Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell Rep ; 27(12): 3547-3560.e5, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31130414

RESUMEN

Orchestrating cell-cycle-dependent mRNA oscillations is critical to cell proliferation in multicellular organisms. Even though our understanding of cell-cycle-regulated transcription has improved significantly over the last three decades, the mechanisms remain untested in vivo. Unbiased transcriptomic profiling of G0, G1-S, and S-G2-M sorted cells from FUCCI mouse embryos suggested a central role for E2Fs in the control of cell-cycle-dependent gene expression. The analysis of gene expression and E2F-tagged knockin mice with tissue imaging and deep-learning tools suggested that post-transcriptional mechanisms universally coordinate the nuclear accumulation of E2F activators (E2F3A) and canonical (E2F4) and atypical (E2F8) repressors during the cell cycle in vivo. In summary, we mapped the spatiotemporal expression of sentinel E2F activators and canonical and atypical repressors at the single-cell level in vivo and propose that two distinct E2F modules relay the control of gene expression in cells actively cycling (E2F3A-8-4) and exiting the cycle (E2F3A-4) during mammalian development.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular , Diferenciación Celular , Factor de Transcripción E2F3/fisiología , Factor de Transcripción E2F4/fisiología , Regulación de la Expresión Génica , Proteínas Represoras/fisiología , Animales , Proteínas de Ciclo Celular/genética , Proliferación Celular , Células Cultivadas , Femenino , Masculino , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Transcriptoma
2.
Gene ; 684: 39-46, 2019 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-30359737

RESUMEN

Sirtuin 5 (SIRT5) belongs to the mitochondrial sirtuin family, which constitutes a highly conserved family of nicotinamide adenine dinucleotide NAD+-dependent deacetylases and ADP-ribosyltransferases that play important regulatory roles in stress resistance and metabolic homeostasis. SIRT5 was shown to have deacetylase, desuccinylase, and demalonylase activities. However, the mechanisms regulating SIRT5 transcription remain unclear. To explore the molecular regulation of bovine SIRT5 expression, we obtained a 500-base pair fragment of the 5'-regulatory region of bovine SIRT5 by molecular cloning, which contained a region with 3 CpG islands. Electrophoretic mobility shift assays and luciferase reporter assays revealed the E2F transcription factor 4 (E2F4) and Kruppel-like factor 6 (KLF6) binding sites as transcriptional activators or repressors in the promoter region of SIRT5. We further verified that E2F4 and KLF6 bind to the SIRT5 promoter by chromatin immunoprecipitation assays. Additionally, methylation and luciferase report assays showed that SIRT5 promoter activity was enhanced by demethylation, and transcriptional activation by E2F4 and transcriptional inhibition by KLF6 of SIRT5 expression was strengthened by demethylation during adipocytes differentiation. This study focused on the mechanism underlying the methylation and transcriptional regulation of SIRT5 expression in bovine adipocytes.


Asunto(s)
Factor de Transcripción E2F4/metabolismo , Factor 6 Similar a Kruppel/metabolismo , Sirtuinas/genética , Adipocitos/metabolismo , Animales , Sitios de Unión/genética , Bovinos , Islas de CpG/genética , Metilación de ADN/genética , Factor de Transcripción E2F4/fisiología , Ensayo de Cambio de Movilidad Electroforética , Regulación de la Expresión Génica/genética , Factor 6 Similar a Kruppel/fisiología , Factores de Transcripción de Tipo Kruppel/genética , Regiones Promotoras Genéticas/genética , Sirtuinas/metabolismo , Factores de Transcripción/genética , Activación Transcripcional
3.
Dev Biol ; 443(2): 165-172, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30218642

RESUMEN

Multiciliated cells (MCCs) differentiate arrays of motile cilia that beat to drive fluid flow over epithelia. Recent studies have established two Geminin family coiled-coil containing nuclear regulatory proteins, Gmnc and Multicilin (Mci), in the specification and differentiation of the MCCs. Both Gmnc and Mci are devoid of a DNA binding domain: they regulate transcription by associating with E2f family transcription factors, notably E2f4 and E2f5. Here, we have studied the relative contribution of these two E2f factors in MCC development using the zebrafish embryo, which differentiates MCCs within kidney tubules and the nose. We found that while E2f4 is fully dispensable, E2f5 is essential for MCCs to form in the kidney tubules. Moreover, using a variety of double mutant combinations we show that E2f5 has a more prominent role in MCC development in the zebrafish than E2f4. This contrasts with current evidence from the mouse, where E2f4 seems to be more important. Thus, distinct combinatorial activities of the E2f4 and E2f5 proteins regulate the specification and differentiation of MCCs in zebrafish and mice.


Asunto(s)
Factor de Transcripción E2F4/metabolismo , Factor de Transcripción E2F5/metabolismo , Pez Cebra/embriología , Animales , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/genética , Cilios/metabolismo , Cilios/fisiología , Factor de Transcripción E2F4/fisiología , Factor de Transcripción E2F5/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Células HEK293 , Humanos , Proteínas Nucleares/metabolismo , Factores de Transcripción , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
4.
Cancer Lett ; 412: 59-68, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28987387

RESUMEN

The members of the miR-17-92 cluster are upregulated in various cancers and function as a cluster of oncogenic miRNA. Our study characterized a new function of miR-17-5p, a member of the miR-17-92 cluster, in regulating cell proliferation in pancreatic cancer. Our results indicate that miR-17-5p was up-regulated in pancreatic adenocarcinoma and directly targeted the retinoblastoma-like protein 2 (RBL2), a tumor suppressor belonging to the Rb family. High levels of miR-17-5p and low levels of RBL2 were associated with poor prognosis. RBL2 interacted with the transcription factor E2F4 and bound to the promoter regions of the E2F target genes. Disruption of the RBL2/E2F4 complex by miR-17-5p overexpression shifted the activity of E2F from gene repressing to gene activating, which induced cell cycle entry and proliferation. These results suggest that miR-17-5p promoted proliferation in pancreatic ductal adenocarcinoma cells (PDAC), and altered cell cycle profiles in vivo and in vitro, by disrupting the RBL2/E2F4-associated gene repressing complexes via direct targeting of RBL2. The new regulatory network, involving miR-17-5p and RBL2, emerges as a new target of PDAC treatment.


Asunto(s)
Adenocarcinoma/patología , Carcinoma Ductal Pancreático/patología , Factor de Transcripción E2F4/fisiología , MicroARNs/fisiología , Neoplasias Pancreáticas/patología , Proteína p130 Similar a la del Retinoblastoma/fisiología , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Factor de Transcripción E2F4/genética , Humanos , Proteína p130 Similar a la del Retinoblastoma/genética
5.
Nat Commun ; 7: 11091, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-27033104

RESUMEN

Germline mutations affecting telomere maintenance or DNA repair may, respectively, cause dyskeratosis congenita or Fanconi anaemia, two clinically related bone marrow failure syndromes. Mice expressing p53(Δ31), a mutant p53 lacking the C terminus, model dyskeratosis congenita. Accordingly, the increased p53 activity in p53(Δ31/Δ31) fibroblasts correlated with a decreased expression of 4 genes implicated in telomere syndromes. Here we show that these cells exhibit decreased mRNA levels for additional genes contributing to telomere metabolism, but also, surprisingly, for 12 genes mutated in Fanconi anaemia. Furthermore, p53(Δ31/Δ31) fibroblasts exhibit a reduced capacity to repair DNA interstrand crosslinks, a typical feature of Fanconi anaemia cells. Importantly, the p53-dependent downregulation of Fanc genes is largely conserved in human cells. Defective DNA repair is known to activate p53, but our results indicate that, conversely, an increased p53 activity may attenuate the Fanconi anaemia DNA repair pathway, defining a positive regulatory feedback loop.


Asunto(s)
Reparación del ADN , Regulación hacia Abajo , Anemia de Fanconi/genética , Proteína p53 Supresora de Tumor/fisiología , Animales , Células Cultivadas , Factor de Transcripción E2F4/genética , Factor de Transcripción E2F4/metabolismo , Factor de Transcripción E2F4/fisiología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/fisiología , Humanos , Ratones , Células 3T3 NIH , Transcriptoma
6.
Mol Med Rep ; 12(6): 7891-8, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26498477

RESUMEN

The present study was aimed at screening the key genes associated with abdominal aortic aneurysm (AAA) in the neck, and to investigate the molecular mechanism underlying the development of AAA. The gene expression profile, GSE47472, including 14 AAA neck samples and eight donor controls, was downloaded from the Gene Expression Omnibus database. The total AAA samples were grouped into two types to avoid bias. Differentially expressed genes (DEGs) were screened in patients with AAA and subsequently compared with donor controls using linear models for microarray data, or the Limma package in R, followed by gene ontology enrichment analysis. Furthermore, a protein­protein interaction (PPI) network based on the DEGs was constructed to detect highly connected regions using a Cytoscape plugin. In total, 388 DEGs in the AAA samples were identified. These DEGs were predominantly associated with limb development, including embryonic limb development and appendage development. Nuclear receptor co­repressor 1 (NCOR1), histone 4 (H4), E2F transcription factor 4 (E2F4) and hepatocyte nuclear factor 4α (HNF4A) were the four transcription factors associated with AAA. Furthermore, HNF4A indirectly interacted with the other three transcription factors. Additionally, six clusters were selected from the PPI network. The DEG screening process and the construction of an interaction network enabled an understanding of the mechanism of AAA to be gleaned. HNF4A may exert an important role in AAA development through its interactions with the three other transcription factors (E2F4, NCOR1 and H4), and the mechanism of this coordinated regulation of the transcription factors in AAA may provide a suitable target for the development of therapeutic intervention strategies.


Asunto(s)
Aneurisma de la Aorta Abdominal/genética , Perfilación de la Expresión Génica , Aneurisma de la Aorta Abdominal/metabolismo , Biología Computacional/métodos , Factor de Transcripción E2F4/genética , Factor de Transcripción E2F4/metabolismo , Factor de Transcripción E2F4/fisiología , Regulación de la Expresión Génica , Estudios de Asociación Genética , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Factor Nuclear 4 del Hepatocito/fisiología , Histonas/genética , Histonas/metabolismo , Histonas/fisiología , Humanos , Co-Represor 1 de Receptor Nuclear/genética , Co-Represor 1 de Receptor Nuclear/metabolismo , Co-Represor 1 de Receptor Nuclear/fisiología , Mapeo de Interacción de Proteínas , Programas Informáticos
7.
Curr Opin Oncol ; 26(4): 415-21, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24840522

RESUMEN

PURPOSE OF REVIEW: Although most gastrointestinal stromal tumors respond well to treatment with the small molecule kinase inhibitor imatinib mesylate (Gleevec), complete remissions are rare and the majority of patients achieve disease stabilization. Furthermore, discontinuation of treatment in the presence of residual tumor mass almost inevitably leads to tumor progression. These observations suggest that a subset of tumor cells not only persists under imatinib treatment, but remains viable. The current article reviews the molecular basis for these findings and explores strategies to exploit them therapeutically. RECENT FINDINGS: Although imatinib induces apoptosis in a subset of gastrointestinal stromal tumor cells, it leads to a reversible exit from the cell division cycle and entry into G0, a cell cycle state called quiescence, in the remaining cells. Mechanistically, this process involves the DREAM complex (DP, p130/RBL2, E2F4 and MuvB), a newly identified key regulator of quiescence. Interfering with DREAM complex formation either by siRNA-mediated knockdown or by pharmacological inhibition of the regulatory kinase dual-specificity tyrosine phosphorylation-regulated kinase 1A was shown to enhance imatinib-induced gastrointestinal stromal tumor cell death. SUMMARY: Targeting the DREAM complex and imatinib-induced quiescence could provide opportunities for future therapeutic interventions toward more efficient imatinib responses.


Asunto(s)
Antineoplásicos/uso terapéutico , Benzamidas/uso terapéutico , Neoplasias Gastrointestinales/tratamiento farmacológico , Piperazinas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/uso terapéutico , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Factor de Transcripción E2F4/fisiología , Neoplasias Gastrointestinales/patología , Humanos , Mesilato de Imatinib , Terapia Molecular Dirigida/métodos , Proteína p130 Similar a la del Retinoblastoma/fisiología
8.
Cell Mol Life Sci ; 71(18): 3583-97, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24573694

RESUMEN

The inhibitor of differentiation Id2, a protein lacking the basic DNA-binding domain, is involved in the modulation of a number of biological processes. The molecular mechanisms explaining Id2 pleiotropic functions are poorly understood. Id2 and E2F4 are known to bind simultaneously to c-myc promoter. To study whether Id2 plays a global role on transcriptional regulation, we performed in vivo genome-wide ChIP/chip experiments for Id2 and E2F4 in adult mouse liver. An Id2-containing complex was bound to a common sequence downstream from the TSS on a subset of 442 E2F4 target genes mainly related to cell development and chromatin structure. We found a positive correlation between Id2 protein levels and the expression of E2F4/Id2 targets in fetal and adult liver. Id2 protein stability increased in fetal liver by interaction with USP1 de-ubiquitinating enzyme, which was induced during development. In adult liver, USP1 and Id2 levels dramatically decreased. In differentiated liver tissue, when Id2 concentration was low, E2F4/Id2 was bound to the same region as paused Pol II and target genes remained transcriptionally inactive. Conversely, in fetal liver when Id2 levels were increased, Id2 and Pol II were released from gene promoters and target genes up-regulated. During liver regeneration after partial hepatectomy, we obtained the same results as in fetal liver. Our results suggest that Id2 might be part of a reversible development-related program involved in the paused-ON/OFF state of Pol II on selected genes that would remain responsive to specific stimuli.


Asunto(s)
Factor de Transcripción E2F4/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Hígado/metabolismo , Animales , Factor de Transcripción E2F4/fisiología , Proteína 2 Inhibidora de la Diferenciación/fisiología , Regeneración Hepática/genética , Ratones , ARN Polimerasa II/metabolismo , ARN Polimerasa II/fisiología
9.
Surg Obes Relat Dis ; 9(1): 94-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22138333

RESUMEN

BACKGROUND: The insulin-like growth factor-1 (IGF-1) signaling pathway promotes adipocyte differentiation and, therefore, insulin sensitivity by suppression of necdin expression, which represses peroxisome proliferator-activated receptor-gamma promoter activity by interaction with E2F4 in mouse adipocytes. The aim of the present study was to test the hypothesis that this pathway represents one of the mechanisms by which Roux-en-Y gastric bypass surgery (RYGB) induces resolution of insulin resistance. METHODS: Clinical samples were collected and the key biomarkers measured to test the hypothesis that the IGF-1 pathway represents 1 of the mechanisms by which RYGB induces resolution of insulin resistance in obese individuals. RESULTS: Free IGF-1 levels were significantly greater in the post-RYGB patients than in the pre-RYGB obese patients (2.55 ± 1.54 versus 1.32 ± .65 µg/L, P = .03) and similar to that in normal weight controls (2.54 ± 1.27 µg/L). Necdin and E2F4 gene expression in the adipose tissue was significantly downregulated after RYGB compared with obese and were similar to the levels observed in the controls. In mature human adipocytes cultured in vitro, treatment with des-IGF-1 induced downregulation of necdin and E2F4 gene expression in a dose-dependent manner (P = .01). CONCLUSION: After RYGB, the insulin/IGF-1 signaling pathway is activated and could account for the observed decrease in the expression of necdin, which represses peroxisome proliferator-activated receptor-gamma promoter activity by interaction with E2F4. This could represent one of the mechanisms that induce resolution of insulin resistance after RYGB.


Asunto(s)
Factor de Transcripción E2F4/fisiología , Derivación Gástrica , Resistencia a la Insulina/fisiología , Proteínas del Tejido Nervioso/fisiología , Proteínas Nucleares/fisiología , Pérdida de Peso/fisiología , Adipocitos/fisiología , Adulto , Estudios de Casos y Controles , Células Cultivadas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Obesidad Mórbida/fisiopatología , Obesidad Mórbida/cirugía , Estudios Prospectivos
10.
Leukemia ; 26(10): 2277-85, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22475873

RESUMEN

Sporadic Burkitt lymphoma (sBL) is a rapidly growing B-cell non-Hodgkin's lymphoma whose treatment requires highly aggressive therapies that often result severely toxic. Identification of proteins whose expression or function is deregulated in sBL and play a role in its formation could facilitate development of less toxic therapies. We have previously shown that E2F1 expression is deregulated in sBL. We have now investigated the mechanisms underlying E2F1 deregulation and found that the E2F sites in its promoter fail to repress its transcriptional activity in BL cells and that the transcriptional repressor E2F4 barely interacts with these sites. We also have found that E2F4 protein levels, but not those of its mRNA, are reduced in sBL cell lines relative to immortal B-cell lines. E2F4 protein expression is also decreased in 24 of 26 sBL tumor samples from patients compared with control tissues. Our data demonstrate that enforced E2F4 expression in BL cells not only diminishes E2F1 levels, but also reduces selectively the tumorigenic properties and proliferation of BL cells, while increasing their accumulation in G(2)/M. Our results therefore point to E2F4 as a target for developing novel and less toxic treatments for sBL.


Asunto(s)
Linfoma de Burkitt/etiología , Transformación Celular Neoplásica , Factor de Transcripción E2F4/fisiología , Animales , Linfoma de Burkitt/química , División Celular , Línea Celular Tumoral , Transformación Celular Neoplásica/química , Proteína Sustrato Asociada a CrK/fisiología , Factor de Transcripción E2F1/análisis , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F4/análisis , Femenino , Fase G2 , Humanos , Ratones , Células 3T3 NIH , Regiones Promotoras Genéticas
11.
J Clin Invest ; 120(10): 3530-44, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20890043

RESUMEN

Human parvovirus B19 (B19V) is the only human pathogenic parvovirus. It causes a wide spectrum of human diseases, including fifth disease (erythema infectiosum) in children and pure red cell aplasia in immunocompromised patients. B19V is highly erythrotropic and preferentially replicates in erythroid progenitor cells (EPCs). Current understanding of how B19V interacts with cellular factors to regulate disease progression is limited, due to a lack of permissive cell lines and animal models. Here, we employed a recently developed primary human CD36(+) EPC culture system that is highly permissive for B19V infection to identify cellular factors that lead to cell cycle arrest after B19V infection. We found that B19V exploited the E2F family of transcription factors by downregulating activating E2Fs (E2F1 to E2F3a) and upregulating repressive E2Fs (E2F4 to E2F8) in the primary CD36(+) EPCs. B19V nonstructural protein 1 (NS1) was a key viral factor responsible for altering E2F1-E2F5 expression, but not E2F6-E2F8 expression. Interaction between NS1 and E2F4 or E2F5 enhanced the nuclear import of these repressive E2Fs and induced stable G2 arrest. NS1-induced G2 arrest was independent of p53 activation and increased viral replication. Downstream E2F4/E2F5 targets, which are potentially involved in the progression from G2 into M phase and erythroid differentiation, were identified by microarray analysis. These findings provide new insight into the molecular pathogenesis of B19V in highly permissive erythroid progenitors.


Asunto(s)
Factores de Transcripción E2F/fisiología , Células Precursoras Eritroides/citología , Parvovirus B19 Humano/patogenicidad , Transporte Activo de Núcleo Celular , Antígenos CD36/análisis , Ciclo Celular , Diferenciación Celular , Células Cultivadas , Factor de Transcripción E2F4/fisiología , Factor de Transcripción E2F5/fisiología , Células Precursoras Eritroides/virología , Humanos , Transducción de Señal , Proteína p53 Supresora de Tumor/fisiología , Proteínas no Estructurales Virales/fisiología , Replicación Viral
12.
Cell Cycle ; 9(2): 371-6, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20023434

RESUMEN

The retinoblastoma tumor suppressor protein pRB functions, at least in part, by directly binding to and modulating the activity of the E2F transcription factors. Previous studies have shown that both E2F4 and pRB play important roles in fetal erythropoiesis. Given that these two proteins interact directly we investigated the overlap of E2F4 and pRB function in this process by analyzing E2f4(-/-), conditional Rb knockout (Rb(1lox/1lox)), and compound E2f4(-/-);Rb(1lox/1lox) embryos. At E15.5 E2f4(-/-) and Rb(1lox/1lox) fetal erythroid cells display distinct abnormalities in their differentiation profiles. When cultured in vitro, both E2f4(-/-) and Rb(1lox/1lox) erythroid cells show defects in cell cycle progression. Surprisingly, analysis of cell cycle profiling suggests that E2F4 and pRB control cell cycle exit through different mechanisms. Moreover, only pRB, but not E2F4, promotes cell survival in erythroid cells. We observed an additive rather than a synergistic impact upon the erythroid defects in the compound E2f4(-/-);Rb(1lox/1lox) embryos. We further found that fetal liver macrophage development is largely normal regardless of genotype. Taken together, our results show that E2F4 and pRB play independent cell-intrinsic roles in fetal erythropoiesis.


Asunto(s)
Factor de Transcripción E2F4/metabolismo , Células Eritroides/citología , Eritropoyesis , Proteína de Retinoblastoma/metabolismo , Animales , Apoptosis , Diferenciación Celular , Supervivencia Celular , Factor de Transcripción E2F4/genética , Factor de Transcripción E2F4/fisiología , Embrión de Mamíferos/citología , Feto , Técnicas de Inactivación de Genes , Macrófagos/citología , Macrófagos/inmunología , Ratones , Ratones Transgénicos , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/fisiología
13.
Int J Cancer ; 125(9): 2086-94, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19585502

RESUMEN

Chlorophyllin (CHL) is a water-soluble derivative of chlorophyll that exhibits cancer chemopreventive properties, but which also has been studied for its possible cancer therapeutic effects. We report here that human colon cancer cells treated with CHL accumulate in S-phase of the cell cycle, and this is associated with reduced expression levels of p53, p21, and other G(1)/S checkpoint controls. At the same time, E2F1 and E2F4 transcription factors become elevated and exhibit increased DNA binding activity. In CHL-treated colon cancer cells, bromodeoxyuridine pulse-chase experiments provided evidence for the inhibition of DNA synthesis. Ribonucleotide reductase (RR), a pivotal enzyme for DNA synthesis and repair, was reduced at the mRNA and protein level after CHL treatment, and the enzymatic activity was inhibited in a concentration-dependent manner both in vitro and in vivo. Immunoblotting revealed that expression levels of RR subunits R1, R2, and p53R2 were reduced by CHL treatment in HCT116 (p53(+/+)) and HCT116 (p53(-/-)) cells, supporting a p53-independent mechanism. Prior studies have shown that reduced levels of RR small subunits can increase the sensitivity of colon cancer cells to clinically used DNA-damaging agents and RR inhibitors. We conclude that by inhibiting R1, R2, and p53R2, CHL has the potential to be effective in the clinical setting, when used alone or in combination with currently available cancer therapeutic agents.


Asunto(s)
Anticarcinógenos/farmacología , Clorofilidas/farmacología , Neoplasias del Colon/patología , Factor de Transcripción E2F4/fisiología , Ribonucleótido Reductasas/fisiología , Fase S/efectos de los fármacos , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , ADN/metabolismo , Factor de Transcripción E2F1/análisis , Factor de Transcripción E2F1/metabolismo , Factor de Transcripción E2F4/análisis , Humanos , Ribonucleótido Reductasas/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/fisiología
14.
J Neurosci ; 27(22): 5926-35, 2007 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-17537963

RESUMEN

Early forebrain development is characterized by extensive proliferation of neural precursors coupled with complex structural transformations; however, little is known regarding the mechanisms by which these processes are integrated. Here, we show that deficiency of the cell cycle regulatory protein, E2F4, results in the loss of ventral telencephalic structures and impaired self-renewal of neural precursor cells. The mechanism underlying aberrant ventral patterning lies in a dramatic loss of Sonic hedgehog (Shh) expression specifically in this region. The E2F4-deficient phenotype can be recapitulated by interbreeding mice heterozygous for E2F4 with those lacking one allele of Shh, suggesting a genetic interaction between these pathways. Treatment of E2F4-deficient cells with a Hh agonist rescues stem cell self-renewal and cells expressing the homeodomain proteins that specify the ventral telencephalic structures. Finally, we show that E2F4 deficiency results in impaired activity of Shh forebrain-specific enhancers. In conclusion, these studies establish a novel requirement for the cell cycle regulatory protein, E2F4, in the development of the ventral telencephalon.


Asunto(s)
Ciclo Celular/fisiología , Factor de Transcripción E2F4/fisiología , Telencéfalo/embriología , Telencéfalo/metabolismo , Animales , Células Cultivadas , Factor de Transcripción E2F4/deficiencia , Femenino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo , Telencéfalo/citología , Telencéfalo/crecimiento & desarrollo
15.
Dev Biol ; 305(2): 564-76, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17383628

RESUMEN

The airway epithelium is comprised of specialized cell types that play key roles in protecting the lungs from environmental insults. The cellular composition of the murine respiratory epithelium is established during development and different cell types populate specific regions along the airway. Here we show that E2f4-deficiency leads to an absence of ciliated cells from the entire airway epithelium and the epithelium of the submucosal glands in the paranasal sinuses. This defect is particularly striking in the nasal epithelium of E2f4-/- mice where ciliated cells are replaced by columnar secretory cells that produce mucin-like substances. In addition, in the proximal lung, E2f4 loss causes a reduction in Clara cell marker expression indicating that Clara cell development is also affected. These defects arise during embryogenesis and, in the nasal epithelium, appear to be independent of any changes in cell proliferation, the principal process regulated by members of the E2f family of transcription factors. We therefore conclude that E2f4 is required to determine the appropriate development of the airway epithelium. Importantly, the combination of no ciliated cells and excess mucous cells can account for the chronic rhinitis and increased susceptibility to opportunistic infections that causes the postnatal lethality of E2f4 mutant mice.


Asunto(s)
Factor de Transcripción E2F4/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Mucosa Respiratoria/embriología , Animales , Proliferación Celular , Cilios/patología , Cilios/ultraestructura , Factor de Transcripción E2F4/deficiencia , Factor de Transcripción E2F4/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mucosa Respiratoria/patología , Mucosa Respiratoria/ultraestructura
16.
Oncogene ; 26(13): 1897-909, 2007 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-17043659

RESUMEN

The retinoblastoma (pRB) family proteins regulate the E2F transcription factors; their complexes regulate critical transitions through the cell cycle. The function of these pRB family/E2F complexes, which includes p130/E2F4, in response to genotoxic agents, is not well understood. We investigated the role of E2F4 in the genotoxic stress response. Following radiation treatment, E2F4 colocalized with p130 in the nucleus during a radiation-induced stable G(2)-phase arrest. Arrested cells had significantly decreased expression of Cyclins A2 and B1 and decreased phosphorylation of mitotic protein monoclonal-2 (MPM-2) mitotic proteins. Small interference RNA (siRNA)-mediated knockdown of E2F4 sensitized cells to subsequent irradiation, resulting in enhanced cellular DNA damage and cell death, as determined by caspase activation and decreased clonogenic cell survival. Downstream E2F4 targets potentially involved in the progression from G(2) into M phase were identified by oligonucleotide microarray expression profiling. Chromatin immunoprecipitation localized E2F4 at promoter regions of the Bub3 and Pttg1 mitotic genes following irradiation, which were among the downregulated genes identified by the microarray. These data suggest that in response to radiation, E2F4 becomes active in the nucleus, enforces a stable G(2) arrest by target gene repression, and thus provides increased cell survival ability by minimizing propagation of cells that have irreparable DNA damage.


Asunto(s)
Factor de Transcripción E2F4/fisiología , Fase G2/efectos de los fármacos , Neoplasias de la Próstata/patología , Secuencia de Bases , División Celular/efectos de la radiación , Cartilla de ADN , Factor de Transcripción E2F4/genética , Citometría de Flujo , Humanos , Masculino , Neoplasias de la Próstata/metabolismo , ARN Interferente Pequeño , Radiación Ionizante
17.
Cell Cycle ; 5(21): 2436-9, 2006 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17102628

RESUMEN

The fundamental role of E2F transcription factors in the regulation of proliferation is well established. According to a widely accepted model, E2F1, E2F2, and E2F3 are classified as "activating" E2Fs since they induce proliferation of quiescent cells whereas E2F4 and E2F5 do not have the power to incite cell cycle progression but are related to differentiation processes and were therefore considered to be "repressive". In addition, it has been postulated that "activating" E2Fs induce apoptosis in a wide variety of cell types depending on their expression level. However, we demonstrated recently that this 'threshold model' does not hold true for cardiomyocytes. In a series of experiments in which we overexpressed individual E2Fs we found that directed expression of E2F2, unlike E2F1, E2F3 and E2F5, did not induce apoptosis but even suppressed expression of several pro-apoptotic genes in primary cardiomyocytes. Furthermore, we established that not only E2F1, E2F2, and E2F3 but also E2F4 was able to induce S-phase entry of primary cardiomyocytes. Our results suggest that it is possible to utilize the proliferation-inducing properties of the E2Fs in cardiomyocytes without activation of potentially harmful pro-apoptotic traits. This finding might open a new access to stimulate regeneration in postmitotic tissues such as the heart.


Asunto(s)
Factores de Transcripción E2F/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Animales , Apoptosis , Proliferación Celular , Factor de Transcripción E2F1/fisiología , Factor de Transcripción E2F2/fisiología , Factor de Transcripción E2F3/fisiología , Factor de Transcripción E2F4/fisiología , Factor de Transcripción E2F5/fisiología , Modelos Biológicos , Ratas , Regeneración , Fase S , Activación Transcripcional
18.
Blood ; 108(3): 886-95, 2006 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16861343

RESUMEN

The E2F proteins are major regulators of the transcriptional program required to coordinate cell cycle progression and exit. In particular, E2f4 has been proposed to be the principal family member responsible for the regulation of cell cycle exit chiefly through its transcriptional repressive properties. We have previously shown that E2f4(-/-) mice display a marked macrocytic anemia implicating E2f4 in the regulation of erythropoiesis. However, these studies could not distinguish whether E2f4 was required for differentiation, survival, or proliferation control. Here, we describe a novel function for E2f4 in the promotion of erythroid proliferation. We show that loss of E2f4 results in an impaired expansion of the fetal erythroid compartment in vivo that is associated with impaired cell cycle progression and decreased erythroid proliferation. Consistent with these observations, cDNA microarray analysis reveals cell cycle control genes as one of the major class of genes down-regulated in E2f4(-/-) FLs, and we provide evidence that E2f4 may directly regulate the transcriptional expression of a number of these genes. We conclude that the macrocytic anemia of E2f4(-/-) mice results primarily from impaired cellular proliferation and that the major role of E2f4 in fetal erythropoiesis is to promote cell cycle progression and cellular proliferation.


Asunto(s)
Proliferación Celular , Factor de Transcripción E2F4/fisiología , Células Eritroides/citología , Eritropoyesis , Anemia Macrocítica/etiología , Animales , Ciclo Celular , Factor de Transcripción E2F4/deficiencia , Embrión de Mamíferos , Perfilación de la Expresión Génica , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Transcripción Genética
19.
Diabetes ; 55(3): 640-50, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16505226

RESUMEN

To identify novel pathways mediating molecular mechanisms of thiazolidinediones (TZDs) in humans, we assessed gene expression in adipose and muscle tissue from six subjects with type 2 diabetes before and after 8 weeks of treatment with rosiglitazone. mRNA was analyzed using Total Gene Expression Analysis (TOGA), an automated restriction-based cDNA display method with quantitative analysis of PCR products. The expression of cell cycle regulatory transcription factors E2F4 and the MAGE protein necdin were similarly altered in all subjects after rosiglitazone treatment. E2F4 expression was decreased by 10-fold in muscle and 2.5-fold in adipose tissue; necdin was identified in adipose tissue only and increased 1.8-fold after TZD treatment. To determine whether changes were related to an effect of the drug or adipogenesis, we evaluated the impact of rosiglitazone and differentiation independently in 3T3-L1 adipocytes. While treatment of differentiated adipocytes with rosiglitazone did not alter E2F4 or necdin, expression of both genes was significantly altered during differentiation. Differentiation was associated with increased cytosolic localization of E2F4. Moreover, necdin overexpression potently inhibited adipocyte differentiation and cell cycle progression. These data suggest that changes in necdin and E2F4 expression after rosiglitazone exposure in humans are associated with altered adipocyte differentiation and may contribute to improved insulin sensitivity in humans treated with TZDs.


Asunto(s)
Adipocitos/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Factor de Transcripción E2F4/genética , Hipoglucemiantes/uso terapéutico , Músculos/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Tiazolidinedionas/uso terapéutico , Células 3T3-L1 , Adulto , Anciano , Animales , Diferenciación Celular , Diabetes Mellitus Tipo 2/metabolismo , Factor de Transcripción E2F4/fisiología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , Persona de Mediana Edad , Proteínas del Tejido Nervioso/fisiología , Proteínas Nucleares/fisiología , ARN Mensajero/análisis , Rosiglitazona
20.
Front Biosci ; 3: d11-24, 1998 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-9405335

RESUMEN

Pocket proteins, including the retinoblastoma susceptibility gene product (pRB) and the related proteins p107 and p130, function at cell cycle regulatory steps that link cyclin/CDK-integrated positive and negative growth signals with E2F transcription factor activity on genes required for cell cycle progression. Protein complex formation between pocket proteins and members of the E2F family of transcription factors determines whether E2F complexes act as transcriptional activators or repressors. Experimental work over the last few years indicates that individual pocket proteins interact with specific E2F members to regulate the transcription of certain genes under diverse cell growth conditions. Among these protein associations, p130-containing E2F complexes seem to be of particular importance in controlling gene transcription in quiescent and differentiating cells by repressing the transcription of a set of E2F-responsive genes. Once the cells are progressing through the G1 phase of the cell cycle, pocket protein-mediated regulation of E2F activity is assumed by pRB and p107. p130-mediated transcriptional regulation thus seems to prevent a gene expression program characteristic of dividing cells at the cell cycle exit and re-entrance transitions and in quiescent cells.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Factor de Transcripción E2F4/fisiología , Proteína p130 Similar a la del Retinoblastoma/fisiología , Animales , Diferenciación Celular/fisiología , Fase G1/fisiología , Regulación de la Expresión Génica/fisiología , Humanos , Fosforilación , Fase de Descanso del Ciclo Celular/fisiología , Proteína p107 Similar a la del Retinoblastoma/fisiología , Transcripción Genética/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...