Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Fundam Clin Pharmacol ; 35(1): 156-164, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32446293

RESUMO

Cervical cancer is the second most common malignancy in women, and the novel therapeutic treatment is needed. Abemaciclib is a FDA-approved drug for breast cancer treatment. In this work, we identified that abemaciclib has potent anti-cervical cancer activity. We demonstrate that abemaciclib is the most effective drug against human papillomavirus (HPV)-negative cervical cancer cells compared to ribociclib and palbociclib, with its IC50 at nanomolar concentration range. This is achieved by the inhibition of proliferation and induction of apoptosis, through specifically suppressing CDK4/6-Rb-E2F and mTOR pathways by abemaciclib in HPV-negative cervical cancer cells. Of note, the combination of abemaciclib with paclitaxel and cisplatin at sublethal concentration results in much greater efficacy than chemotherapy alone. In addition, we confirm the efficacy of abemaciclib and its combination with paclitaxel or cisplatin at the doses that are not toxic to mice in HPV-negative cervical cancer xenograft mouse model. Interestingly, we show that abemaciclib and other CDK4/6 inhibitors are not effective in targeting HPV-positive cervical cancer cells, and this is likely to be associated with the high p16 and low Rb expression in HPV-positive cervical cancer cells. Our work is the first to provide the preclinical evidence to demonstrate the potential of abemaciclib for the treatment of HPV-negative cervical cancer. The mechanism analysis highlights the therapeutic value of inhibiting CDK4/6 in HPV-negative but not HPV-positive cervical cancer.


Assuntos
Aminopiridinas/farmacologia , Benzimidazóis/farmacologia , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Fatores de Transcrição E2F/antagonistas & inibidores , Proteína do Retinoblastoma/antagonistas & inibidores , Serina-Treonina Quinases TOR/antagonistas & inibidores , Neoplasias do Colo do Útero/tratamento farmacológico , Alphapapillomavirus/isolamento & purificação , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Neoplasias do Colo do Útero/virologia , Ensaios Antitumorais Modelo de Xenoenxerto
2.
PLoS One ; 13(12): e0208110, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30540809

RESUMO

The propensity for differentiation varies substantially across human pluripotent stem cell (hPSC) lines, greatly restricting the use of hPSCs for cell replacement therapy or disease modeling. Here, we investigate the underlying mechanisms and demonstrate that activation of the retinoblastoma (Rb) pathway in a transient manner is important for differentiation. In prior work, we demonstrated that pre-treating hPSCs with dimethylsulfoxide (DMSO) before directed differentiation enhanced differentiation potential across all three germ layers. Here, we show that exposure to DMSO improves the efficiency of hPSC differentiation through Rb and by repressing downstream E2F-target genes. While transient inactivation of the Rb family members (including Rb, p107, and p130) suppresses DMSO's capacity to enhance differentiation across all germ layers, transient expression of a constitutively active (non-phosphorylatable) form of Rb increases the differentiation efficiency similar to DMSO. Inhibition of downstream targets of Rb, such as E2F signaling, also promotes differentiation of hPSCs. More generally, we demonstrate that the duration of Rb activation plays an important role in regulating differentiation capacity.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Dimetil Sulfóxido/farmacologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais/efeitos dos fármacos , Aminopiridinas/farmacologia , Técnicas de Cultura de Células , Diferenciação Celular/genética , Linhagem Celular , Fatores de Transcrição E2F/antagonistas & inibidores , Fatores de Transcrição E2F/metabolismo , Técnicas de Silenciamento de Genes , Camadas Germinativas/citologia , Camadas Germinativas/efeitos dos fármacos , Camadas Germinativas/fisiologia , Humanos , Hidroxiquinolinas/farmacologia , Células-Tronco Pluripotentes/fisiologia , Proteína do Retinoblastoma/genética , Proteína p107 Retinoblastoma-Like/genética , Proteína p107 Retinoblastoma-Like/metabolismo , Proteína p130 Retinoblastoma-Like/genética , Proteína p130 Retinoblastoma-Like/metabolismo , Transdução de Sinais/genética , Fatores de Tempo
3.
Sci Rep ; 8(1): 8330, 2018 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-29844366

RESUMO

The main goal of this study was to find out strategies of clinical relevance to classify patients with a pancreatic ductal adenocarcinoma (PDAC) for individualized treatments. In the present study a set of 55 patient-derived xenografts (PDX) were obtained and their transcriptome were analyzed by using an Affymetrix approach. A supervised bioinformatics-based analysis let us to classify these PDX in two main groups named E2F-highly dependent and E2F-lowly dependent. Afterwards their characterization by using a Kaplan-Meier analysis demonstrated that E2F high patients survived significantly less than E2F low patients (9.5 months vs. 16.8 months; p = 0.0066). Then we tried to establish if E2F transcriptional target levels were associated to the response to cytotoxic treatments by comparing the IC50 values of E2F high and E2F low cells after gemcitabine, 5-fluorouracil, oxaliplatin, docetaxel or irinotecan treatment, and no association was found. Then we identified an E2F inhibitor compound, named ly101-4B, and we observed that E2F-higly dependent cells were more sensitive to its treatment (IC50 of 19.4 ± 1.8 µM vs. 44.1 ± 4.4 µM; p = 0.0061). In conclusion, in this work we describe an E2F target expression-based classification that could be predictive for patient outcome, but more important, for the sensitivity of tumors to the E2F inhibitors as a treatment. Finally, we can assume that phenotypic characterization, essentially by an RNA expression analysis of the PDAC, can help to predict their clinical outcome and their response to some treatments when are rationally selected.


Assuntos
Carcinoma Ductal Pancreático/classificação , Fatores de Transcrição E2F/metabolismo , Neoplasias Pancreáticas/classificação , Animais , Antimetabólitos Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Fatores de Transcrição E2F/antagonistas & inibidores , Fatores de Transcrição E2F/genética , Feminino , Humanos , Estimativa de Kaplan-Meier , Masculino , Camundongos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Análise de Sobrevida , Transcriptoma , Ensaios Antitumorais Modelo de Xenoenxerto , Neoplasias Pancreáticas
4.
Mol Cancer Ther ; 17(5): 908-920, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29483206

RESUMO

The cyclin dependent kinase (CDK)-retinoblastoma (RB)-E2F pathway plays a critical role in the control of cell cycle in estrogen receptor-positive (ER+) breast cancer. Small-molecule inhibitors of CDK4/6 have shown promise in this tumor type in combination with hormonal therapies, reflecting the particular dependence of this subtype of cancer on cyclin D1 and E2F transcription factors. mTOR inhibitors have also shown potential in clinical trials in this disease setting. Recent data have suggested cooperation between the PI3K/mTOR pathway and CDK4/6 inhibition in preventing early adaptation and eliciting growth arrest, but the mechanisms of the interplay between these pathways have not been fully elucidated. Here we show that profound and durable inhibition of ER+ breast cancer growth is likely to require multiple hits on E2F-mediated transcription. We demonstrate that inhibition of mTORC1/2 does not affect ER function directly, but does cause a decrease in cyclin D1 protein, RB phosphorylation, and E2F-mediated transcription. Combination of an mTORC1/2 inhibitor with a CDK4/6 inhibitor results in more profound effects on E2F-dependent transcription, which translates into more durable growth arrest and a delay in the onset of resistance. Combined inhibition of mTORC1/2, CDK4/6, and ER delivers even more profound and durable regressions in breast cancer cell lines and xenografts. Furthermore, we show that CDK4/6 inhibitor-resistant cell lines reactivate the CDK-RB-E2F pathway, but remain sensitive to mTORC1/2 inhibition, suggesting that mTORC1/2 inhibitors may represent an option for patients that have relapsed on CDK4/6 therapy. Mol Cancer Ther; 17(5); 908-20. ©2018 AACR.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Fatores de Transcrição E2F/antagonistas & inibidores , Serina-Treonina Quinases TOR/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Benzamidas , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quinase 4 Dependente de Ciclina/metabolismo , Quinase 6 Dependente de Ciclina/metabolismo , Fatores de Transcrição E2F/metabolismo , Feminino , Humanos , Células MCF-7 , Camundongos SCID , Morfolinas/administração & dosagem , Piperazinas/administração & dosagem , Piridinas/administração & dosagem , Pirimidinas , Receptores de Estrogênio/metabolismo , Serina-Treonina Quinases TOR/metabolismo
5.
Sci Rep ; 7(1): 5198, 2017 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-28701722

