Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Res ; 29(4): 265-273, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30814679

RESUMO

The re-emergence of Zika virus (ZIKV) in the Western Hemisphere has resulted in global public health crisis since 2015. ZIKV preferentially infects and targets human neural progenitor cells (hNPCs) and causes fetal microcephaly upon maternal infection. hNPCs not only play critical roles during fetal brain development, but also persist in adult brain throughout life. Yet the mechanism of innate antiviral immunity in hNPCs remains largely unknown. Here, we show that ZIKV infection triggers the abundant production of virus-derived small interfering RNAs in hNPCs, but not in the more differentiated progenies or somatic cells. Ablation of key RNAi machinery components significantly enhances ZIKV replication in hNPCs. Furthermore, enoxacin, a broad-spectrum antibiotic that is known as an RNAi enhancer, exerts potent anti-ZIKV activity in hNPCs and other RNAi-competent cells. Strikingly, enoxacin treatment completely prevents ZIKV infection and circumvents ZIKV-induced microcephalic phenotypes in brain organoid models that recapitulate human fetal brain development. Our findings highlight the physiological importance of RNAi-mediated antiviral immunity during the early stage of human brain development, uncovering a novel strategy to combat human congenital viral infections through enhancing RNAi.


Assuntos
Encéfalo/imunologia , Células-Tronco Neurais/imunologia , Organoides/imunologia , RNA Viral/imunologia , Infecção por Zika virus/imunologia , Zika virus/genética , Animais , Antivirais/farmacologia , Encéfalo/patologia , Linhagem Celular , Enoxacino/farmacologia , Humanos , Imunidade Inata , Células-Tronco Neurais/patologia , Organoides/patologia , Interferência de RNA , Replicação Viral , Zika virus/imunologia , Zika virus/fisiologia
2.
Cell ; 176(6): 1447-1460.e14, 2019 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-30799039

RESUMO

The presence of DNA in the cytoplasm is normally a sign of microbial infections and is quickly detected by cyclic GMP-AMP synthase (cGAS) to elicit anti-infection immune responses. However, chronic activation of cGAS by self-DNA leads to severe autoimmune diseases for which no effective treatment is available yet. Here we report that acetylation inhibits cGAS activation and that the enforced acetylation of cGAS by aspirin robustly suppresses self-DNA-induced autoimmunity. We find that cGAS acetylation on either Lys384, Lys394, or Lys414 contributes to keeping cGAS inactive. cGAS is deacetylated in response to DNA challenges. Importantly, we show that aspirin can directly acetylate cGAS and efficiently inhibit cGAS-mediated immune responses. Finally, we demonstrate that aspirin can effectively suppress self-DNA-induced autoimmunity in Aicardi-Goutières syndrome (AGS) patient cells and in an AGS mouse model. Thus, our study reveals that acetylation contributes to cGAS activity regulation and provides a potential therapy for treating DNA-mediated autoimmune diseases.


Assuntos
DNA/imunologia , Nucleotidiltransferases/metabolismo , Tolerância a Antígenos Próprios/imunologia , Acetilação , Sequência de Aminoácidos , Animais , Aspirina/farmacologia , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Doenças Autoimunes do Sistema Nervoso/genética , Doenças Autoimunes do Sistema Nervoso/imunologia , Doenças Autoimunes do Sistema Nervoso/metabolismo , Autoimunidade , Linhagem Celular , DNA/genética , DNA/metabolismo , Modelos Animais de Doenças , Exodesoxirribonucleases/metabolismo , Células HEK293 , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Mutação , Malformações do Sistema Nervoso/genética , Malformações do Sistema Nervoso/imunologia , Malformações do Sistema Nervoso/metabolismo , Nucleotidiltransferases/antagonistas & inibidores , Nucleotidiltransferases/química , Nucleotidiltransferases/genética , Células THP-1
3.
Zhonghua Yi Xue Za Zhi ; 93(14): 1099-103, 2013 Apr 09.
Artigo em Chinês | MEDLINE | ID: mdl-23902846

RESUMO

OBJECTIVE: To establish Gankyrin knocking down 4T1-luc cell model and detect the effects of Gankyrin expression on breast cancer metastasis. METHODS: 4T1-luc cells carrying shGankyrin construct were established by lentivirus infection and antibiotic screening. Western blotting and real-time PCR were used to check the expression levels of Gankyrin. In vivo imaging system was used to monitor the effects of Gankyrin knocked down on cell growth and tumor metastasis after the in situ implantation of Gankyrin knocking down 4T1-luc cells in BALB/c mice. RESULTS: The cell expression decreased at the protein and mRNA levels. Gankyrin mRNA expression in different shGankyrin 4T1-luc cells was respectively 4.9%, 25.1% and 69.8% versus the control cells. ShGankyrin#2 4T1-luc cells were chosen for in situ implantation into BAL/c mice because luminescent intensity was consistent with cell numbers. The photon flux of lung metastatic tumor induced by Gankyrin knocking down 4T1-luc cell was 3.02 × 10(6), while that of lung metastasis induced by control cells was 10.9 × 10(6). The differences between two groups were significant. In pathology, Gankyrin was detected positive in lung metastasis tumors induced by control group. However, Gankyrin was negative in the Gankyrin knockdown group. CONCLUSIONS: Lentivirus infection may be effectively used to establish Gankyrin knocking down 4T1-luc cell model. Because of its involvement in the in vivo pulmonary metastasis of breast cancers, Gankyrin should be a novel target for tumor therapy.


Assuntos
Neoplasias Mamárias Experimentais/genética , Complexo de Endopeptidases do Proteassoma/genética , Proteínas Proto-Oncogênicas/genética , Animais , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica
4.
Proc Natl Acad Sci U S A ; 110(27): 11017-22, 2013 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-23776205

RESUMO

DNA damage triggers cell cycle arrest to provide a time window for DNA repair. Failure of arrest could lead to genomic instability and tumorigenesis. DNA damage-induced G1 arrest is generally achieved by the accumulation of Cyclin-dependent kinase inhibitor 1 (p21). However, p21 is degraded and does not play a role in UV-induced G1 arrest. The mechanism of UV-induced G1 arrest thus remains elusive. Here, we have identified a critical role for CUE domain-containing protein 2 (CUEDC2) in this process. CUEDC2 binds to and inhibits anaphase-promoting complex/cyclosome-Cdh1 (APC/C(Cdh1)), a critical ubiquitin ligase in G1 phase, thereby stabilizing Cyclin A and promoting G1-S transition. In response to UV irradiation, CUEDC2 undergoes ERK1/2-dependent phosphorylation and ubiquitin-dependent degradation, leading to APC/C(Cdh1)-mediated Cyclin A destruction, Cyclin-dependent kinase 2 inactivation, and G1 arrest. A nonphosphorylatable CUEDC2 mutant is resistant to UV-induced degradation. Expression of this stable mutant effectively overrides UV-induced G1-S block. These results establish CUEDC2 as an APC/C(Cdh1) inhibitor and indicate that regulated CUEDC2 degradation is critical for UV-induced G1 arrest.


Assuntos
Proteínas de Transporte/metabolismo , Pontos de Checagem da Fase G1 do Ciclo Celular/fisiologia , Proteínas de Membrana/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Ciclossomo-Complexo Promotor de Anáfase , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Ciclina A/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Pontos de Checagem da Fase G1 do Ciclo Celular/genética , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos da radiação , Células HEK293 , Células HeLa , Humanos , Sistema de Sinalização das MAP Quinases/genética , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Mutagênese Sítio-Dirigida , Fosforilação/genética , Complexos Ubiquitina-Proteína Ligase/genética , Raios Ultravioleta
5.
J Biol Chem ; 287(1): 382-392, 2012 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-22084247

RESUMO

Janus kinase 1/signal transducers and activators of transcription 3 (JAK1/STAT3) pathway is one of the recognized oncogenic signaling pathways that frequently overactivated in a variety of human tumors. Despite rapid progress in elucidating the molecular mechanisms of activation of JAK/STAT pathway, the processes that regulate JAK/STAT deactivation need to be further clarified. Here we demonstrate that CUE domain-containing 2 (CUEDC2) inhibits cytokine-induced phosphorylation of JAK1 and STAT3 and the subsequent STAT3 transcriptional activity. Further analysis by a yeast two-hybrid assay showed that CUEDC2 could engage in a specific interaction with a key JAK/STAT inhibitor, SOCS3 (suppressors of cytokine signaling 3). The interaction between CUEDC2 and SOCS3 is required for the inhibitory effect of CUEDC2 on JAK1 and STAT3 activity. Additionally, we found CUEDC2 functions collaboratively with SOCS3 to inhibit JAK1/STAT3 signaling by increasing SOCS3 stability via enhancing its association with Elongin C. Therefore, our findings revealed a new biological activity for CUEDC2 as the regulator of JAK1/STAT3 signaling and paved the way to a better understanding of the mechanisms by which SOCS3 has been linked to suppression of the JAK/STAT pathway.


Assuntos
Proteínas de Transporte/metabolismo , Janus Quinase 1/metabolismo , Proteínas de Membrana/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Linhagem Celular , Elonguina , Ativação Enzimática , Humanos , Fosforilação , Estabilidade Proteica , Proteólise , Proteína 3 Supressora da Sinalização de Citocinas , Fatores de Transcrição/metabolismo , Transcrição Gênica
6.
Nat Cell Biol ; 13(8): 924-33, 2011 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-21743465

RESUMO

Aneuploidy and chromosomal instability are major characteristics of human cancer. These abnormalities can result from defects in the spindle assembly checkpoint (SAC), which is a surveillance mechanism for accurate chromosome segregation through restraint of the activity of the anaphase-promoting complex/cyclosome (APC/C). Here, we show that a CUE-domain-containing protein, CUEDC2, is a cell-cycle regulator that promotes spindle checkpoint inactivation and releases APC/C from checkpoint inhibition. CUEDC2 is phosphorylated by Cdk1 during mitosis. Depletion of CUEDC2 causes a checkpoint-dependent delay of the metaphase-anaphase transition. Phosphorylated CUEDC2 binds to Cdc20, an activator of APC/C, and promotes the release of Mad2 from APC/C-Cdc20 and subsequent APC/C activation. CUEDC2 overexpression causes earlier activation of APC/C, leading to chromosome missegregation and aneuploidy. Interestingly, CUEDC2 is highly expressed in many types of tumours. These results suggest that CUEDC2 is a key regulator of mitosis progression, and that CUEDC2 dysregulation might contribute to tumour development by causing chromosomal instability.


Assuntos
Proteína Quinase CDC2/metabolismo , Proteínas de Transporte/metabolismo , Instabilidade Cromossômica/fisiologia , Proteínas de Membrana/metabolismo , Fuso Acromático/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Ciclossomo-Complexo Promotor de Anáfase , Aneuploidia , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas Cdc20 , Proteínas de Ciclo Celular/metabolismo , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Instabilidade Cromossômica/genética , Células HeLa , Humanos , Proteínas Mad2 , Mitose/genética , Mitose/fisiologia , Complexos Multiproteicos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Fosforilação , Proteínas Repressoras/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo
7.
Nat Med ; 17(6): 708-14, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21572428

RESUMO

Endocrine resistance is a major obstacle to hormonal therapy for breast cancers. Although reduced expression of estrogen receptor-α (ER-α) is a known contributing factor to endocrine resistance, the mechanism of ER-α downregulation in endocrine resistance is still not fully understood. Here we report that CUE domain-containing protein-2 (CUEDC2), a ubiquitin-binding motif-containing protein, is a key factor in endocrine resistance in breast cancer. We show that CUEDC2 modulates ER-α protein stability through the ubiquitin-proteasome pathway. Through the study of specimens from a large cohort of subjects with breast cancer, we found a strong inverse correlation between CUEDC2 and ER-α protein expression. Notably, subjects with tumors that highly expressed CUEDC2 had poor responsiveness to tamoxifen treatment and high potential for relapse. We further show that ectopic CUEDC2 expression impaired the responsiveness of breast cancer cells to tamoxifen. Therefore, our findings suggest that CUEDC2 is a crucial determinant of resistance to endocrine therapies in breast cancer.


Assuntos
Antineoplásicos Hormonais/uso terapêutico , Neoplasias da Mama/fisiopatologia , Proteínas de Transporte/fisiologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Proteínas de Membrana/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Neoplasias da Mama/tratamento farmacológico , Proteínas de Transporte/biossíntese , Linhagem Celular Tumoral , Regulação para Baixo , Receptor alfa de Estrogênio/metabolismo , Receptor alfa de Estrogênio/fisiologia , Feminino , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Quinase I-kappa B/metabolismo , Quinase I-kappa B/fisiologia , Proteínas de Membrana/biossíntese , Fosforilação , Receptores de Progesterona/metabolismo , Receptores de Progesterona/fisiologia , Tamoxifeno/uso terapêutico , Ubiquitinação
8.
J Clin Invest ; 120(8): 2829-41, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20628200

RESUMO

Activating mutations in Ras proteins are present in about 30% of human cancers. Despite tremendous progress in the study of Ras oncogenes, many aspects of the molecular mechanisms underlying Ras-induced tumorigenesis remain unknown. Through proteomics analysis, we previously found that the protein Gankyrin, a known oncoprotein in hepatocellular carcinoma, was upregulated during Ras-mediated transformation, although the functional consequences of this were not clear. Here we present evidence that Gankyrin plays an essential role in Ras-initiated tumorigenesis in mouse and human cells. We found that the increased Gankyrin present following Ras activation increased the interaction between the RhoA GTPase and its GDP dissociation inhibitor RhoGDI, which resulted in inhibition of the RhoA effector kinase Rho-associated coiled coil-containing protein kinase (ROCK). Importantly, Gankyrin-mediated ROCK inhibition led to prolonged Akt activation, a critical step in activated Ras-induced transformation and tumorigenesis. In addition, we found that Gankyrin is highly expressed in human lung cancers that have Ras mutations and that increased Gankyrin expression is required for the constitutive activation of Akt and tumorigenesis in these lung cancers. Our findings suggest that Gankyrin is a key regulator of Ras-mediated activation of Akt through inhibition of the downstream RhoA/ROCK pathway and thus plays an essential role in Ras-induced tumorigenesis.


Assuntos
Transformação Celular Neoplásica , Genes ras , Neoplasias Pulmonares/etiologia , Transdução de Sinais , Fatores de Transcrição/fisiologia , Proteínas rho de Ligação ao GTP/fisiologia , Quinases Associadas a rho/fisiologia , Animais , Inibidores de Dissociação do Nucleotídeo Guanina/fisiologia , Humanos , Camundongos , Células NIH 3T3 , PTEN Fosfo-Hidrolase/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho , Inibidores da Dissociação do Nucleotídeo Guanina rho-Específico , Proteína rhoA de Ligação ao GTP
9.
Proc Natl Acad Sci U S A ; 106(10): 3788-93, 2009 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-19234109

RESUMO

DNA damage response (DDR) acts as a tumorigenesis barrier, and any defects in the DDR machinery may lead to cancer. SOX4 expression is elevated in many types of tumors; however, its role in DDR is still largely unknown. Here, we show that SOX4, a new DNA damage sensor, is required for the activation of p53 tumor suppressor in response to DNA damage. Notably, SOX4 interacts with and stabilizes p53 protein by blocking Mdm2-mediated p53 ubiquitination and degradation. Furthermore, SOX4 enhances p53 acetylation by interacting with p300/CBP and facilitating p300/CBP/p53 complex formation. In concert with these results, SOX4 promotes cell cycle arrest and apoptosis, and it inhibits tumorigenesis in a p53-dependent manner. Therefore, these findings highlight SOX4 as a potential key factor in regulating DDR-associated cancer.


Assuntos
Dano ao DNA , Fatores de Transcrição SOXC/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Acetilação , Apoptose , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Ligação Proteica , Estabilidade Proteica , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Ubiquitinação
10.
Nat Immunol ; 9(5): 533-41, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18362886

RESUMO

Despite rapid progress in elucidating the molecular mechanisms of activation of the kinase IKK, the processes that regulate IKK deactivation are still unknown. Here we demonstrate that CUE domain-containing 2 (CUEDC2) interacted with IKKalpha and IKKbeta and repressed activation of the transcription factor NF-kappaB by decreasing phosphorylation and activation of IKK. Notably, CUEDC2 also interacted with GADD34, a regulatory subunit of protein phosphatase 1 (PP1). We found that IKK, CUEDC2 and PP1 existed in a complex and that IKK was released from the complex in response to inflammatory stimuli such as tumor necrosis factor. CUEDC2 deactivated IKK by recruiting PP1 to the complex. Therefore, CUEDC2 acts as an adaptor protein to target IKK for dephosphorylation and inactivation by recruiting PP1.


Assuntos
Proteínas de Transporte/metabolismo , Quinase I-kappa B/metabolismo , Proteínas de Membrana/metabolismo , Proteína Fosfatase 1/metabolismo , Proteínas Repressoras/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Proteínas de Transporte/imunologia , Domínio Catalítico , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Feminino , Humanos , Quinase I-kappa B/química , Inflamação/imunologia , Interleucina-6/biossíntese , Interleucina-6/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/genética , NF-kappa B/metabolismo , Fosforilação , Ligação Proteica , Proteínas Repressoras/imunologia , Regulação para Cima
11.
J Proteome Res ; 6(11): 4397-406, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17894482

RESUMO

A global understanding of ubiquitinated proteins in vivo is key to unraveling the biological significance of ubiquitination. There are, however, a few effective screening methods for rapid analysis of ubiquitinated proteins. In the current study, we designed a cell-based cDNA expression array combined with cell imaging for the rapid identification of polyubiquitinated proteins, which normally accumulate to form the unique "dot" structure following inhibition of ubiquitin proteasomes. The array consisted of 112 cDNAs encoding key components of major cellular pathways and potential targets of polyubiquitination. Among them, 40 proteins formed accumulation dots in response to proteasome inhibitor, MG-132, treatment. More importantly, 24 of those 40 proteins, such as MAPKAPK3, NLK, and RhoGDI2, are previously not known as the targets of ubiquitin. We further validated our findings by examining the endogenous counterparts of some of these proteins and found that those endogenous proteins form a similar "dot" structure. Immunoprecipitation assays confirmed that these accumulated proteins are polyubiquitinated. Our results demonstrate that this large-scale application of cell-based arrays represents a novel global approach in identifying candidates of the polyubiquitinated proteins. Therefore, the technique utilized here will facilitate future research on ubiquitination-regulated cell signaling.


Assuntos
Proteínas/química , Proteômica/métodos , Ubiquitina/química , Linhagem Celular Tumoral , DNA Complementar/metabolismo , Eletroforese em Gel Bidimensional , Inibidores de Dissociação do Nucleotídeo Guanina/metabolismo , Células HeLa , Humanos , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Leupeptinas/farmacologia , Inibidores de Proteassoma , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/metabolismo , Inibidor beta de Dissociação do Nucleotídeo Guanina rho , Inibidores da Dissociação do Nucleotídeo Guanina rho-Específico
12.
EMBO J ; 26(7): 1831-42, 2007 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-17347654

RESUMO

Accumulated evidence indicates that progesterone receptors (PR) are involved in proliferation of breast cancer cells and are implicated in the development of breast cancer. In this paper, a yeast two-hybrid screen for PR led to the identification of CUE domain containing 2 (CUEDC2), whose function is unknown. Our results demonstrate that CUEDC2 interacts with PR and promotes progesterone-induced PR degradation by the ubiquitin-proteasome pathway. The inhibition of endogenous CUEDC2 by siRNA nearly abrogated the progesterone-induced degradation of PR, suggesting that CUEDC2 is involved in progesterone-induced PR ubiquitination and degradation. Moreover, we identify the sumoylation site Lys-388 of PR as the target of CUEDC2-promoted ubiquitination. CUEDC2 decreases the sumoylation while promoting ubiquitination on Lys-388 of PRB. We also show that CUEDC2 represses PR transactivation, inhibits the ability of PR to stimulate rapid MAPK activity, and impairs the effect of progesterone on breast cancer cell growth. Therefore, our results identify a key post-translational mechanism that controls PR protein levels and for the first time provide an important insight into the function of CUEDC2 in breast cancer proliferation.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Membrana/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Processamento de Proteína Pós-Traducional , Receptores de Progesterona/metabolismo , Ubiquitina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proteínas de Transporte/genética , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Ligantes , Proteínas de Membrana/genética , Proteínas Mutantes/metabolismo , Progesterona/farmacologia , Ligação Proteica/efeitos dos fármacos , Mapeamento de Interação de Proteínas , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Estrutura Terciária de Proteína/efeitos dos fármacos , Receptores de Progesterona/genética , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/genética
13.
Nucleic Acids Res ; 34(19): 5552-66, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17020914

RESUMO

Progesterone receptor (PR) plays a critical role in cell proliferation and differentiation, and its transcriptional activity is known to be modulated by cofactor proteins. In the present study, we demonstrated that in the presence of progesterone, protein inhibitor of activated STAT-3 (PIAS3) significantly inhibited the PR transcriptional activity and the expression of progesterone-responsive genes. Reduction of endogenous PIAS3 by PIAS3 small-interfering RNA enhanced PR transactivation in a ligand-dependent manner. PIAS3 interacted with PR both in vitro and in vivo and the interaction was enhanced by progesterone. Furthermore, our findings suggested that PIAS3 strongly induced PRB sumoylation at three sites, Lys-7, Lys-388 and Lys-531. In addition, novel roles in PRB nuclear retention and transactivation were identified for these sites. Our data also suggested that PIAS3 was recruited in a largely hormone-dependent manner in response to a progesterone-responsive promoter. Finally, we demonstrated that PIAS3 inhibited the DNA-binding activity of PR and influenced its nuclear export as well as PR transactivation. Taken together, these data strongly suggested that PIAS3 played an important physiological role in PR function.


Assuntos
Núcleo Celular/química , Chaperonas Moleculares/metabolismo , Proteínas Inibidoras de STAT Ativados/metabolismo , Receptores de Progesterona/metabolismo , Ativação Transcricional , Animais , Humanos , Progesterona/antagonistas & inibidores , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional , Receptores de Progesterona/análise , Receptores de Progesterona/antagonistas & inibidores , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo
14.
J Proteome Res ; 5(10): 2815-23, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17022653

RESUMO

We implemented a proteomics approach to the systematical analysis of the alterations in the proteome of NIH3T3 cells transformed by oncogenic H-RasV12. Forty-four proteins associated with Ras-mediated transformation have been identified, and 28 proteins were not previously reported. RT-PCR analysis showed that approximately 44% of target proteins identified showed concomitant changes in mRNA abundance. A principal finding was the up-regulation of gankyrin, which was the first evidence to show that gankyrin pathway was implicated in Ras-activated transformation.


Assuntos
Transformação Celular Neoplásica/metabolismo , Proteína Oncogênica p21(ras)/genética , Proteoma/análise , Proteômica , Animais , Western Blotting , Transformação Celular Neoplásica/genética , Análise por Conglomerados , Humanos , Camundongos , Células NIH 3T3 , Proteoma/genética , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica , Transfecção
15.
Biochem Biophys Res Commun ; 345(1): 355-61, 2006 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-16678796

RESUMO

Recent reports have shown that MDM2 may attenuate hypertrophy of cardiac myocytes. However, mechanism of MDM2 involving in this process is unclear. In this study, we identified a novel specific MDM2-binding protein TCAP by the yeast two-hybrid screen. It was validated by GST pull-down and co-immunoprecipitation assays. Confocal analysis showed that MDM2 and TCAP co-localized in the nucleus, and elevated MDM2 expression could alter the subcellular localization of TCAP. Notably, MDM2 downregulated the protein level of TCAP through the proteasomal pathway, and this downregulation was inhibited by p14(ARF). In addition, our results suggested that the degradation of TCAP by MDM2 was through the ubiquitin-independent pathway. Given that TCAP is a key component involving in the cardiac hypertrophy, the degradation of TCAP by MDM2 might be connected with the roles of MDM2 in cardiac hypertrophy. Further investigation will focus on the biological significance of MDM2-TCAP interaction in cardiac hypertrophy.


Assuntos
Proteínas Musculares/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Sarcômeros/metabolismo , Frações Subcelulares/metabolismo , Conectina , Regulação para Baixo , Proteínas Musculares/química , Ligação Proteica , Mapeamento de Interação de Proteínas , Proteínas Proto-Oncogênicas c-mdm2/química , Sarcômeros/química , Frações Subcelulares/química
16.
J Biol Chem ; 279(47): 49251-8, 2004 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-15371445

RESUMO

Although eukaryotic translation initiation factor 5A (eIF5A) was originally designated as an "initiation factor," recent data have shown it to be also involved in apoptosis. However, the actual function of eIF5A in apoptosis is still unknown. In this study, we performed yeast two-hybrid screens to identify eIF5A-interacting proteins to help us understand the mechanisms of eIF5A. Our results demonstrated that eIF5A and syntenin could engage in a specific interaction both in vitro and in vivo and functioned collaboratively to regulate p53 activity. Our findings, for the first time, revealed a new biological activity for eIF5A as the regulator of p53. Overexpression of eIF5A or its EFP domain resulted in up-regulation of p53, and silencing eIF5A by small interfering RNA reduced the p53 protein level. Further analysis by reverse transcription PCR showed eIF5A-activated p53 transcription. The effect of eIF5A on p53 transcriptional activity was further demonstrated by the increasing expressions of p21 and Bax, well known target genes of p53. In contrast, a point mutant of eIF5A, hypusination being abolished, was revealed to be functionally defective in p53 up-regulation. Overexpression of eIF5A led to a p53-dependent apoptosis or sensitized cells to induction of apoptosis by chemotherapeutic agents. However, when eIF5A interacted with its novel partner, syntenin, the eIF5A-induced increase in p53 protein level was significantly inhibited. Therefore, eIF5A seems to be a previously unrecognized regulator of p53 that may define a new pathway for p53-dependent apoptosis, and syntenin might regulate p53 by balancing the regulation of eIF5A signaling to p53 for apoptosis.


Assuntos
Apoptose , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas de Membrana/fisiologia , Fatores de Iniciação de Peptídeos/fisiologia , Proteínas de Ligação a RNA/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Western Blotting , Células COS , Linhagem Celular , Linhagem Celular Tumoral , DNA Complementar/metabolismo , Citometria de Fluxo , Transferência Ressonante de Energia de Fluorescência , Regulação da Expressão Gênica , Inativação Gênica , Glutationa Transferase/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imunoprecipitação , Mutação , Fatores de Iniciação de Peptídeos/química , Fosforilação , Plasmídeos/metabolismo , Mutação Puntual , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/química , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sinteninas , Fatores de Tempo , Transfecção , Técnicas do Sistema de Duplo-Híbrido , Regulação para Cima , Proteína X Associada a bcl-2 , Fator de Iniciação de Tradução Eucariótico 5A
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...