Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Cancer Sci ; 108(4): 563-569, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28165648

RESUMO

"Cellular senescence" is a state in which cells undergo irreversible cell cycle arrest in response to a variety of cellular stresses. Once cells senesce, they are strongly resistant to any mitogens, including oncogenic stimuli. Therefore, cellular senescence has been assumed to be a potent anticancer mechanism. Although irreversible cell-cycle arrest is traditionally considered the major characteristic of senescent cells, recent studies have revealed some additional functions. Most noteworthy is the increased secretion of various secretory proteins, such as inflammatory cytokines, chemokines, growth factors, and MMPs, into the surrounding extracellular fluid. These newly recognized senescent phenotypes, termed senescence-associated secretory phenotypes (SASPs), reportedly contribute to tumor suppression, wound healing, embryonic development, and even tumorigenesis promotion. Thus, SASPs appear to be beneficial or deleterious, depending on the biological context. As senescent cells are known to accumulate during the aging process in vivo, it is quite possible that their accumulation in aged tissues promotes age-associated functional decline and various diseases, including cancers, at least to some extent. Here, we focus on and discuss the functional and regulatory network of SASPs toward opening up new possibilities for controlling aging and aging-associated diseases.


Assuntos
Envelhecimento/genética , Senescência Celular/genética , Doença/genética , Neoplasias/genética , Envelhecimento/metabolismo , Animais , Pontos de Checagem do Ciclo Celular/genética , Transformação Celular Neoplásica/genética , Humanos , Neoplasias/metabolismo , Fenótipo , Via Secretória/genética
2.
Nat Commun ; 14(1): 6420, 2023 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-37828010

RESUMO

Identification of factors that regulate chromatin condensation is important for understanding of gene regulation. High-mobility group AT-hook (HMGA) proteins 1 and 2 are abundant nonhistone chromatin proteins that play a role in many biological processes including tissue stem-progenitor cell regulation, but the nature of their protein function remains unclear. Here we show that HMGA2 mediates direct condensation of polynucleosomes and forms droplets with nucleosomes. Consistently, most endogenous HMGA2 localized to transposase 5- and DNase I-inaccessible chromatin regions, and its binding was mostly associated with gene repression, in mouse embryonic neocortical cells. The AT-hook 1 domain was necessary for chromatin condensation by HMGA2 in vitro and in cellulo, and an HMGA2 mutant lacking this domain was defective in the ability to maintain neuronal progenitors in vivo. Intrinsically disordered regions of other proteins could substitute for the AT-hook 1 domain in promoting this biological function of HMGA2. Taken together, HMGA2 may regulate neural cell fate by its chromatin condensation activity.


Assuntos
Cromatina , Regulação da Expressão Gênica , Camundongos , Animais , Nucleossomos , Células-Tronco , Diferenciação Celular/genética
3.
Cancer Sci ; 103(3): 439-47, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22106824

RESUMO

High-mobility group A1 (Hmga1) protein is an architectural chromatin factor, and aberrant Hmga1 expression in mice causes hematopoietic malignancies with defects in cellular differentiation. However, the functional involvement of Hmga1 in hematopoietic development and leukemic cells remains to be elucidated. Using Hmga1-green fluorescent protein (GFP) knock-in mice that endogenously express an Hmga1-GFP fusion protein, we examined Hmga1 expression in undifferentiated and differentiated populations of hematopoietic cells. During early T cell development in the thymus, Hmga1 is highly expressed in CD4/CD8-double negative (DN) cells and is transiently downregulated in CD4/CD8-double positive (DP) cells. Consistently, Hmga1 directly binds to cis-regulatory elements in the CD4/CD8 loci and the heterochromatin foci in DN-stage cells, but not in DP cells. Interestingly, CD4/CD8 expression in DN-stage leukemic cells is induced by inhibition of Hmga1 binding to nuclear DNA or RNA interference-mediated Hmga1 knockdown. In addition, Hmga1-depleted leukemic T cells markedly diminish proliferation, with transcriptional activation of cyclin-dependent kinase inhibitor genes as a direct target of Hmga1. The data in the present study reveal a role of Hmga1 in transcriptional silencing in T cell lineages and leukemic cells.


Assuntos
Antígenos CD4/biossíntese , Antígenos CD8/biossíntese , Linhagem da Célula , Proteína HMGA1a/genética , Proteína HMGA1a/metabolismo , Leucemia/metabolismo , Subpopulações de Linfócitos T/metabolismo , Animais , Western Blotting , Antígenos CD4/genética , Antígenos CD8/genética , Diferenciação Celular/genética , Linhagem da Célula/genética , Proliferação de Células , Imunoprecipitação da Cromatina , Citometria de Fluxo , Imunofluorescência , Técnicas de Introdução de Genes , Inativação Gênica , Humanos , Células Jurkat , Leucemia/genética , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Subpopulações de Linfócitos T/patologia , Transcrição Gênica , Transfecção
4.
Am J Pathol ; 174(3): 854-68, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19179606

RESUMO

Pancreatic cancer is a highly aggressive malignancy due to elevated mitotic activities and epithelial-mesenchymal transition (EMT). Oncogenic RAS and transforming growth factor-beta signaling are implicated in these malignant features. The mechanisms that underlie EMT need to be addressed since it promotes tissue invasion and metastasis. The high-mobility group A protein 2 (HMGA2) is a non-histone chromatin factor that is primarily expressed in undifferentiated tissues and tumors of mesenchymal origin. However, its role in EMT in pancreatic cancer is largely unknown. Here we report that HMGA2 is involved in EMT maintenance in human pancreatic cancer cells. Specific knockdown of HMGA2 inhibited cell proliferation, leading to an epithelial-state transition that restores cell-cell contact due to E-cadherin up-regulation. Consistently, an inverse correlation between HMGA2-positive cells and E-cadherin-positive cells was found in cancer tissues. Inhibition of the RAS/MEK pathway also induced an epithelial transition, together with HMGA2 down-regulation. Transcriptional repressors of the E-cadherin gene, such as SNAIL, decreased after HMGA2 knockdown since HMGA2 directly activated the SNAlL gene promoter. The decrease of SNAIL after RAS/MEK inhibition was suppressed by HMGA2 overexpression. Further, let-7 microRNA-mediated HMGA2 down-regulation had no effect on the prevention of the transformed phenotype in these cells. These data shed light on the importance of HMGA2 in reversibly maintaining EMT, suggesting that HMGA2 is a potential therapeutic target for the treatment of pancreatic cancer.


Assuntos
Células Epiteliais/patologia , Proteína HMGA2/fisiologia , Mesoderma/fisiologia , Neoplasias Pancreáticas/patologia , Animais , Linhagem Celular Tumoral , Primers do DNA , Desoxirribonuclease I , Células Epiteliais/fisiologia , Amplificação de Genes , Regulação Neoplásica da Expressão Gênica , Genes ras , Proteína HMGA2/deficiência , Proteína HMGA2/genética , Células HeLa , Humanos , Mesoderma/patologia , Camundongos , Camundongos Knockout , MicroRNAs/genética , Plasmídeos , Regiões Promotoras Genéticas , RNA Neoplásico/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética
5.
Am J Pathol ; 175(4): 1675-85, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19729480

RESUMO

The development of stomach cancer is closely associated with chronic inflammation, and the Wnt/beta-catenin signaling pathway is activated in most cases of this cancer. High-mobility group A (HMGA) proteins are oncogenic chromatin factors that are primarily expressed not only in undifferentiated tissues but also in various tumors. Here we report that HMGA1 is induced by the Wnt/beta-catenin pathway and maintains proliferation of gastric cancer cells. Specific knockdown of HMGA1 resulted in marked reduction of cell growth. The loss of beta-catenin or its downstream c-myc decreased HMGA1 expression, whereas Wnt3a treatment increased HMGA1 and c-myc transcripts. Furthermore, Wnt3a-induced expression of HMGA1 was inhibited by c-myc knockdown, suggesting that HMGA1 is a downstream target of the Wnt/beta-catenin pathway. Enhanced expression of HMGA1 coexisted with the nuclear accumulation of beta-catenin in about 30% of gastric cancer tissues. To visualize the expression of HMGA1 in vivo, transgenic mice expressing endogenous HMGA1 fused to enhanced green fluorescent protein were generated and then crossed with K19-Wnt1/C2mE mice, which develop gastric tumors through activation of both the Wnt and prostaglandin E2 pathways. Expression of HMGA1-enhanced green fluorescent protein was normally detected in the forestomach, along the upper border of the glandular stomach, but its expression was also up-regulated in cancerous glandular stomach. These data suggest that HMGA1 is involved in proliferation and gastric tumor formation via the Wnt/beta-catenin pathway.


Assuntos
Proteína HMGA1a/metabolismo , Transdução de Sinais , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Introdução de Genes , Proteínas de Fluorescência Verde/metabolismo , Humanos , Queratina-19/metabolismo , Camundongos , Proteínas Proto-Oncogênicas c-myc/metabolismo
6.
Sci Rep ; 8(1): 10888, 2018 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-30022091

RESUMO

Chromatin dynamics mediated by post-translational modifications play a crucial role in cellular response to genotoxic stress for the maintenance of genome integrity. MDC1 is a pivotal chromatin adaptor in DNA damage response (DDR) and its methylation is essential to recruit repair factors at DNA double-strand break (DSB) sites, yet their precise molecular mechanisms remain elusive. Here we identified euchromatic histone-lysine N-methyltransferase 1 (EHMT1) and EHMT2 as novel regulators of MDC1, which is required for the accumulation of DDR factors e.g. 53BP1 and RAP80, at the DSB sites. MDC1 interacts mainly with EHMT1, which is facilitated by DNA damage-initiated ATM signalling, and EHMT2 dominantly modulates methylation of MDC1 lysine 45. This regulatory modification promotes the interaction between MDC1 and ATM to expand activated ATM on damaged chromatin and dysfunctional telomere. These findings identify EHMT1 and EHMT2 as DDR components, with implications for genome-integrity maintenance through proper dynamic methylation of MDC1.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Dano ao DNA , Metilação de DNA , Antígenos de Histocompatibilidade/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Lisina/química , Proteínas Nucleares/química , Transativadores/química , Proteínas Adaptadoras de Transdução de Sinal , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas de Ciclo Celular , Cromatina/genética , Cromatina/metabolismo , Reparo do DNA , Instabilidade Genômica , Antígenos de Histocompatibilidade/genética , Histona-Lisina N-Metiltransferase/genética , Histonas , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Transativadores/genética , Transativadores/metabolismo , Células Tumorais Cultivadas
7.
Nat Commun ; 9(1): 4109, 2018 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-30294002

RESUMO

This Article contains errors in Fig. 4. In panel d, the lanes of the western blot should have been labeled '1.05','1.06, '1.09', '1.11' '1.13', '1.16', '1.19', '1.22', '1.24', '1.25'. The correct version of Figure 4 appears in the associated Publisher Correction.

8.
Nat Commun ; 9(1): 1249, 2018 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-29593264

RESUMO

Accumulating evidence indicates that the senescence-associated secretory phenotype (SASP) contributes to many aspects of physiology and disease. Thus, controlling the SASP will have tremendous impacts on our health. However, our understanding of SASP regulation is far from complete. Here, we show that cytoplasmic accumulation of nuclear DNA plays key roles in the onset of SASP. Although both DNase2 and TREX1 rapidly remove the cytoplasmic DNA fragments emanating from the nucleus in pre-senescent cells, the expression of these DNases is downregulated in senescent cells, resulting in the cytoplasmic accumulation of nuclear DNA. This causes the aberrant activation of cGAS-STING cytoplasmic DNA sensors, provoking SASP through induction of interferon-ß. Notably, the blockage of this pathway prevents SASP in senescent hepatic stellate cells, accompanied by a decline of obesity-associated hepatocellular carcinoma development in mice. These findings provide valuable new insights into the roles and mechanisms of SASP and possibilities for their control.


Assuntos
Senescência Celular , Citoplasma/enzimologia , Desoxirribonucleases/metabolismo , Regulação para Baixo , Animais , Carcinoma Hepatocelular/enzimologia , Linhagem Celular , Dano ao DNA , DNA de Cadeia Simples/metabolismo , Exodesoxirribonucleases/metabolismo , Células Estreladas do Fígado/enzimologia , Humanos , Interferon beta/metabolismo , Fígado/enzimologia , Neoplasias Hepáticas/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Fenótipo , Fosfoproteínas/metabolismo , Fosforilação , Interferência de RNA
9.
Cancer Sci ; 98(12): 1893-901, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17877762

RESUMO

Retinoblastoma protein (RB) acts as a tumor suppressor in many tissue types, by promoting cell arrest via E2F-mediated transcriptional repression. In addition to the aberrant forms of the RB gene found in different types of cancers, many viral oncoproteins including the simian virus 40 large T antigen target RB. However, cellular factors that inhibit RB function remain to be elucidated. Here, we report that RB interacts with the high mobility group protein A1 (HMGA1), a-non-histone architectural chromatin factor that is frequently overexpressed in cancer cells. HMGA1 binds the small pocket domain of RB, and competes with HDAC1. Subsequently, overexpression of HMGA1 abolishes the inhibitory effect of RB on E2F-activated transcription from the cyclin E promoter. Under serum starvation, T98G cells had been previously shown to be arrested in the G0 phase in an RB-mediated manner. The G0 phase was characterized by growth arrest and low levels of transcription, together with the hypophosphorylation of RB and the downregulation of HMGA1. In contrast, such serum-depleted G0 arrest was abrogated in T98G cells overexpressing HMGA1. The overexpressed HMGA1 was found to form complexes with cellular RB, suggesting that downregulation of HMGA1 is required for G0 arrest. There were no phenotypic changes in HMGA1-expressing T98G cells in the presence of serum, but the persistent expression of HMGA1 under serum starvation caused various nuclear abnormalities, which were similarly induced in T antigen-expressing T98G cells. Our present findings indicate that overexpression of HMGA1 disturbs RB-mediated cell arrest, suggesting a negative control of RB by HMGA1.


Assuntos
Proteína HMGA1a/fisiologia , Proteína do Retinoblastoma/antagonistas & inibidores , Animais , Clonagem Molecular , Genes Reporter , Vetores Genéticos , Glutationa Transferase/genética , Proteína HMGA1a/genética , Humanos , Camundongos , Plasmídeos , Fase de Repouso do Ciclo Celular , Transfecção
10.
Mol Cell Biol ; 23(8): 2834-43, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12665582

RESUMO

DNA methylation is involved in a variety of genome functions, including gene control and chromatin dynamics. MBD1 is a transcriptional regulator through the cooperation of a methyl-CpG binding domain, cysteine-rich CXXC domains, and a transcriptional repression domain. A yeast two-hybrid screen was performed to investigate the role of MBD1 in methylation-based transcriptional repression. We report a mediator, MBD1-containing chromatin-associated factor (MCAF), that interacts with the transcriptional repression domain of MBD1. MCAF harbors two conserved domains that allow it to interact with MBD1 and enhancer-like transactivator Sp1. MCAF possesses a coactivator-like activity, and it seems to facilitate Sp1-mediated transcription. In contrast, the MBD1-MCAF complex blocks transcription through affecting Sp1 on methylated promoter regions. These data provide a mechanistic basis for direct inhibition of gene expression via methylation-dependent and histone deacetylation-resistant processes.


Assuntos
Quimiocina CCL2/metabolismo , Proteínas de Ligação a DNA , Proteínas Repressoras/metabolismo , Sítios de Ligação/genética , Linhagem Celular , Quimiocina CCL2/genética , DNA/genética , DNA/metabolismo , Metilação de DNA , Inibidores Enzimáticos/farmacologia , Células HeLa , Inibidores de Histona Desacetilases , Humanos , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/química , Proteínas Repressoras/genética , Fatores de Transcrição , Transcrição Gênica/efeitos dos fármacos , Técnicas do Sistema de Duplo-Híbrido
11.
Nat Commun ; 8(1): 2050, 2017 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-29234059

RESUMO

p16Ink4a and p21Cip1/Waf1 act as tumour suppressors through induction of cellular senescence. However, senescence-independent roles of these CDK inhibitors are not well understood. Here, we report an unexpected function of p16Ink4 and p21Cip1/Waf1, namely, tumour promotion through chemotaxis. In monocytic myeloid-derived suppressor cells (Mo-MDSCs), p16Ink4 and p21Cip1/Waf1 are highly expressed and stimulate CX3CR1 chemokine receptor expression by preventing CDK-mediated phosphorylation and inactivation of SMAD3. Thus, deletion of p16 Ink4 and p21 Cip1/Waf1 reduces CX3CR1 expression, thereby inhibiting Mo-MDSC accumulation in tumours expressing CX3CL1 and suppressing the tumour progression in mice. Notably, blockade of the CX3CL1/CX3CR1 axis suppresses tumour growth, whereas inactivation of CDKs elicits the opposite effect. These findings reveal an unexpected function of p16 Ink4a and p21 Waf1/Cip1 and indicate that regulation of Mo-MDSCs chemotaxis is a valuable potential strategy for control of tumour development.


Assuntos
Quimiotaxia , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Células Supressoras Mieloides/patologia , Neoplasias/patologia , Animais , Receptor 1 de Quimiocina CX3C/antagonistas & inibidores , Receptor 1 de Quimiocina CX3C/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/metabolismo , Dimetil Sulfóxido/farmacologia , Progressão da Doença , Feminino , Flavonoides/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Piperidinas/farmacologia , Proteína Smad3/metabolismo , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Nat Commun ; 8: 15729, 2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28585531

RESUMO

Cellular senescence prevents the proliferation of cells at risk for neoplastic transformation. However, the altered secretome of senescent cells can promote the growth of the surrounding cancer cells. Although extracellular vesicles (EVs) have emerged as new players in intercellular communication, their role in the function of senescent cell secretome has been largely unexplored. Here, we show that exosome-like small EVs (sEVs) are important mediators of the pro-tumorigenic function of senescent cells. sEV-associated EphA2 secreted from senescent cells binds to ephrin-A1, that is, highly expressed in several types of cancer cells and promotes cell proliferation through EphA2/ephrin-A1 reverse signalling. sEV sorting of EphA2 is increased in senescent cells because of its enhanced phosphorylation resulting from oxidative inactivation of PTP1B phosphatase. Our results demonstrate a novel mechanism of reactive oxygen species (ROS)-regulated cargo sorting into sEVs, which is critical for the potentially deleterious growth-promoting effect of the senescent cell secretome.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Efrina-A2/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Senescência Celular , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Espectrometria de Massas , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptor EphA2 , Proteínas Recombinantes/metabolismo , Transdução de Sinais
13.
Nat Commun ; 8: 15287, 2017 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-28508895

RESUMO

Emerging evidence is revealing that exosomes contribute to many aspects of physiology and disease through intercellular communication. However, the biological roles of exosome secretion in exosome-secreting cells have remained largely unexplored. Here we show that exosome secretion plays a crucial role in maintaining cellular homeostasis in exosome-secreting cells. The inhibition of exosome secretion results in the accumulation of nuclear DNA in the cytoplasm, thereby causing the activation of cytoplasmic DNA sensing machinery. This event provokes the innate immune response, leading to reactive oxygen species (ROS)-dependent DNA damage response and thus induce senescence-like cell-cycle arrest or apoptosis in normal human cells. These results, in conjunction with observations that exosomes contain various lengths of chromosomal DNA fragments, indicate that exosome secretion maintains cellular homeostasis by removing harmful cytoplasmic DNA from cells. Together, these findings enhance our understanding of exosome biology, and provide valuable new insights into the control of cellular homeostasis.


Assuntos
Citoplasma/metabolismo , DNA/metabolismo , Exossomos/metabolismo , Homeostase , Animais , Apoptose , Pontos de Checagem do Ciclo Celular , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Citoplasma/genética , Células HEK293 , Células HeLa , Humanos , Camundongos , Modelos Biológicos , Espécies Reativas de Oxigênio/metabolismo
14.
Nat Commun ; 7: 11117, 2016 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-27030108

RESUMO

Temporal regulation of microtubule dynamics is essential for proper progression of mitosis and control of microtubule plus-end tracking proteins by phosphorylation is an essential component of this regulation. Here we show that Aurora B and CDK1 phosphorylate microtubule end-binding protein 2 (EB2) at multiple sites within the amino terminus and a cluster of serine/threonine residues in the linker connecting the calponin homology and end-binding homology domains. EB2 phosphorylation, which is strictly associated with mitotic entry and progression, reduces the binding affinity of EB2 for microtubules. Expression of non-phosphorylatable EB2 induces stable kinetochore microtubule dynamics and delays formation of bipolar metaphase plates in a microtubule binding-dependent manner, and leads to aneuploidy even in unperturbed mitosis. We propose that Aurora B and CDK1 temporally regulate the binding affinity of EB2 for microtubules, thereby ensuring kinetochore microtubule dynamics, proper mitotic progression and genome stability.


Assuntos
Aurora Quinase B/fisiologia , Quinases Ciclina-Dependentes/fisiologia , Proteínas Associadas aos Microtúbulos/fisiologia , Mitose/fisiologia , Aurora Quinase B/análise , Aurora Quinase B/metabolismo , Sítios de Ligação , Proteína Quinase CDC2 , Linhagem Celular , Quinases Ciclina-Dependentes/análise , Quinases Ciclina-Dependentes/metabolismo , Instabilidade Genômica , Humanos , Cinetocoros/metabolismo , Cinetocoros/ultraestrutura , Proteínas Associadas aos Microtúbulos/análise , Proteínas Associadas aos Microtúbulos/química , Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Mitose/genética , Fosforilação
15.
J Cell Biol ; 212(3): 281-8, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26811421

RESUMO

Topoisomerase IIß-binding protein 1 (TOPBP1) participates in DNA replication and DNA damage response; however, its role in DNA repair and relevance for human cancer remain unclear. Here, through an unbiased small interfering RNA screen, we identified and validated TOPBP1 as a novel determinant whose loss sensitized human cells to olaparib, an inhibitor of poly(ADP-ribose) polymerase. We show that TOPBP1 acts in homologous recombination (HR) repair, impacts olaparib response, and exhibits aberrant patterns in subsets of human ovarian carcinomas. TOPBP1 depletion abrogated RAD51 loading to chromatin and formation of RAD51 foci, but without affecting the upstream HR steps of DNA end resection and RPA loading. Furthermore, TOPBP1 BRCT domains 7/8 are essential for RAD51 foci formation. Mechanistically, TOPBP1 physically binds PLK1 and promotes PLK1 kinase-mediated phosphorylation of RAD51 at serine 14, a modification required for RAD51 recruitment to chromatin. Overall, our results provide mechanistic insights into TOPBP1's role in HR, with potential clinical implications for cancer treatment.


Assuntos
Proteínas de Transporte/metabolismo , Montagem e Desmontagem da Cromatina , Cromatina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Recombinação Homóloga , Proteínas Nucleares/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Ftalazinas/farmacologia , Piperazinas/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Rad51 Recombinase/metabolismo , Proteínas de Transporte/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/genética , Relação Dose-Resposta a Droga , Feminino , Células HEK293 , Células HeLa , Humanos , Proteínas Nucleares/genética , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Fosforilação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Interferência de RNA , Rad51 Recombinase/genética , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transfecção , Quinase 1 Polo-Like
16.
Mol Cancer Ther ; 14(4): 1004-13, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25700705

RESUMO

Trifluridine (FTD) is a key component of the novel oral antitumor drug TAS-102, which consists of FTD and a thymidine phosphorylase inhibitor. Like 5-fluoro-2'-deoxyuridine (FdUrd), a deoxynucleoside form of 5-fluorouracil metabolite, FTD is sequentially phosphorylated and not only inhibits thymidylate synthase activity, but is also incorporated into DNA. Although TAS-102 was effective for the treatment of refractory metastatic colorectal cancer in clinical trials, the mechanism of FTD-induced cytotoxicity is not completely understood. Here, we show that FTD as well as FdUrd induce transient phosphorylation of Chk1 at Ser345, and that this is followed by accumulation of p53 and p21 proteins in p53-proficient human cancer cell lines. In particular, FTD induced p53-dependent sustained arrest at G2 phase, which was associated with a proteasome-dependent decrease in the Cyclin B1 protein level and the suppression of CCNB1 and CDK1 gene expression. In addition, a p53-dependent increase in p21 protein was associated with an FTD-induced decrease in Cyclin B1 protein. Although numerous ssDNA and dsDNA breaks were induced by FdUrd, few DNA strand breaks were detected in FTD-treated HCT-116 cells despite massive FTD misincorporation into genomic DNA, suggesting that the antiproliferative effect of FTD is not due to the induction of DNA strand breaks. These distinctive effects of FTD provide insights into the cellular mechanism underlying its antitumor effect and may explain the clinical efficacy of TAS-102.


Assuntos
Quebras de DNA , Pontos de Checagem da Fase G2 do Ciclo Celular , Trifluridina/metabolismo , Trifluridina/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Proteína Quinase CDC2/genética , Proteína Quinase CDC2/metabolismo , Linhagem Celular Tumoral , Ciclina B1/genética , Ciclina B1/metabolismo , Replicação do DNA , Desoxiuridina/análogos & derivados , Pontos de Checagem da Fase G2 do Ciclo Celular/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Inativação de Genes , Humanos , Concentração Inibidora 50 , Proteína Oncogênica p21(ras)/genética , Proteína Oncogênica p21(ras)/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Transcrição Gênica , Proteína Supressora de Tumor p53/genética
17.
Mol Cancer Ther ; 14(10): 2332-42, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26208523

RESUMO

Platinum-based chemotherapeutic drugs are widely used as components of combination chemotherapy in the treatment of cancer. One such drug, oxaliplatin, exerts a synergistic effect against advanced colorectal cancer in combination with 5-fluorouracil (5-FU) and leucovorin. In the p53-proficient colorectal cancer cell line HCT116, oxaliplatin represses the expression of deoxyuridine triphosphatase (dUTPase), a ubiquitous pyrophosphatase that catalyzes the hydrolysis of dUTP to dUMP and inhibits dUTP-mediated cytotoxicity. However, the underlying mechanism of this activity has not been completely elucidated, and it remains unclear whether factors other than downregulation of dUTPase contribute to the synergistic effect of 5-FU and oxaliplatin. In this study, we found that oxaliplatin and dachplatin, platinum-based drugs containing the 1,2-diaminocyclohexane (DACH) carrier ligand, repressed the expression of nuclear isoform of dUTPase (DUT-N), whereas cisplatin and carboplatin did not. Oxaliplatin induced early p53 accumulation, upregulation of primary miR-34a transcript expression, and subsequent downregulation of E2F3 and E2F1. Nutlin-3a, which activates p53 nongenotoxically, had similar effects. Introduction of miR-34a mimic also repressed E2F1 and DUT-N expression, indicating that this miRNA plays a causative role. In addition to DUT-N, oxaliplatin repressed, in a p53-dependent manner, the expression of genes encoding enzymes involved in thymidylate biosynthesis. Consequently, oxaliplatin significantly decreased the level of dTTP in the dNTP pool in a p53-dependent manner. These data indicate that the DACH carrier ligand in oxaliplatin triggers signaling via the p53-miR-34a-E2F axis, leading to transcriptional regulation that ultimately results in accumulation of dUTP and reduced dTTP biosynthesis, potentially enhancing 5-FU cytotoxicity.


Assuntos
Inativação Gênica/efeitos dos fármacos , Compostos Organoplatínicos/farmacologia , Timidina Monofosfato/biossíntese , Transcrição Gênica/efeitos dos fármacos , Proteína Supressora de Tumor p53/fisiologia , Vias Biossintéticas , Replicação do DNA , Regulação para Baixo , Sinergismo Farmacológico , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/metabolismo , Fator de Transcrição E2F3/genética , Fator de Transcrição E2F3/metabolismo , Fluoruracila/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Humanos , Imidazóis/farmacologia , MicroRNAs/genética , MicroRNAs/metabolismo , Compostos Organoplatínicos/química , Oxaliplatina , Piperazinas/farmacologia , Pirofosfatases/genética , Pirofosfatases/metabolismo , Fator de Transcrição Sp1/metabolismo
18.
Nat Struct Mol Biol ; 20(12): 1425-33, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24240613

RESUMO

Chromatin ubiquitylation flanking DNA double-strand breaks (DSBs), mediated by RNF8 and RNF168 ubiquitin ligases, orchestrates a two-branch pathway, recruiting repair factors 53BP1 or the RAP80-BRCA1 complex. We report that human demethylase JMJD1C regulates the RAP80-BRCA1 branch of this DNA-damage response (DDR) pathway. JMJD1C was stabilized by interaction with RNF8, was recruited to DSBs, and was required for local ubiquitylations and recruitment of RAP80-BRCA1 but not 53BP1. JMJD1C bound to RNF8 and MDC1, and demethylated MDC1 at Lys45, thereby promoting MDC1-RNF8 interaction, RNF8-dependent MDC1 ubiquitylation and recruitment of RAP80-BRCA1 to polyubiquitylated MDC1. Furthermore, JMJD1C restricted formation of RAD51 repair foci, and JMJD1C depletion caused resistance to ionizing radiation and PARP inhibitors, phenotypes relevant to aberrant loss of JMJD1C in subsets of breast carcinomas. These findings identify JMJD1C as a DDR component, with implications for genome-integrity maintenance, tumorigenesis and cancer treatment.


Assuntos
Proteína BRCA1/fisiologia , Quebras de DNA , Proteínas de Ligação a DNA/fisiologia , Histona Desmetilases com o Domínio Jumonji/fisiologia , Proteínas Nucleares/metabolismo , Oxirredutases N-Desmetilantes/fisiologia , Transativadores/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Proteína BRCA1/química , Proteína BRCA1/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Metilação de DNA , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Feminino , Células HeLa , Chaperonas de Histonas , Humanos , Histona Desmetilases com o Domínio Jumonji/química , Histona Desmetilases com o Domínio Jumonji/metabolismo , Proteínas Nucleares/química , Oxirredutases N-Desmetilantes/química , Oxirredutases N-Desmetilantes/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases , Rad51 Recombinase/química , Rad51 Recombinase/metabolismo , Rad51 Recombinase/fisiologia , Transativadores/química , Células Tumorais Cultivadas , Ubiquitina-Proteína Ligases , Ubiquitinação
19.
Cell Cycle ; 12(11): 1688-95, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23656789

RESUMO

Genotoxic insults, such as ionizing radiation (IR), cause DNA damage that evokes a multifaceted cellular DNA damage response (DDR). DNA damage signaling events that control protein activity, subcellular localization, DNA binding, protein-protein interactions, etc. rely heavily on time-dependent posttranslational modifications (PTMs). To complement our previous analysis of IR-induced temporal dynamics of nuclear phosphoproteome, we now identify a range of human nuclear proteins that are dynamically regulated by acetylation, and predominantly deacetylation, during IR-induced DDR by using mass spectrometry-based proteomic approaches. Apart from cataloging acetylation sites through SILAC proteomic analyses before IR and at 5 and 60 min after IR exposure of U2OS cells, we report that: (1) key components of the transcriptional machinery, such as EP300 and CREBBP, are dynamically acetylated; (2) that nuclear acetyltransferases themselves are regulated, not on the protein abundance level, but by (de)acetylation; and (3) that the recently reported p53 co-activator and methyltransferase MLL3 is acetylated on five lysines during the DDR. For selected examples, protein immunoprecipitation and immunoblotting were used to assess lysine acetylation status and thereby validate the mass spectrometry data. We thus present evidence that nuclear proteins, including those known to regulate cellular functions via epigenetic modifications of histones, are regulated by (de)acetylation in a timely manner upon cell's exposure to genotoxic insults. Overall, these results present a resource of temporal profiles of a spectrum of protein acetylation sites during DDR and provide further insights into the highly dynamic nature of regulatory PTMs that help orchestrate the maintenance of genome integrity.


Assuntos
Dano ao DNA/efeitos da radiação , Reparo do DNA , Proteínas Nucleares/metabolismo , Radiação Ionizante , Acetilação/efeitos da radiação , Acetiltransferases/metabolismo , Proteína de Ligação a CREB/metabolismo , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/metabolismo , Proteína p300 Associada a E1A/metabolismo , Histonas/metabolismo , Humanos , Lisina/metabolismo , Processamento de Proteína Pós-Traducional , Proteômica , Proteína Supressora de Tumor p53/metabolismo
20.
Mol Cell Biol ; 32(8): 1529-41, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22354988

RESUMO

The three-dimensional context of endogenous chromosomal regions may contribute to the regulation of gene clusters by influencing interactions between transcriptional regulatory elements. In this study, we investigated the effects of tumor necrosis factor (TNF) signaling on spatiotemporal enhancer-promoter interactions in the human tumor necrosis factor (TNF)/lymphotoxin (LT) gene locus, mediated by CCCTC-binding factor (CTCF)-dependent chromatin insulators. The cytokine genes LTα, TNF, and LTß are differentially regulated by NF-κB signaling in inflammatory and oncogenic responses. We identified at least four CTCF-enriched sites with enhancer-blocking activities and a TNF-responsive TE2 enhancer in the TNF/LT locus. One of the CTCF-enriched sites is located between the early-inducible LTα/TNF promoters and the late-inducible LTß promoter. Depletion of CTCF reduced TNF expression and accelerated LTß induction. After TNF stimulation, via intrachromosomal dynamics, these insulators mediated interactions between the enhancer and the LTα/TNF promoters, followed by interaction with the LTß promoter. These results suggest that insulators mediate the spatiotemporal control of enhancer-promoter associations in the TNF/LT gene cluster.


Assuntos
Carcinoma Hepatocelular/genética , Cromatina/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/genética , Linfotoxina-alfa/genética , Família Multigênica , Fator de Necrose Tumoral alfa/genética , Idoso , Fator de Ligação a CCCTC , Linhagem Celular Tumoral , Citocinas/genética , Elementos Facilitadores Genéticos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , NF-kappa B/genética , Regiões Promotoras Genéticas , Proteínas Repressoras/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA