Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2445: 139-169, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34972991

RESUMO

Anticancer therapy is complicated by the ability of malignant cells to activate cytoprotective autophagy that rescues treated cells. This protocol describes methods for analysis of autophagic process in apoptosis-resistant tumor cells treated with damaging agents. Induction of autophagy in these cells can activate apoptotic death. Protocol provides methods for Western blotting, immunofluorescent analysis, and transfection of cells with fluorescent protein-tagged LC3-encoding plasmids to analyze autophagy. Different approaches to change autophagy in tumor cells are suggested. A special approach is connected with induction of cellular senescence. Senescent cells, which are resistant to apoptosis, are vulnerable to certain damaging agents, in particular, to kinase inhibitors. Methods to induce and analyze senescence are considered. They include detection of proliferation arrest by different ways, mTORC1 activity assay and fluorescent analysis of mTORC1 and lysosome localization as a novel senescence hallmark. Incapability of senescent cells to complete autophagy after damage allows to force them to apoptosis. To demonstrate apoptotic cell death, analysis of caspase activity, Annexin V-FITC binding, DNA fragmentation, and mitochondria and lysosome damage are suggested. The methods described can be applied in studies aimed on developing different strategies of tumor cell elimination through changing autophagy.


Assuntos
Apoptose , Autofagia , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular Tumoral , Senescência Celular , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo
2.
Int J Mol Sci ; 22(9)2021 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-33925224

RESUMO

BACKGROUND: Cancer stem cells' (CSCs) self-maintenance is regulated via the pluripotency pathways promoting the most aggressive tumor phenotype. This study aimed to use the activity of these pathways for the CSCs' subpopulation enrichment and separating cells characterized by the OCT4 and SOX2 expression. METHODS: To select and analyze CSCs, we used the SORE6x lentiviral reporter plasmid for viral transduction of colon adenocarcinoma cells. Additionally, we assessed cell chemoresistance, clonogenic, invasive and migratory activity and the data of mRNA-seq and intrinsic disorder predisposition protein analysis (IDPPA). RESULTS: We obtained the line of CSC-like cells selected on the basis of the expression of the OCT4 and SOX2 stem cell factors. The enriched CSC-like subpopulation had increased chemoresistance as well as clonogenic and migration activities. The bioinformatic analysis of mRNA seq data identified the up-regulation of pluripotency, development, drug resistance and phototransduction pathways, and the downregulation of pathways related to proliferation, cell cycle, aging, and differentiation. IDPPA indicated that CSC-like cells are predisposed to increased intrinsic protein disorder. CONCLUSION: The use of the SORE6x reporter construct for CSCs enrichment allows us to obtain CSC-like population that can be used as a model to search for the new prognostic factors and potential therapeutic targets for colon cancer treatment.


Assuntos
Adenocarcinoma/patologia , Neoplasias do Colo/patologia , Células-Tronco Neoplásicas/metabolismo , Adenocarcinoma/genética , Adulto , Biomarcadores Tumorais/isolamento & purificação , Técnicas de Cultura de Células/métodos , Ciclo Celular , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Neoplasias do Colo/genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Masculino , Pessoa de Meia-Idade , Células-Tronco Neoplásicas/patologia , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Cultura Primária de Células , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo
4.
Cell Death Discov ; 5: 61, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30729040

RESUMO

Resveratrol, a natural polyphenolic compound, shows many beneficial effects in various animal models. It increases efficiency of somatic cell reprograming into iPSCs and contributes to cell differentiation. Here, we studied the effect of resveratrol on proliferation and pluripotency of mouse embryonic stem cells (mESCs). Our results demonstrate that resveratrol induces autophagy in mESCs that is provided by the activation of the AMPK/Ulk1 pathway and the concomitant suppression of the activity of the mTORC1 signaling cascade. These events correlate with the enhanced expression of pluripotency markers Oct3/4, Sox2, Nanog, Klf4, SSEA-1 and alkaline phosphatase. Pluripotency is retained under resveratrol-caused retardation of cell proliferation. Given that the Ulk1 overexpression enhances pluripotency of mESCs, the available data evidence that mTOR/Ulk1/AMPK-autophagy network provides the resveratrol-mediated regulation of mESC pluripotency. The capability of resveratrol to support the mESC pluripotency provides a new approach for developing a defined medium for ESC culturing as well as for better understanding signaling events that govern self-renewal and pluripotency.

5.
Biochem Biophys Res Commun ; 503(3): 2180-2185, 2018 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-30098786

RESUMO

Resveratrol is a natural polyphenol with several therapeutic effects, in particular, inducing p53-dependent cell cycle arrest and/or apoptosis in tumor cells. Resveratrol-induced p53 activation may trigger differentiation and apoptosis in embryonic stem cells (ESCs). We show that resveratrol activates p53 that is negatively regulated by SIRT1 deacetylation on Lys379 and positively by AMPK phosphorylation on Ser15 in mouse ESCs (mESCs). Surprisingly, the resveratrol-activated p53 is not associated with either G1/S cell cycle checkpoint or apoptosis in mESCs. Instead, it stimulates autophagy in a transcriptional-dependent manner involving up-regulation of dram1 gene expression. This study demonstrates a novel mechanism of resveratrol-dependent p53 activation in mESCs.


Assuntos
Autofagia/efeitos dos fármacos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Resveratrol/farmacologia , Proteína Supressora de Tumor p53/agonistas , Animais , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Células Cultivadas , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima/efeitos dos fármacos
6.
Aging (Albany NY) ; 9(11): 2352-2375, 2017 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-29140794

RESUMO

The Ras-Raf-MEK-ERK pathway plays a central role in tumorigenesis and is a target for anticancer therapy. The successful strategy based on the activation of cell death in Ras-expressing cells is associated with the suppression of kinases involved in Ras pathway. However, activation of cytoprotective autophagy overcomes antiproliferative effect of the inhibitors and develops drug resistance. We studied whether cellular senescence induced by HDAC inhibitor sodium butyrate in E1a+cHa-Ras-transformed rat embryo fibroblasts (ERas) and A549 human Ki-Ras mutated lung adenocarcinoma cells would enhance the tumor suppressor effect of MEK/ERK inhibition. Treatment of control ERas cells with PD0325901 for 24 h results in mitochondria damage and apoptotic death of a part of cellular population. However, the activation of AMPK-dependent autophagy overcomes pro-apoptotic effects of MEK/ERK inhibitor and results in restoration of the mitochondria and rescue of viability. Senescent ERas cells do not develop cytoprotective autophagy upon inhibition of MEK/ERK pathway due to spatial dissociation of lysosomes and autophagosomes in the senescent cells. Senescent cells are unable to form the autophagolysosomes and to remove the damaged mitochondria resulting in apoptotic death. Our data show that suppression of MEK/ERK pathway in senescent cells provides a new strategy for elimination of Ras-expressing cells.


Assuntos
Adenocarcinoma/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Senescência Celular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibroblastos/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , MAP Quinase Quinase Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Células A549 , Proteínas Quinases Ativadas por AMP/metabolismo , Adenocarcinoma/enzimologia , Adenocarcinoma/genética , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Fibroblastos/enzimologia , Fibroblastos/patologia , Humanos , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , MAP Quinase Quinase Quinases/antagonistas & inibidores , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Ratos , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
7.
Cell Cycle ; 15(1): 52-63, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26636245

RESUMO

Mouse embryonic stem cells (mESCs) lack of G1 checkpoint despite that irradiation (IR) activates ATM/ATR-mediated DDR signaling pathway. The IR-induced p53 localizes in the nuclei and up-regulates p21/Waf1 transcription but that does not lead to accumulation of p21/Waf1 protein. The negative control of the p21Waf1 expression appears to occur at 2 levels of regulation. First, both p21/Waf1 gene transcription and the p21/Waf1 protein content increase in mESCs treated with histone-deacetylase inhibitors, implying its epigenetic regulation. Second, proteasome inhibitors cause the p21/Waf1 accumulation, indicating that the protein is a subject of proteasome-dependent degradation in ESСs. Then, the dynamics of IR-induced p21Waf1 protein show its accumulation at long-term time points (3 and 5 days) that coincides with an increase in the proportion of G1-phase cells, down-regulation of Oct4 and Nanog pluripotent gene transcription and activation of endoderm-specific genes sox17 and afp. In addition, nutlin-dependent stabilization of p53 in mESC was also accompanied by the accumulation of p21/Waf1 as well as restoration of G1 checkpoint and an onset of differentiation. Thus, the lack of functional p21/Waf1 is indispensable for maintaining self-renewal and pluripotency of mESCs.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Células-Tronco Embrionárias/metabolismo , Pontos de Checagem da Fase G1 do Ciclo Celular/fisiologia , Transdução de Sinais/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Sobrevivência Celular/fisiologia , Camundongos , Células NIH 3T3
8.
Oncotarget ; 6(42): 44905-26, 2015 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-26636543

RESUMO

mTOR is a critical target for controlling cell cycle progression, senescence and cell death in mammalian cancer cells. Here we studied the role of mTOR-dependent autophagy in implementating the antiprolifrative effect of mTORC1-specific inhibitor rapamycin and ATP-competitive mTOR kinase inhibitor pp242. We carried out a comprehensive analysis of pp242- and rapamycin-induced autophagy in ERas tumor cells. Rapamycin exerts cytostatic effect on ERas tumor cells, thus causing a temporary and reversible cell cycle arrest, activation of non-selective autophagy not accompanied by cell death. The rapamycin-treated cells are able to continue proliferation after drug removal. The ATP-competitive mTORC1/mTORC2 kinase inhibitor pp242 is highly cytotoxic by suppressing the function of mTORC1-4EBP1 axis and mTORC1-dependent phosphorylation of mTORC1 target--ULK1-Ser757 (Atg1). In contrast to rapamycin, pp242 activates the selective autophagy targeting mitochondria (mitophagy). The pp242-induced mitophagy is accompanied by accumulation of LC3 and conversion of LC3-I form to LC3-II. However reduced degradation of p62/SQSTM indicates abnormal flux of autophagic process. According to transmission electron microscopy data, short-term pp242-treated ERas cells exhibit numerous heavily damaged mitochondria, which are included in single membrane-bound autophagic/autolysophagic vacuoles (mitophagy). Despite the lack of typical for apoptosis features, ERas-treated cells with induced mitophagy revealed the activation of caspase 3, 9 and nucleosomal DNA fragmentation. Thus, pp242 activates autophagy with suppressed later stages, leading to impaired recycling and accumulation of dysfunctional mitochondria and cell death. Better understanding of how autophagy determines the fate of a cell--survival or cell death, can help to development of new strategy for cancer therapy.


Assuntos
Proteínas E1A de Adenovirus/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Indóis/farmacologia , Mitocôndrias/efeitos dos fármacos , Mitofagia/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Purinas/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Proteínas E1A de Adenovirus/genética , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Transformada , Proliferação de Células/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Fibroblastos/enzimologia , Fibroblastos/ultraestrutura , Humanos , Mitocôndrias/enzimologia , Mitocôndrias/ultraestrutura , Proteínas Proto-Oncogênicas p21(ras)/genética , Ratos , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Fatores de Tempo , Transfecção
9.
Int Rev Cell Mol Biol ; 312: 53-78, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25262238

RESUMO

ß-Catenin is involved both in cadherin-mediated intercellular adhesion and transcriptional coactivation downstream of the Wnt signaling pathway. Accumulation of ß-catenin by inhibition or knockout of its negative regulator GSK3 is known to sustain pluripotency in conjunction with other factors. However, dual function of ß-catenin and context-dependence of its activities make it difficult to dissect the mechanisms underlying this phenomenon. ß-Catenin transactivation function, which is considered to be associated with Wnt signaling, proved to be largely dispensable for the self-renewal of naïve embryonic stem cells, but required for differentiation. Instead, ß-catenin-mediated adhesion is beneficial for self-renewal, though presumably its main role is to stabilize LIF/STAT3 pathway rather than to maintain intercellular contacts per se. Yet recent report implicates E-cadherin-independent cytoplasm activity of ß-catenin in pluripotency maintenance. This review focuses on the new data concerning adhesion- and transcription-related activities of ß-catenin in control of self-renewal versus differentiation in pluripotent stem cells, as well as analyzing binding partners of ß-catenin in embryonic stem cells, which include key pluripotency regulators.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Pluripotentes/metabolismo , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo , Animais , Caderinas/genética , Caderinas/metabolismo , Adesão Celular/fisiologia , Células-Tronco Embrionárias/citologia , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Fator Inibidor de Leucemia/genética , Fator Inibidor de Leucemia/metabolismo , Células-Tronco Pluripotentes/citologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Ativação Transcricional/fisiologia , beta Catenina/genética
10.
Oncotarget ; 5(8): 2176-86, 2014 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-24742962

RESUMO

The adenoviral oncoprotein E1A influences cellular regulation by interacting with a number of cellular proteins. In collaboration with complementary oncogenes, E1A fully transforms primary cells. As part of this action, E1A inhibits transcription of c-Jun:Fos target genes while promoting that of c-Jun:ATF2-dependent genes including jun. Both c-Jun and ATF2 are hyperphosphorylated in response to E1A. In the current study, E1A was fused with the ligand binding domain of the estrogen receptor (E1A-ER) to monitor the immediate effect of E1A activation. With this approach we now show that E1A activates c-Jun N-terminal kinase (JNK), the upstream kinases MKK4 and MKK7, as well as the small GTPase Rac1. Activation of the JNK pathway requires the N-terminal domain of E1A, and, importantly, is independent of transcription. In addition, it requires the presence of ERM proteins. Downregulation of signaling components upstream of JNK inhibits E1A-dependent JNK/c-Jun activation. Taking these findings together, we show that E1A activates the JNK/c-Jun signaling pathway upstream of Rac1 in a transcription-independent manner, demonstrating a novel mechanism of E1A action.


Assuntos
Proteínas E1A de Adenovirus/metabolismo , Transformação Celular Viral/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Animais , Linhagem Celular , Ativação Enzimática/fisiologia , Receptor alfa de Estrogênio/metabolismo , Humanos , Immunoblotting , Imunoprecipitação , Camundongos , Células NIH 3T3 , Proteínas de Fusão Oncogênica/metabolismo , RNA Interferente Pequeno , Transfecção
11.
Oncotarget ; 5(5): 1157-61, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24727648

RESUMO

Here we discuss the latest progress in development of some kinase inhibitors such as inhibitors of c-MET, LIM and Bcr-Abl kinases. Importantly, many oncogenic kinases signal via the mTOR pathway, suggesting a common target for drug combinations.


Assuntos
Neoplasias/tratamento farmacológico , Proteínas Tirosina Quinases/antagonistas & inibidores , Humanos , Quinases Lim/antagonistas & inibidores , NF-kappa B/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Transdução de Sinais , Serina-Treonina Quinases TOR/antagonistas & inibidores
12.
Cell Cycle ; 13(9): 1424-39, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24626185

RESUMO

Cells respond to genotoxic stress by activating the DNA damage response (DDR). When injury is severe or irreparable, cells induce apoptosis or cellular senescence to prevent transmission of the lesions to the daughter cells upon cell division. Resistance to apoptosis is a hallmark of cancer that challenges the efficacy of cancer therapy. In this work, the effects of ionizing radiation on apoptosis-resistant E1A + E1B transformed cells were investigated to ascertain whether the activation of cellular senescence could provide an alternative tumor suppressor mechanism. We show that irradiated cells arrest cell cycle at G 2/M phase and resume DNA replication in the absence of cell division followed by formation of giant polyploid cells. Permanent activation of DDR signaling due to impaired DNA repair results in the induction of cellular senescence in E1A + E1B cells. However, irradiated cells bypass senescence and restore the population by dividing cells, which have near normal size and ploidy and do not express senescence markers. Reversion of senescence and appearance of proliferating cells were associated with downregulation of mTOR, activation of autophagy, mitigation of DDR signaling, and expression of stem cell markers.


Assuntos
Apoptose/fisiologia , Senescência Celular/fisiologia , Dano ao DNA , Células-Tronco/efeitos da radiação , Serina-Treonina Quinases TOR/metabolismo , Proteínas E1A de Adenovirus/genética , Proteínas E1B de Adenovirus/genética , Autofagia , Biomarcadores/metabolismo , Linhagem Celular Transformada , Proliferação de Células , Reparo do DNA , Replicação do DNA , Regulação para Baixo , Pontos de Checagem da Fase G2 do Ciclo Celular , Humanos , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco/metabolismo , Serina-Treonina Quinases TOR/genética , Fatores de Transcrição/metabolismo
13.
Cell Cycle ; 12(24): 3841-51, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24296616

RESUMO

Primary rodent cells undergo replicative senescence, independent from telomere shortening. We have recently shown that treatment with rapamycin during passages 3-7 suppressed replicative senescence in rat embryonic fibroblasts (REFs), which otherwise occurred by 10-14 passages. Here, we further investigated rapamycin-primed cells for an extended number of passages. Rapamycin-primed cells continued to proliferate without accumulation of senescent markers. Importantly, these cells retained the ability to undergo serum starvation- and etoposide-induced cell cycle arrest. The p53/p21 pathway was functional. This indicates that rapamycin did not cause either transformation or loss of cell cycle checkpoints. We found that rapamycin activated transcription of pluripotent genes, oct-4, sox-2, nanog, as well as further upregulated telomerase (tert) gene. The rapamycin-derived cells have mostly non-rearranged, near-normal karyotype. Still, when cultivated for a higher number of passages, these cells acquired a chromosomal marker within the chromosome 3. We conclude that suppression mTORC1 activity may prevent replicative senescence without transformation of rodent cells.


Assuntos
Senescência Celular/efeitos dos fármacos , Proteínas de Homeodomínio/genética , Fator 3 de Transcrição de Octâmero/genética , Fatores de Transcrição SOXB1/genética , Sirolimo/farmacologia , Telomerase/genética , Animais , Autofagia , Técnicas de Cultura de Células , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Proteínas de Homeodomínio/metabolismo , Cariótipo , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/metabolismo , Ratos , Fatores de Transcrição SOXB1/metabolismo , Telomerase/metabolismo
14.
Methods Mol Biol ; 965: 383-408, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23296673

RESUMO

Cellular senescence is considered as a crucial mechanism of tumor suppression that helps to prevent the growth of cells at risk for neoplastic transformation. In normal cells, cellular senescence induces an irreversible cell cycle arrest in response to telomere dysfunction, oncogene activation, genotoxic stress and a persistent DNA damage response (DDR). This process is accompanied by dramatic changes in cell morphology as well as in the activity of several signaling pathways. The senescent phenotype is multifaceted. In addition to an obligatory proliferation arrest, senescent cells manifest various senescence markers: mTOR-mediated hypertrophic growth (cell size increase), cell flattening, senescence-associated ß galactosidase (SA-ß gal) staining, expression of negative cell cycle regulators p53, p21(Waf1) and p16(Ink4a), specific chromatin reorganization including DNA segments with chromatin alterations reinforcing senescence (DNA-SCARS), senescence-associated secretory phenotype (SASP) and other features. Here, we describe the protocols that are used to study histone deacetylase inhibitor (HDACI)-induced cellular senescence in transformed cells with a special emphasis on the morphological features of senescence.


Assuntos
Senescência Celular , Técnicas Citológicas/métodos , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Animais , Ciclo Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Senescência Celular/genética , Amarelo de Eosina-(YS)/metabolismo , Epigênese Genética/efeitos dos fármacos , Fibroblastos/citologia , Citometria de Fluxo , Imunofluorescência , Adesões Focais/efeitos dos fármacos , Adesões Focais/metabolismo , Genes ras/genética , Hematoxilina/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Histonas/genética , Proteínas de Membrana/metabolismo , Camundongos , Oncogenes/genética , Coloração e Rotulagem , Cicatrização , beta-Galactosidase/metabolismo
15.
Int Rev Cell Mol Biol ; 299: 161-98, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22959303

RESUMO

Embryonic stem cells (ESCs) have unlimited proliferative potential, while retaining the ability to differentiate into descendants of all three embryonic layers. High proliferation rate of ESCs is accompanied by a shortening of the G(1) phase and the lack of G(1) checkpoint following DNA damage. The absence of G(1) arrest in ESCs after DNA damage is likely caused by a dysfunction of the p53-dependent p21Waf1 pathway that is a key event for the maintenance of pluripotency. There are controversial data on the functional status of p53, but it is well established that one of the key p53 target-p21Waf1-is expressed in ESCs at a very low level. Despite the lack of G(1) checkpoint, ESCs are capable to repair DNA defects; moreover the DNA damage response (DDR) signaling operates very effectively throughout the cell cycle. This review covers also the results obtained with the reprogramming of somatic cells into the induced pluripotent stem cells, for which have been shown that a partial dysfunction of the p53Waf1 pathway increases the frequency of generation of pluripotent cells. In summary, these results indicate that the G(1) checkpoint control and DDR are distinct from somatic cells and their status is tightly connected with maintaining of pluripotency and self-renewal.


Assuntos
Ciclo Celular , Dano ao DNA , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Animais , Humanos , Transdução de Sinais
16.
Cell Cycle ; 11(12): 2402-7, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22672902

RESUMO

The TOR (target of rapamycin) pathway is involved in aging in diverse organisms from yeast to mammals. We have previously demonstrated in human and rodent cells that mTOR converts stress-induced cell cycle arrest to irreversible senescence (geroconversion), whereas rapamycin decelerates or suppresses geroconversion during cell cycle arrest. Here, we investigated whether rapamycin can suppress replicative senescence of rodent cells. Mouse embryonic fibroblasts (MEFs) gradually acquired senescent morphology and ceased proliferation. Rapamycin decreased cellular hypertrophy, and SA-ß-Gal staining otherwise developed by 4-6 passages, but it blocked cell proliferation, masking its effects on replicative lifespan. We determined that rapamycin inhibited pS6 at 100-300 pM and inhibited proliferation with IC(50) around 30 pM. At 30 pM, rapamycin partially suppressed senescence. However, the gerosuppressive effect was balanced by the cytostatic effect, making it difficult to suppress senescence without causing quiescence. We also investigated rat embryonic fibroblasts (REFs), which exhibited markers of senescence at passage 7, yet were able to slowly proliferate until 12-14 passages. REFs grew in size, acquired a large, flat cell morphology, SA-ß-Gal staining and components of DNA damage response (DDR), in particular, γH2AX/53BP1 foci. Incubation of REFs with rapamycin (from passage 7 to passage 10) allowed REFs to overcome the replicative senescence crisis. Following rapamycin treatment and removal, a fraction of proliferating REFs gradually increased and senescent phenotype disappeared completely by passage 24.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Senescência Celular/efeitos dos fármacos , Sirolimo/farmacologia , Animais , Proteínas Reguladoras de Apoptose , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Fibroblastos/metabolismo , Proteínas de Choque Térmico/metabolismo , Histonas/metabolismo , Camundongos , Proteínas Nucleares/metabolismo , Fosforilação , Ratos , Proteínas Quinases S6 Ribossômicas/metabolismo
17.
Biochimie ; 93(9): 1408-14, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21554922

RESUMO

Cyclin-dependent kinase inhibitor p21(Waf1) is known to have alternative functions associated with positive regulation of proliferation, actin cytoskeleton remodeling and suppression of apoptosis. The goal of the present study was to assess the role of p21(Waf1) in the establishment of the transformed phenotype of mouse embryo fibroblasts with stable expression of E1Aad5 and c-Ha-ras complementary oncogenes. Herein, we demonstrate that E1A/c-Ha-Ras-transformed p21(Waf1)-null fibroblasts possess some characteristic features of transformed cells, such as loss of contact inhibition, high saturation density, shortened cell cycle, inability to undergo cell-cycle arrest after DNA damage and serum deprivation, but, at the same time, they are not completely transformed in that they are unable to proliferate at clonal density, are anchorage-dependent, retain a fibroblast-like morphology with pronounced actin cytoskeleton and show reduced migration and invasion. Our data support the concept of p21(Waf1) "tumor suppressor" having alternative oncogenic functions in the cytoplasm and for the first time indicate that p21(Waf1) can be indispensable for complete oncogenic transformation.


Assuntos
Proteínas E1A de Adenovirus/metabolismo , Transformação Celular Neoplásica/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Fibroblastos/citologia , Oncogenes , Proteínas E1A de Adenovirus/genética , Animais , Apoptose , Ciclo Celular , Linhagem Celular Transformada , Movimento Celular , Proliferação de Células , Transformação Celular Neoplásica/patologia , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Citoplasma/metabolismo , Fibroblastos/metabolismo , Genes ras , Camundongos , Transdução Genética
18.
Cancer Biol Ther ; 12(12): 1069-77, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22236879

RESUMO

HDAC inhibitors (HDACi) suppress the growth of tumor cells due to induction of cell cycle arrest, senescence or apoptosis. Recent data demonstrate that HDACi can interfere with DNA Damage Response (DDR) thereby sensitizing the cells to DNA damaging agents. Here, we show that HDACi sodium butyrate (NaBut) potentiates the formation of γH2AX foci predominantly in S-phase E1A+Ras cells. Accumulation of γH2AX foci sensitizes the cells toward such DNA damaging agents as irradiation (IR) and adriamycin. In fact, NaBut potentiates the persistence of γH2AX foci induced by genotoxic agents. The synergizing effects depend on DNA damaging factors and on the order of NaBut treatment. Indeed, NaBut treatment for 24 h leads to an accumulation of G 1-phase cells and a lack of S-phase cells, therefore, adriamycin, a powerful S-phase-specific inhibitor, when added to NaBut-treated cells, is unable to substantially add γH2AX foci. In contrast, IR produces both single- and double-strand DNA breaks at any stage of the cell cycle and was shown to increase γH2AX foci in NaBut-treated cells. Further, a lifetime of IR-induced γH2AX foci depends on the subsequent presence of HDACi. Correspondingly, NaBut withdrawal leads to the extinction of IR-induced γH2AX foci. This necessitates HDACi to hold the IR-induced γH2AX foci unrepaired. However, the IR-induced γH2AX foci persist after long-term NaBut treatment (72 h) even after washing the drug. Thus, although signaling pathways regulating H2AX phosphorylation in NaBut-treated cells remain to be investigated, the obtained results show that NaBut potentiates effects of DNA damaging agents by facilitating formation and persistence of γH2AX foci.


Assuntos
Proteínas E1A de Adenovirus/metabolismo , Butiratos/farmacologia , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Histonas/metabolismo , Proteína Oncogênica p21(ras)/metabolismo , Proteínas E1A de Adenovirus/genética , Apoptose , Quebras de DNA de Cadeia Dupla , Histonas/genética , Humanos , Proteína Oncogênica p21(ras)/genética , Fosforilação , Processamento de Proteína Pós-Traducional , Transdução de Sinais
19.
Biol Cell ; 102(10): 549-60, 2010 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-20626347

RESUMO

BACKGROUND: GSK3beta (glycogen synthase kinase 3beta) regulates the expression level and activity of various target proteins, including beta-catenin. beta-Catenin is a co-activator of Wnt-dependent genes as well as a partner for transmembrane cadherins to mediate cell-to-cell adhesion. In some cases, inhibition of GSK3beta activity was shown to promote self-renewal of ESCs (embryonic stem cells), but immediate effects of GSK3beta inhibitors in these cells still remain elusive. RESULTS: Here, we address the effects of GSK3beta inhibitors BIO (6-bromoindirubin-3'-oxime) and CHIR99021 on mESCs (mouse ESCs), focusing on modulation of beta-catenin activities. We found that, upon GSK3beta inhibition, the colonies of undifferentiated mESCs acquire a more compact morphology. This change is paralleled by two somewhat polar effects: (i) the accumulation of the beta-catenin, which is co-localized with E-cadherin at the plasma membrane, and the cytoplasmic, tyrosine unphosphorylated beta-catenin, which is able to bind the GST (glutathione transferase)-fused cytoplasmic domain of E-cadherin; and (ii) the accumulation of the tyrosine phosphorylated beta-catenin and its nuclear translocation that is accompanied by activation of the Tcf (T-cell factor)/beta-catenin-dependent transcription of Top-Flash reporter. The Tcf-mediated activation, however, does not affect most of the analysed Wnt-responsive genes involved in EMT (epithelial-mesenchymal transition) or cell-cycle progression, suggesting that the adhesive function of beta-catenin is dominant over transcription in undifferentiated mESCs. Treatment with BIO decreases proliferation rates of mESCs. This is not due to apoptosis, but rather to accumulation of cells in G1 phase of the cell cycle and is accompanied by down-regulation of the c-myc mRNA content. CONCLUSION: Our results suggest that inhibition of GSK3beta activity in mESCs enhances both the beta-catenin/E-cadherin-mediated adhesion and the Tcf/beta-catenin-dependent transcription, but does not activate transcription in most of the examined genes involved in EMT and cell cycle progression.


Assuntos
Células-Tronco Embrionárias/citologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/metabolismo , Transdução de Sinais , beta Catenina/metabolismo , Animais , Caderinas/metabolismo , Adesão Celular , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Transição Epitelial-Mesenquimal , Imunofluorescência , Glicogênio Sintase Quinase 3 beta , Camundongos
20.
Biochem Biophys Res Commun ; 391(1): 142-6, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19900401

RESUMO

HDAC inhibitors induce cell cycle arrest of E1A+Ras-transformed cells accompanied by e2f1 gene down-regulation and activation of Wnt pathway. Here we show that e2f1 expression is regulated through the Wnt/Tcf-pathway: e2f1 promoter activity is inhibited by sodium butyrate (NaB) and by overexpression of beta-catenin/Tcf. The e2f1 promoter was found to contain two putative Tcf-binding elements: the proximal one competes well with canonical Tcf element in DNA-binding assay. Being inserted into luciferase reporter vector, the identified element provides positive transcriptional regulation in response to beta-catenin/Tcf co-transfection and NaB treatment. Thus we have firstly demonstrated that e2f1 belongs to genes regulated through Wnt/beta-catenin/Tcf pathway.


Assuntos
Fator de Transcrição E2F1/genética , Regulação da Expressão Gênica , Fatores de Transcrição TCF/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Sequência de Bases , Butiratos/farmacologia , Linhagem Celular , Inibidores de Histona Desacetilases/farmacologia , Camundongos , Dados de Sequência Molecular , Regiões Promotoras Genéticas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...