RESUMO

Although ASXL1 mutations are frequently found in human diseases, including myeloid leukemia, the cell proliferation-associated function of ASXL1 is largely unknown. Here, we explored the molecular mechanism underlying the growth defect found in Asxl1-deficient mouse embryonic fibroblasts (MEFs). We found that Asxl1, through amino acids 371 to 655, interacts with the kinase domain of AKT1. In Asxl1-null MEFs, IGF-1 was unable to induce AKT1 phosphorylation and activation; p27Kip1, which forms a ternary complex with ASXL1 and AKT1, therefore remained unphosphorylated. Hypophosphorylated p27Kip1 is able to enter the nucleus, where it prevents the phosphorylation of Rb; this ultimately leads to the down-regulation of E2F target genes as confirmed by microarray analysis. We also found that senescence-associated (SA) genes were upregulated and that SA ß-gal staining was increased in Asxl1 -/- MEFs. Further, the treatment of an AKT inhibitor not only stimulated nuclear accumulation of p27Kip1 leading to E2F inactivation, but also promoted senescence. Finally, Asxl1 disruption augmented the expression of p16Ink4a as result of the defect in Asxl1-Ezh2 cooperation. Overall, our study provides the first evidence that Asxl1 both activates the AKT-E2F pathway and cooperates with Ezh2 through direct interactions at early embryonic stages, reflecting that Asxl1 disruption causes cellular senescence.


Assuntos
Senescência Celular , Fatores de Transcrição E2F/antagonistas & inibidores , Embrião de Mamíferos/patologia , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Fibroblastos/patologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Repressoras/fisiologia , Animais , Proliferação de Células , Células Cultivadas , Fatores de Transcrição E2F/genética , Fatores de Transcrição E2F/metabolismo , Embrião de Mamíferos/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Fibroblastos/metabolismo , Camundongos , Camundongos Knockout , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
6.
G3 (Bethesda) ; 6(3): 709-20, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26772748

RESUMO

Centrioles play critical roles in the organization of microtubule-based structures, from the mitotic spindle to cilia and flagella. In order to properly execute their various functions, centrioles are subjected to stringent copy number control. Central to this control mechanism is a precise duplication event that takes place during S phase of the cell cycle and involves the assembly of a single daughter centriole in association with each mother centriole . Recent studies have revealed that posttranslational control of the master regulator Plk4/ZYG-1 kinase and its downstream effector SAS-6 is key to ensuring production of a single daughter centriole. In contrast, relatively little is known about how centriole duplication is regulated at a transcriptional level. Here we show that the transcription factor complex EFL-1-DPL-1 both positively and negatively controls centriole duplication in the Caenorhabditis elegans embryo. Specifically, we find that down regulation of EFL-1-DPL-1 can restore centriole duplication in a zyg-1 hypomorphic mutant and that suppression of the zyg-1 mutant phenotype is accompanied by an increase in SAS-6 protein levels. Further, we find evidence that EFL-1-DPL-1 promotes the transcription of zyg-1 and other centriole duplication genes. Our results provide evidence that in a single tissue type, EFL-1-DPL-1 sets the balance between positive and negative regulators of centriole assembly and thus may be part of a homeostatic mechanism that governs centriole assembly.


Assuntos
Caenorhabditis elegans/metabolismo , Centríolos/metabolismo , Fatores de Transcrição E2F/metabolismo , Complexos Multiproteicos/metabolismo , Fator de Transcrição DP1/metabolismo , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Divisão Celular/genética , Fatores de Transcrição E2F/antagonistas & inibidores , Regulação da Expressão Gênica , Genes Letais , Genoma Helmíntico , Mutação , Fator de Transcrição DP1/antagonistas & inibidores , Transcrição Gênica
7.
Ann Thorac Surg ; 101(6): 2407-16, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26801060

RESUMO

The concept of gene therapy was introduced in the 1970s after the development of recombinant DNA technology. Despite the initial great expectations, this field experienced early setbacks. Recent years have seen a revival of clinical programs of gene therapy in different fields of medicine. There are many promising targets for genetic therapy as an adjunct to cardiac surgery. The first positive long-term results were published for adenoviral administration of vascular endothelial growth factor with coronary artery bypass grafting. In this review we analyze the past, present, and future of gene therapy in cardiac surgery. The articles discussed were collected through PubMed and from author experience. The clinical trials referenced were found through the Wiley clinical trial database (http://www.wiley.com/legacy/wileychi/genmed/clinical/) as well as the National Institutes of Health clinical trial database (Clinicaltrials.gov).


Assuntos
Procedimentos Cirúrgicos Cardíacos , Ensaios Clínicos como Assunto , Terapia Genética , Proteínas Angiogênicas/genética , Animais , Terapia Combinada , Ponte de Artéria Coronária , Fatores de Transcrição E2F/antagonistas & inibidores , Determinação de Ponto Final , Vetores Genéticos/administração & dosagem , Vetores Genéticos/uso terapêutico , Oclusão de Enxerto Vascular/prevenção & controle , Insuficiência Cardíaca/cirurgia , Insuficiência Cardíaca/terapia , Transplante de Coração , Humanos , Injeções , Estudos Multicêntricos como Assunto , Miocárdio , Neovascularização Fisiológica/genética , Seleção de Pacientes , Resultado do Tratamento
8.
J Cell Biochem ; 115(10): 1779-86, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24820054

RESUMO

In several skeletal dysplasias defects in extracellular matrix molecules affect not only the structural and mechanical properties of cartilage, but also the complex network of signaling pathways involved in cell proliferation and differentiation. Sulfated proteoglycans, besides playing an important structural role in cartilage, are crucial in modulating the transport, diffusion, and interactions of growth factors with their specific targets, taking part in the regulation of signaling pathways involved in skeletal development and growth. In this work, we investigated by real time PCR and Western blots of the microdissected growth plate and by immunohistochemistry the molecular basis of reduced chondrocyte proliferation in the growth plate of the dtd mouse, a chondrodysplastic model with defective chondroitin sulfate proteoglycan sulfation of articular and growth plate cartilage. We detected activation of the Wnt pathway, leading to an increase in the non-phosphorylated form of nuclear ß-catenin and subsequent up-regulation of cyclin D1 expression in the G1 phase of the cell cycle. ß-Catenin was further stabilized by up-regulation of Smad3 expression through TGF-ß pathway synergistic activation. We demonstrate that notwithstanding cyclin D1 expression increase, cell cycle progression is compromised in the G1 phase due to reduced phosphorylation of the pocket protein p130 leading to inhibition of transcription factors of the E2F family which are crucial for cell cycle progression and DNA replication. These data, together with altered Indian hedgehox signaling detected previously, explain at the molecular level the reduced chondrocyte proliferation rate of the dtd growth plate leading to reduced skeletal growth.


Assuntos
Desenvolvimento Ósseo/genética , Condrócitos/metabolismo , Ciclina D1/biossíntese , Fatores de Transcrição E2F/antagonistas & inibidores , Proteína p130 Retinoblastoma-Like/metabolismo , Animais , Doenças Ósseas/genética , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Diferenciação Celular/genética , Proliferação de Células/genética , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Matriz Extracelular/patologia , Fase G1/genética , Técnicas de Introdução de Genes , Lâmina de Crescimento/metabolismo , Proteínas Hedgehog/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Transdução de Sinais/genética , Proteína Smad3/biossíntese , Fator de Crescimento Transformador beta/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo
9.
Mol Biol Cell ; 25(8): 1374-83, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24554762

RESUMO

Geminin performs a central function in regulating cellular proliferation and differentiation in development and also in stem cells. Of interest, down-regulation of Geminin induces gene transcription regulated by E2F, indicating that Geminin is involved in regulation of E2F-mediated transcriptional activity. Because transcription of the Geminin gene is reportedly regulated via an E2F-responsive region (E2F-R) located in the first intron, we first used a reporter vector to examine the effect of Geminin on E2F-mediated transcriptional regulation. We found that Geminin transfection suppressed E2F1- and E2F2-mediated transcriptional activation and also mildly suppressed such activity in synergy with E2F5, 6, and 7, suggesting that Geminin constitutes a negative-feedback loop for the Geminin promoter. Of interest, Geminin also suppressed nuclease accessibility, acetylation of histone H3, and trimethylation of histone H3 at lysine 4, which were induced by E2F1 overexpression, and enhanced tri-methylation of histone H3 at lysine 27 and monoubiquitination of histone H2A at lysine 119 in E2F-R. However, Geminin5EQ, which does not interact with Brahma or Brg1, did not suppress accessibility to nuclease digestion or transcription but had an overall dominant-negative effect. These findings suggest that E2F-mediated activation of Geminin transcription is negatively regulated by Geminin through the inhibition of chromatin remodeling.


Assuntos
Fatores de Transcrição E2F/genética , Retroalimentação Fisiológica , Geminina/genética , Ativação Transcricional/genética , Células 3T3 , Acetilação , Animais , Anticorpos/imunologia , Ciclo Celular/genética , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Montagem e Desmontagem da Cromatina/genética , DNA Helicases , Proteínas de Ligação a DNA , Fatores de Transcrição E2F/antagonistas & inibidores , Fatores de Transcrição E2F/biossíntese , Geminina/biossíntese , Geminina/metabolismo , Regulação da Expressão Gênica , Células HEK293 , Histonas/imunologia , Histonas/metabolismo , Humanos , Metilação , Camundongos , Proteínas Nucleares , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Interferência de RNA , RNA Interferente Pequeno , Células-Tronco/metabolismo , Fatores de Transcrição , Ubiquitinação
10.
J Mol Biol ; 426(1): 245-55, 2014 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-24103329

RESUMO

The retinoblastoma protein C-terminal domain (RbC) is necessary for the tumor suppressor protein's activities in growth suppression and E2F transcription factor inhibition. Cyclin-dependent kinase phosphorylation of RbC contributes to Rb inactivation and weakens the Rb-E2F inhibitory complex. Here we demonstrate two mechanisms for how RbC phosphorylation inhibits E2F binding. We find that phosphorylation of S788 and S795 weakens the direct association between the N-terminal portion of RbC (RbC(N)) and the marked-box domains of E2F and its heterodimerization partner DP. Phosphorylation of these sites and S807/S811 also induces an intramolecular association between RbC and the pocket domain, which overlaps with the site of E2F transactivation domain binding. A reduction in E2F binding affinity occurs with S788/S795 phosphorylation that is additive with the effects of phosphorylation at other sites, and we propose a structural mechanism that explains this additivity. We find that different Rb phosphorylation events have distinct effects on activating E2F family members, which suggests a novel mechanism for how Rb may differentially regulate E2F activities.


Assuntos
Fatores de Transcrição E2F/antagonistas & inibidores , Fatores de Transcrição E2F/metabolismo , Proteína do Retinoblastoma/metabolismo , Humanos , Modelos Biológicos , Modelos Moleculares , Fosforilação , Ligação Proteica , Conformação Proteica
11.
Plant Physiol ; 157(3): 1440-51, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21908689

RESUMO

Endoreduplication represents a variation on the cell cycle in which multiple rounds of DNA replication occur without subsequent chromosome separation and cytokinesis, thereby increasing the cellular DNA content. It is known that the DNA ploidy level of cells is controlled by external stimuli such as light; however, limited knowledge is available on how environmental signals regulate the endoreduplication cycle at the molecular level. Previously, we had demonstrated that the conversion from a mitotic cell cycle into an endoreduplication cycle is controlled by the atypical E2F transcription factor, DP-E2F-LIKE1 (DEL1), that represses the endocycle onset. Here, the Arabidopsis (Arabidopsis thaliana) DEL1 gene was identified as a transcriptional target of the classical E2Fb and E2Fc transcription factors that antagonistically control its transcript levels through competition for a single E2F cis-acting binding site. In accordance with the reported opposite effects of light on the protein levels of E2Fb and E2Fc, DEL1 transcription depended on the light regime. Strikingly, modified DEL1 expression levels uncoupled the link between light and endoreduplication in hypocotyls, implying that DEL1 acts as a regulatory connection between endocycle control and the photomorphogenic response.


Assuntos
Proteínas de Arabidopsis/antagonistas & inibidores , Arabidopsis/genética , Arabidopsis/efeitos da radiação , Fatores de Transcrição E2F/antagonistas & inibidores , Regulação da Expressão Gênica de Plantas/efeitos da radiação , Luz , Fatores de Transcrição/genética , Proteínas de Arabidopsis/metabolismo , Sequência de Bases , Fatores de Transcrição E2F/metabolismo , Hipocótilo/genética , Hipocótilo/efeitos da radiação , Modelos Biológicos , Dados de Sequência Molecular , Mutação/genética , Plantas Geneticamente Modificadas , Ploidias , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos da radiação , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
12.
Biochim Biophys Acta ; 1799(10-12): 788-94, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20637913

RESUMO

The retinoblastoma tumor suppressor protein, Rb, plays a major role in the regulation of mammalian cell cycle progression. It has been shown that Rb function is essential for the proper modulation of G1/S transition and inactivation of Rb contributes to deregulated cell proliferation. Rb exerts its cell cycle regulatory functions mainly by targeting the E2F family of transcription factors and Rb has been shown to physically interact with E2Fs 1, 2 and 3, repressing their transcriptional activity. Multiple genes involved in DNA synthesis and cell cycle progression are regulated by E2Fs, and Rb prevents their expression by inhibiting E2F activity, inducing growth arrest. It has been established that inactivation of Rb by phosphorylation, mutation, or by the interaction of viral oncoproteins leads to a release of the repression of E2F activity, facilitating cell cycle progression. Rb-mediated repression of E2F activity involves the recruitment of a variety of transcriptional co-repressors and chromatin remodeling proteins, including histone deacetylases, DNA methyltransferases and Brg1/Brm chromatin remodeling proteins. Inactivation of Rb by sequential phosphorylation events during cell cycle progression leads to a dissociation of these co-repressors from Rb, facilitating transcription. It has been found that small molecules that prevent the phosphorylation of Rb prevent the dissociation of certain co-repressors from Rb, especially Brg1, leading to the maintenance of Rb-mediated transcriptional repression and cell cycle arrest. Such small molecules have anti-cancer activities and will also act as valuable probes to study chromatin remodeling and transcriptional regulation.


Assuntos
Antineoplásicos , Montagem e Desmontagem da Cromatina/efeitos dos fármacos , Fatores de Transcrição E2F , Proteína do Retinoblastoma , Transcrição Gênica/efeitos dos fármacos , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Ciclo Celular/efeitos dos fármacos , DNA/biossíntese , Metilases de Modificação do DNA/metabolismo , Fatores de Transcrição E2F/antagonistas & inibidores , Fatores de Transcrição E2F/metabolismo , Humanos , Fosforilação/efeitos dos fármacos , Proteína do Retinoblastoma/antagonistas & inibidores , Proteína do Retinoblastoma/metabolismo , Fatores de Transcrição/metabolismo
13.
Nucleic Acids Res ; 38(13): e140, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20453028

RESUMO

MicroRNAs (miRNAs) are naturally occurring small RNAs that regulate the expression of several genes. MiRNAs' targeting rules are based on sequence complementarity between their mature products and targeted genes' mRNAs. Based on our present understanding of those rules, we developed an algorithm to design artificial miRNAs to target simultaneously a set of predetermined genes. To validate in silico our algorithm, we tested different sets of genes known to be targeted by a single miRNA. The algorithm finds the seed of the corresponding miRNA among the solutions, which also include the seeds of new artificial miRNA sequences potentially capable of targeting these genes as well. We also validated the functionality of some artificial miRNAs designed to target simultaneously members of the E2F family. These artificial miRNAs reproduced the effects of E2Fs inhibition in both normal human fibroblasts and prostate cancer cells where they inhibited cell proliferation and induced cellular senescence. We conclude that the current miRNA targeting rules based on the seed sequence work to design multiple-target artificial miRNAs. This approach may find applications in both research and therapeutics.


Assuntos
Algoritmos , Regulação da Expressão Gênica , MicroRNAs/química , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Senescência Celular , Fatores de Transcrição E2F/antagonistas & inibidores , Humanos , MicroRNAs/metabolismo
14.
Cell Cycle ; 9(8): 1590-600, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20372067

RESUMO

Abnormalities in the p16INK4a/ cyclin-dependent kinase (Cdk)4, 6/ Retinoblastoma (Rb) pathway frequently occur in various human cancers. Thus, Cdk4/6 is an attractive target for cancer therapy. Here we report the biological characterization of a 2-aminothiazole-derived Cdk4/6 selective inhibitor, named Compound A in vitro and in vivo. Compound A potently inhibits Cdk4 and Cdk6 with high selectivity (more than 57-fold) against other Cdks and 45 serine/threonine and tyrosine kinases. Compound A inhibits Rb protein (pRb) phosphorylation at Ser780, inhibits E2F-dependent transcription, and induces cell-cycle arrest at G1 in the T98G human glioma cell line. Among 82 human cells derived from various tissues, cell lines derived from hematological cancers (leukemia/lymphoma) tended to be more sensitive to Compound A in cell proliferation assay. Rb-negative cells tended to be insensitive to Compound A, as we had expected. In a nude rat xenograft model, Compound A inhibited pRb phosphorylation and bromodeoxyuridine (BrdU) incorporation in Eol-1 xenograft tumor at plasma concentration of 510 nM. Interestingly Compound A only moderately inhibited those pharmacodynamic and cell cycle parameters of normal crypt cells in small intestine even at 5 times higher plasma concentration. In F344 rats, Compound A did not cause immunosuppression even at 17 times higher plasma conc. These results suggest that Cdk4/6 selective inhibitors only moderately affects on the cell cycle of normal proliferating tissues and has a safer profile than pan-Cdk inhibitor in vivo.


Assuntos
Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Tiazóis/farmacologia , Animais , Linhagem Celular Tumoral , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Quinase 2 Dependente de Ciclina/genética , Quinase 2 Dependente de Ciclina/metabolismo , Quinase 4 Dependente de Ciclina/metabolismo , Quinase 6 Dependente de Ciclina/metabolismo , Fatores de Transcrição E2F/antagonistas & inibidores , Fatores de Transcrição E2F/metabolismo , Fase G1 , Humanos , Masculino , Fosforilação , Inibidores de Proteínas Quinases/química , Pirimidinas/química , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Nus , Proteína do Retinoblastoma/antagonistas & inibidores , Proteína do Retinoblastoma/metabolismo , Tiazóis/química , Transplante Heterólogo
16.
J Cell Biochem ; 109(4): 693-701, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-20082327

RESUMO

Primarily, E2F factors such as E2F1, -2, and -3 stimulate cell-cycle progression, while ARF tumor suppressor mediates growth suppression. The ARF gene can be induced by oncogenic signal through activating E2F-dependent transcription. In turn, ARF may target E2F for its degradation via a p53-dependent mechanism. However, it remains unclear how the cell keeps the balance between the functional opposites of E2F and ARF. In this study, we demonstrate that p14ARF interacts with E2F1-3 factors to directly repress their transcriptional activities through forming p14ARF-E2F/partner-DNA super complexes, regardless of E2F protein degradation. The inhibition of E2F transcriptional activities by p14ARF in this manner occurs commonly in a variety of cell types, including p53-deficient and p53-wild type cells. Thus, E2F-mediated activation of the ARF gene and ARF-mediated functional inhibition of E2F compose a feedback loop, by which the two opposites act in concert to regulate cell proliferation and apoptosis, depending on the cellular context and the environment.


Assuntos
DNA/metabolismo , Fatores de Transcrição E2F/antagonistas & inibidores , Transcrição Gênica , Proteína Supressora de Tumor p14ARF/metabolismo , Apoptose , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Fatores de Transcrição E2F/genética , Fatores de Transcrição E2F/metabolismo , Retroalimentação Fisiológica , Humanos , Proteína Supressora de Tumor p53
17.
Cancer Res ; 68(15): 6292-9, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18676853

RESUMO

HLM006474 was identified using a computer-based virtual screen and the known crystal structure of the DNA-bound E2F4/DP2 heterodimer. Treatment of multiple cell lines with HLM006474 resulted in the loss of intracellular E2F4 DNA-binding activity as measured by electrophoretic mobility shift assay within hours. Overnight exposure to HLM006474 resulted in down-regulation of total E2F4 protein as well as known E2F targets. The effects of HLM006474 treatment on different cell lines varied but included a reduction in cell proliferation and an increase in apoptosis. HLM006474 induced apoptosis in a manner distinct from cisplatin and doxorubicin. E2F4-null mouse embryonic fibroblasts were less sensitive than wild-type counterparts to the apoptosis-inducing activity of the compound, revealing its biological specificity. A375 cells were extremely sensitive to the apoptosis-inducing activity of the compound in two-dimensional culture, and HLM006474 was a potent inhibitor of melanocytes proliferation and subsequent invasion in a three-dimensional tissue culture model system. Together, these results suggest that interference with E2F activity using small molecules may have clinical application in cancer therapy.


Assuntos
Fatores de Transcrição E2F/antagonistas & inibidores , Melanoma Experimental/patologia , Aminopiridinas/farmacologia , Animais , Antineoplásicos/farmacologia , Apoptose , Ciclo Celular , Linhagem Celular Tumoral , Hidroxiquinolinas/farmacologia , Camundongos
18.
Oncogene ; 27(43): 5696-705, 2008 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-18542057

RESUMO

E2Fs are key regulators of cell-cycle progression, and their transcriptional activities are regulated by histone acetyltransferases (HATs). Retinoblastoma (Rb) family proteins (pRb, p107 and p130) bind to E2Fs and inhibit their transcriptional activities by disrupting HAT binding and recruitment of histone deacetylases. In this study, we show that IkappaB kinases (IKKalpha or IKKbeta) activation inhibits cell growth and E2F-dependent transcription in normal human fibroblasts. The inhibition of E2F by IKKs was not observed in cells lacking nuclear factor (NF)-kappaB/p65; however, it was observed in cells lacking three Rb family genes. p65 disrupted the physical interaction between activator E2Fs (F2F1, E2F2 and E2F3) and the HAT cofactor transactivation/transformation-domain associated protein, resulting in a reduction in E2F-responsive gene expression. Furthermore, IKKalpha and IKKbeta directly phosphorylated E2F4, resulting in nuclear accumulation and enhanced DNA binding of the E2F4/p130 repressor complex. Our study describes a novel growth inhibitory system that functions by Rb-independent suppression of E2Fs by the IKK/NF-kappaB signaling pathway.


Assuntos
Ciclo Celular , Fatores de Transcrição E2F/antagonistas & inibidores , Quinase I-kappa B/fisiologia , NF-kappa B/fisiologia , Proteína do Retinoblastoma/fisiologia , Transdução de Sinais/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Núcleo Celular/metabolismo , DNA/metabolismo , Histona Acetiltransferases , Humanos , Camundongos , Células NIH 3T3 , Proteínas Nucleares/metabolismo , Fosforilação , Regiões Promotoras Genéticas , Fator de Transcrição RelA/fisiologia , Fator de Necrose Tumoral alfa/farmacologia
19.
Cardiovasc Drug Rev ; 25(3): 221-34, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17919257

RESUMO

The era of genomics and recombinant DNA technology has ushered in an entirely new class of therapeutic agents designed to influence disease progression at a genetic level. The scope and utility of this technology is not fully realized. However, multiple trials of therapeutic agents have been completed and many more are ongoing. Here we report on edifoligide, a double-stranded oligodeoxynucleotide (ODN) that competitively inhibits the transcription factor E2F, a critical regulator of the cell cycle. Edifoligide has undergone extensive clinical testing for the treatment of intimal hyperplasia following vascular bypass procedures. In this review we address the rationale for targeting E2F in vascular disease, the pharmacology of edifoligide, and the results of preclinical and clinical studies using this novel compound.


Assuntos
Proliferação de Células/efeitos dos fármacos , Oclusão de Enxerto Vascular/prevenção & controle , Músculo Liso Vascular/efeitos dos fármacos , Oligonucleotídeos/farmacologia , Animais , Ponte de Artéria Coronária , Fatores de Transcrição E2F/antagonistas & inibidores , Fatores de Transcrição E2F/genética , Humanos , Modelos Biológicos , Músculo Liso Vascular/citologia , Oligonucleotídeos/genética
20.
Proc Natl Acad Sci U S A ; 104(32): 12988-93, 2007 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-17652516

RESUMO

Intimal hyperplasia (IH) and restenosis limit the long-term utility of bypass surgery and angioplasty due to pathological proliferation and migration of vascular smooth muscle cells (VSMCs) into the intima of treated vessels. Consequently, much attention has been focused on developing inhibitory agents that reduce this pathogenic process. The E2F transcription factors are key cell cycle regulators that play important roles in modulating cell proliferation and cell fate. Nonselective E2F inhibitors have thus been extensively evaluated for this purpose. Surprisingly, these E2F inhibitors have failed to reduce IH. These findings prompted us to evaluate the roles of different E2Fs during IH to determine how selective targeting of E2F isoforms impacts VSMC proliferation. Importantly, we show that E2F3 promotes proliferation of VSMCs leading to increased IH, whereas E2F4 inhibits this pathological response. Furthermore, we use RNA probes to show that selective inhibition of E2F3, not global inhibition of E2F activity, significantly reduces VSMC proliferation and limits IH in murine bypass grafts.


Assuntos
Fatores de Transcrição E2F/fisiologia , Músculo Liso Vascular/patologia , Túnica Íntima/patologia , Animais , Aptâmeros de Nucleotídeos/farmacologia , Proliferação de Células , Células Cultivadas , Fatores de Transcrição E2F/antagonistas & inibidores , Hiperplasia , Camundongos , RNA Interferente Pequeno/farmacologia , Veia Cava Inferior/transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA