In a second set of experiments, we demonstrate that E2F3 ablation alone does not cause abnormalities in lens development but rescues phenotypic defects caused by loss of Rb, a binding partner of E2F known to recruit histone deacetylases, SWI/SNF and CtBP-polycomb complexes, methyltransferases, and other co-repressors to gene promoters. Together, these data implicate E2F1-3 in mediating transcriptional repression by Rb during cell cycle exit and point to a critical role for their repressive functions in cell survival.


Subject(s)
Cell Proliferation , E2F1 Transcription Factor/physiology , E2F2 Transcription Factor/physiology , E2F3 Transcription Factor/physiology , Repressor Proteins/physiology , Animals , Apoptosis , Cell Survival , DNA Breaks, Double-Stranded , E2F1 Transcription Factor/deficiency , E2F2 Transcription Factor/deficiency , E2F3 Transcription Factor/deficiency , Epithelial Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Retinoblastoma Protein/physiology , Tumor Suppressor Protein p53/physiology
14.
Dev Biol ; 346(1): 80-9, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20659447

ABSTRACT

The ability of ionizing radiation (IR) to induce apoptosis independent of p53 is crucial for successful therapy of cancers bearing p53 mutations. p53-independent apoptosis, however, remains poorly understood relative to p53-dependent apoptosis. IR induces both p53-dependent and p53-independent apoptoses in Drosophila melanogaster, making studies of both modes of cell death possible in a genetically tractable model. Previous studies have found that Drosophila E2F proteins are generally pro-death or neutral with regard to p53-dependent apoptosis. We report here that dE2F1 promotes IR-induced p53-independent apoptosis in larval imaginal discs. Using transcriptional reporters, we provide evidence that, when p53 is mutated, dE2F1 becomes necessary for the transcriptional induction of the pro-apoptotic gene hid after irradiation. In contrast, the second E2F homolog, dE2F2, as well as the net E2F activity, which can be depleted by mutating the common cofactor, dDp, is inhibitory for p53-independent apoptosis. We conclude that p53-dependent and p53-independent apoptoses show differential reliance on E2F activity in Drosophila.


Subject(s)
Apoptosis/radiation effects , Drosophila Proteins/physiology , Drosophila melanogaster/physiology , E2F1 Transcription Factor/physiology , E2F2 Transcription Factor/physiology , Tumor Suppressor Protein p53/physiology , Animals , Checkpoint Kinase 2 , DNA-Binding Proteins/physiology , Drosophila melanogaster/cytology , Drosophila melanogaster/radiation effects , Mitotic Index , Protein Serine-Threonine Kinases/physiology
15.
Mol Cell Proteomics ; 9(10): 2184-94, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20573986

ABSTRACT

E2F transcription factors (E2F1-8) are best known for their role in cell proliferation, although it is clear that they regulate many other biological processes through the transcriptional modulation of distinct target genes. However, the specific set of genes regulated by each E2F remains to be characterized. To gain insight into the molecular pathways regulated by E2F2, we have analyzed the proteome of antigen receptor-activated T cells lacking E2F2. We report that loss of E2F2 results in a deregulated Aryl-hydrocarbon-receptor pathway. Proliferating E2F2(-/-) T lymphocytes expressed significantly higher levels of Aip, Ahr, and Arnt relative to wild-type (WT)(1) controls. The mechanism for increased levels of Aip appears straightforward, involving direct regulation of the Aip gene promoter by E2F2. Although the Ahr and Arnt promoters also bind E2F2, their regulation appears to be more complex. Nevertheless, exposure to the environmental xenobiotic 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a well-known exogenous ligand of the Ahr pathway, led to overexpression of the Ahr target gene Cyp1a1, and to increased sensitivity to TCDD-triggered apoptosis in E2F2(-/-) T cells compared with WT controls. These results suggest that E2F2 modulates cellular sensitivity to xenobiotic signals through the negative regulation of the Ahr pathway.


Subject(s)
E2F2 Transcription Factor/physiology , Proteomics , Receptors, Aryl Hydrocarbon/metabolism , T-Lymphocytes/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Electrophoresis, Gel, Two-Dimensional , Flow Cytometry , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Phosphorylation , Polychlorinated Dibenzodioxins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
16.
Mol Carcinog ; 49(2): 152-6, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19798698

ABSTRACT

Deregulation of E2F transcriptional activity as a result of alterations in the p16-cyclin D-Rb pathway is a hallmark of cancer. However, the roles of the different E2F family members in the process of tumorigenesis are still being elucidated. Studies in mice and humans suggest that E2F2 functions as a tumor suppressor. Here we demonstrate that E2f2 inactivation cooperates with transgenic expression of Myc to enhance tumor development in the skin and oral cavity. In fact, hemizygosity at the E2f2 locus was sufficient to increase tumor incidence in this model. Loss of E2F2 enhanced proliferation in Myc transgenic tissue but did not affect Myc-induced apoptosis. E2F2 did not behave as a simple activator of transcription in epidermal keratinocytes but instead appeared to differentially regulate gene expression dependent on the individual target. E2f2 inactivation also altered the changes in gene expression in Myc transgenic cells by enhancing the increase of some genes, such as cyclin E, and reversing the repression of other genes. These findings demonstrate that E2F2 can function as a tumor suppressor in epithelial tissues, perhaps by limiting proliferation in response to Myc.


Subject(s)
Cell Proliferation , Cell Transformation, Neoplastic , E2F2 Transcription Factor/physiology , Genes, myc , Animals , Blotting, Western , Immunohistochemistry , Mice , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction
17.
J Immunol ; 180(6): 3699-707, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18322175

ABSTRACT

The E2F4 protein is involved in gene repression and cell cycle exit, and also has poorly understood effects in differentiation. We analyzed the impact of E2F4 deficiency on early steps in mouse hematopoietic development, and found defects in early hematopoietic progenitor cells that were propagated through common lymphoid precursors to the B and T lineages. In contrast, the defects in erythromyeloid precursor cells were self-correcting over time. This suggests that E2F4 is important in early stages of commitment to the lymphoid lineage. The E2F4-deficient progenitor cells showed reduced expression of several key lymphoid-lineage genes, and overexpression of two erythromyeloid lineage genes. However, we did not detect effects on cell proliferation. These findings emphasize the significance of E2F4 in controlling gene expression and cell fate.


Subject(s)
Cell Differentiation/immunology , Cell Lineage/immunology , E2F2 Transcription Factor/physiology , Gene Expression Regulation/immunology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Lymphocytes/cytology , Lymphocytes/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Cell Differentiation/genetics , Cell Lineage/genetics , E2F2 Transcription Factor/deficiency , E2F2 Transcription Factor/genetics , Female , Fetus , Ikaros Transcription Factor/deficiency , Ikaros Transcription Factor/genetics , Liver/cytology , Liver/immunology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cells/cytology , Myeloid Cells/immunology , Myeloid Cells/metabolism
18.
Carcinogenesis ; 29(1): 194-201, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17916908

ABSTRACT

The inhibitor of apoptosis protein survivin is a dual mediator of apoptosis resistance and cell cycle progression and is highly expressed in cancer. We have shown previously that survivin is up-regulated in melanoma compared with normal melanocytes, is required for melanoma cell viability, and that melanocyte expression of survivin predisposes mice to ultraviolet-induced melanoma and metastasis. The mechanism of survivin up-regulation in the course of melanocyte transformation and its repression in normal melanocytes, however, has not been clearly defined. We show here that p53 and retinoblastoma (Rb), at basal levels and in the absence of any activating stimuli, are both required to repress survivin transcription in normal human melanocytes. Survivin repression in melanocytes does not involve alterations in protein stability or promoter methylation. p53 and Rb (via E2Fs) regulate survivin expression by direct binding to the survivin promoter; p53 also affects survivin expression by activating p21. We demonstrate a novel role for E2F2 in the negative regulation of survivin expression. In addition, we identify a novel E2F-binding site in the survivin promoter and show that mutation of either the p53- or E2F-binding sites is sufficient to increase promoter activity. These studies suggest that compromise of either p53 or Rb pathways during melanocyte transformation leads to up-regulation of survivin expression in melanoma.


Subject(s)
E2F2 Transcription Factor/physiology , Melanocytes/metabolism , Microtubule-Associated Proteins/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Retinoblastoma Protein/physiology , Tumor Suppressor Protein p53/physiology , Cells, Cultured , DNA Methylation , Humans , Inhibitor of Apoptosis Proteins , Promoter Regions, Genetic , Survivin
19.
Proc Natl Acad Sci U S A ; 104(39): 15400-5, 2007 Sep 25.
Article in English | MEDLINE | ID: mdl-17881568

ABSTRACT

Deregulation of the Myc pathway and deregulation of the Rb pathway are two of the most common abnormalities in human malignancies. Recent in vitro experiments suggest a complex cross-regulatory relationship between Myc and Rb that is mediated through the control of E2F. To evaluate the functional connection between Myc and E2Fs in vivo, we used a bitransgenic mouse model of Myc-induced T cell lymphomagenesis and analyzed tumor progression in mice deficient for E2f1, E2f2, or E2f3. Whereas the targeted inactivation of E2f1 or E2f3 had no significant effect on tumor progression, loss of E2f2 accelerated lymphomagenesis. Interestingly, loss of a single copy of E2f2 also accelerated tumorigenesis, albeit to a lesser extent, suggesting a haploinsufficient function for this locus. The combined ablation of E2f1 or E2f3, along with E2f2, did not further accelerate tumorigenesis. Myc-overexpressing T cells were more resistant to apoptosis in the absence of E2f2, and the reintroduction of E2F2 into these tumor cells resulted in an increase of apoptosis and inhibition of tumorigenesis. These results identify the E2f2 locus as a tumor suppressor through its ability to modulate apoptosis.


Subject(s)
E2F2 Transcription Factor/physiology , Lymphoma/metabolism , Proto-Oncogene Proteins c-myc/metabolism , T-Lymphocytes/metabolism , Animals , Apoptosis , Cell Cycle , Disease Progression , E2F1 Transcription Factor/metabolism , E2F2 Transcription Factor/metabolism , E2F3 Transcription Factor/metabolism , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , Lymphoma/pathology , Mice , Mice, Transgenic
20.
Mol Cell Biol ; 27(1): 65-78, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17167174

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.


Subject(s)
E2F1 Transcription Factor/physiology , E2F2 Transcription Factor/physiology , E2F3 Transcription Factor/physiology , Gene Expression Regulation , Tumor Suppressor Protein p53/metabolism , Animals , Cell Proliferation , Fibroblasts/metabolism , Gene Targeting , Mice , Mice, Knockout , Mutation , Phosphorylation , Retinoblastoma Protein/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...