Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Nat Med ; 78(2): 285-295, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38082192

RESUMO

The natural product Honokiol exhibits robust antitumor activity against a range of cancers, and it has also received approval to undergo phase I clinical trial testing. We confrmed that honokiol can promote the apoptotic death of tumor cells through cell experiments. Then siRNA constructs specific for PIAS3, PIAS3 overexpression plasmid and the mutation of the STAT3 Tyr705 residue were used to confirm the mechanism of Honokiol-induced apoptosis. Finally, we confrmed that honokiol can promote PIAS3 upregulation, in turn suppressing STAT3 Tyr705 phosphorylation through the in vivo and in vitro experiments. Honokiol was ultimately found to reduce tumor cell viability by promoting apoptosis through a mechanism dependent on the ability of Honokiol to promote PIAS3 upregulation and the selective inhibition of p-STAT3 (Tyr705) without affecting p-STAT3 (Ser727) or p-STAT1 (Tyr701) levels. PIAS3 knockdown and overexpression in tumor cells altered STAT3 activation and associated DNA binding activity through the control of Tyr705 phosphorylation via PIAS3-STAT3 complex formation, ultimately shaping Honokiol-induced tumor cell apoptosis. Honokiol was also confirmed to significantly prolong the survival of mice bearing xenograft tumors in a PIAS3-dependent fashion. Together, these findings highlight a novel pathway through which Honokiol can promote PIAS3 upregulation, in turn suppressing STAT3 Tyr705 phosphorylation and promoting the apoptotic death of tumor cells.


Assuntos
Compostos Alílicos , Apoptose , Compostos de Bifenilo , Fenóis , Tirosina , Humanos , Animais , Camundongos , Fosforilação , Regulação para Cima , Linhagem Celular Tumoral , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Proteínas Inibidoras de STAT Ativados/genética , Proteínas Inibidoras de STAT Ativados/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo
2.
PLoS Genet ; 16(11): e1009106, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33151932

RESUMO

Hirschsprung disease (HSCR, OMIM 142623) involves congenital intestinal obstruction caused by dysfunction of neural crest cells and their progeny during enteric nervous system (ENS) development. HSCR is a multifactorial disorder; pathogenetic variants accounting for disease phenotype are identified only in a minority of cases, and the identification of novel disease-relevant genes remains challenging. In order to identify and to validate a potential disease-causing relevance of novel HSCR candidate genes, we established a complementary study approach, combining whole exome sequencing (WES) with transcriptome analysis of murine embryonic ENS-related tissues, literature and database searches, in silico network analyses, and functional readouts using candidate gene-specific genome-edited cell clones. WES datasets of two patients with HSCR and their non-affected parents were analysed, and four novel HSCR candidate genes could be identified: ATP7A, SREBF1, ABCD1 and PIAS2. Further rare variants in these genes were identified in additional HSCR patients, suggesting disease relevance. Transcriptomics revealed that these genes are expressed in embryonic and fetal gastrointestinal tissues. Knockout of these genes in neuronal cells demonstrated impaired cell differentiation, proliferation and/or survival. Our approach identified and validated candidate HSCR genes and provided further insight into the underlying pathomechanisms of HSCR.


Assuntos
Doença de Hirschsprung/genética , Membro 1 da Subfamília D de Transportadores de Cassetes de Ligação de ATP/genética , Animais , Diferenciação Celular/genética , Linhagem Celular , Proliferação de Células/genética , Sobrevivência Celular/genética , Simulação por Computador , ATPases Transportadoras de Cobre/genética , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Lactente , Masculino , Camundongos , Proteínas Inibidoras de STAT Ativados/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Sequenciamento do Exoma
3.
Br J Cancer ; 117(12): 1777-1786, 2017 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-29123263

RESUMO

BACKGROUND: Although chemotherapy is the cornerstone treatment for patients with metastatic colorectal cancer (mCRC), acquired chemoresistance is common and constitutes the main reason for treatment failure. Monoclonal antibodies against insulin-like growth factor-1 receptor (IGF-1R) have been tested in pre-treated mCRC patients, but results have been largely deceiving. METHODS: We analysed time to progression, overall survival, and the mutational status of RAS, BRAF and nuclear p-IGF-1R expression by immunohistochemistry, in 470 metastatic CRC patients. The effect of IGF-1R activation and distribution was also assessed using cellular models of CRC and RNAi for functional validation. RESULTS: Nuclear IGF-1R increased in metastatic tumours compared to paired untreated primary tumours, and significantly correlated with poor overall survival in mCRC patients. In vitro, chemo-resistant cell lines presented significantly higher levels of IGF-1R expression within the nuclear compartment, and PIAS3, a protein implicated also in the sumoylation process of intranuclear proteins, contributed to IGF-1R nuclear sequestration, highlighting the essential role of PIAS3 in this process. Intriguingly, we observed that ganitumab, an IGF-1R blocking-antibody used in several clinical trials, and dasatinib, an SRC inhibitor, increased the nuclear localisation of IGF-1R. CONCLUSIONS: Our study demonstrates that IGF-1R nuclear location might lead to chemotherapy and targeted agent resistance.


Assuntos
Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Núcleo Celular/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Transporte Proteico/efeitos dos fármacos , Receptor IGF Tipo 1/metabolismo , Idoso , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Bevacizumab/administração & dosagem , Camptotecina/administração & dosagem , Camptotecina/análogos & derivados , Sobrevivência Celular/efeitos dos fármacos , Cetuximab/administração & dosagem , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Curcumina/farmacologia , Dasatinibe/farmacologia , Ácidos Graxos Insaturados/farmacologia , Feminino , Fluoruracila/administração & dosagem , Fluoruracila/farmacologia , Inativação Gênica , Células HCT116 , Células HT29 , Humanos , Leucovorina/administração & dosagem , Masculino , Pessoa de Meia-Idade , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Terapia de Alvo Molecular , Niacinamida/análogos & derivados , Niacinamida/farmacologia , Compostos Organoplatínicos/administração & dosagem , Compostos Organoplatínicos/farmacologia , Oxaliplatina , Panitumumabe , Compostos de Fenilureia/farmacologia , Proteínas Inibidoras de STAT Ativados/genética , Proteínas Inibidoras de STAT Ativados/metabolismo , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Pirimidinas/farmacologia , Pirróis/farmacologia , Transdução de Sinais/efeitos dos fármacos , Sorafenibe
4.
Artif Cells Nanomed Biotechnol ; 44(3): 859-64, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-25650648

RESUMO

Research studies in recent years have found that isoquercetin has an inhibiting effect on multiple carcinogens, but research studies filed on isoquercetin in bladder cancer are quite few. This paper observed the influence of isoquercetin on biological activity of the EJ cell of bladder cancer through HC dyeing and trypan blue counting, studied the EJ cell cycle by flow cytometry (FCM), and then analyzed the influence of isoquercetin and its effect on the protein expression of STAT3 and STAT3-inhibiting factors (PIAS3) in EJ cells. Research has shown that isoquercetin has an inhibitory effect on the EJ cells of bladder cancer, but it is not obvious.


Assuntos
Plantas Medicinais/química , Quercetina/análogos & derivados , Neoplasias da Bexiga Urinária/tratamento farmacológico , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Chaperonas Moleculares/biossíntese , Proteínas de Neoplasias/biossíntese , Proteínas Inibidoras de STAT Ativados/biossíntese , Quercetina/química , Quercetina/isolamento & purificação , Quercetina/farmacologia , Fator de Transcrição STAT3/biossíntese , Neoplasias da Bexiga Urinária/metabolismo
5.
Toxicol Appl Pharmacol ; 275(3): 244-56, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24486434

RESUMO

Non-receptor protein tyrosine kinases (NRPTKs)-dependent inflammatory signal transduction cascades play key roles in immunoregulation. However, drug intervention through NRPTKs-involved immunoregulation mechanism in microglia (the major immune cells of the central nervous system) has not been widely investigated. A main aim of the present study is to elucidate the contribution of two major NRPTKs (Syk and Jak2) in neuroinflammation suppression by a bioactive sesquiterpene dimmer (DSF-27). We found that LPS-stimulated BV-2 cells activated Syk and further initiated Akt/NF-κB inflammatory pathway. This Syk-dependent Akt/NF-κB inflammatory pathway can be effectively ameliorated by DSF-27. Moreover, Jak2 was activated by LPS, which was followed by transcriptional factor Stat3 activation. The Jak2/Stat3 signal was suppressed by DSF-27 through inhibition of Jak2 and Stat3 phosphorylation, promotion of Jak/Stat3 inhibitory factors PIAS3 expression, and down-regulation of ERK and p38 MAPK phosphorylation. Furthermore, DSF-27 protected cortical and mesencephalic dopaminergic neurons against neuroinflammatory injury. Taken together, our findings indicate NRPTK signaling pathways including Syk/NF-κB and Jak2/Stat3 cascades are potential anti-neuroinflammatory targets in microglia, and may also set the basis for the use of sesquiterpene dimmer as a therapeutic approach for neuroinflammation via interruption of these pathways.


Assuntos
Anti-Inflamatórios/farmacologia , Mediadores da Inflamação/metabolismo , Inflamação/prevenção & controle , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Janus Quinase 2/antagonistas & inibidores , Microglia/efeitos dos fármacos , Proteínas Tirosina Quinases/antagonistas & inibidores , Sesquiterpenos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Anti-Inflamatórios/química , Artemisia/química , Linhagem Celular , Técnicas de Cocultura , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/enzimologia , Neurônios Dopaminérgicos/imunologia , Relação Dose-Resposta a Droga , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Inflamação/enzimologia , Inflamação/genética , Inflamação/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Janus Quinase 2/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Microglia/enzimologia , Microglia/imunologia , Modelos Moleculares , Simulação de Acoplamento Molecular , Estrutura Molecular , NF-kappa B/metabolismo , Fosforilação , Proteínas Inibidoras de STAT Ativados/metabolismo , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Fator de Transcrição STAT3 , Sesquiterpenos/química , Quinase Syk , Fatores de Tempo , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
Mol Cell Biol ; 27(10): 3780-92, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17353273

RESUMO

As a multifunctional protein, Yin Yang 1 (YY1) has been demonstrated to regulate both gene expression and protein posttranslational modifications. However, gaps still exist in our knowledge of how YY1 can be modified and what the consequences of its modifications are. Here we report that YY1 protein can be sumoylated both in vivo and in vitro. We have identified lysine 288 as the major sumoylation site of YY1. We also discovered that PIASy, a SUMO E3 ligase, is a novel YY1-interacting protein and can stimulate the sumoylation of YY1 both in vitro and in vivo. Importantly, the effects of PIASy mutants on in vivo YY1 sumoylation correlate with the YY1-PIASy interaction but do not depend on the RING finger domain of PIASy. This regulation is unique to YY1 sumoylation because PIASy-mediated p53 sumoylation still relies on the integrity of PIASy, which is also true of all of the previously identified substrates of PIASy. In addition, PIASy colocalizes with YY1 in the nucleus, stabilizes YY1 in vivo, and differentially regulates YY1 transcriptional activity on different target promoters. This study demonstrates that YY1 is a target of SUMOs and reveals a novel feature of a SUMO E3 ligase in the PIAS family that selectively stimulates protein sumoylation independent of the RING finger domain.


Assuntos
Proteínas Inibidoras de STAT Ativados/metabolismo , Proteína SUMO-1/metabolismo , Fator de Transcrição YY1/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Regulação da Expressão Gênica , Humanos , Lisina/metabolismo , Dados de Sequência Molecular , Proteínas de Ligação a Poli-ADP-Ribose , Proteínas Inibidoras de STAT Ativados/genética , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteína SUMO-1/genética , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/metabolismo , Fator de Transcrição YY1/genética
7.
J Neuroimmunol ; 173(1-2): 12-22, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16494952

RESUMO

Functional interactions between neuroendocrine and immune systems are mediated by similar ligands and receptors, which establish a bi-directional communication that is relevant for homeostasis. We investigated herein the hypothalamus-pituitary-adrenal (HPA) axis in mice acutely infected by Trypanosoma cruzi, the causative agent of Chagas' disease. Parasites were seen in the adrenal gland, whereas T. cruzi specific PCR gene amplification product was found in both adrenal and pituitary glands of infected mice. Histological and immunohistochemical analyses of pituitary and adrenal glands of infected animals revealed several alterations including vascular stasis, upregulation of the extracellular matrix proteins fibronectin and laminin, as well as T cell and macrophage infiltration. Functionally, we detected a decrease in CRH and an increase in corticosterone contents, in hypothalamus and serum respectively. In contrast, we did not find significant changes in the amounts of ACTH in sera of infected animals, whereas the serum levels of the glucocorticoid-stimulating cytokine, IL-6 (interleukin-6), were increased as compared to controls. When we analyzed the effects of T. cruzi in ACTH-producing AtT-20 cell line, infected cultures presented lower levels of ACTH and pro-opiomelanocortin production when compared to controls. In these cells we observed a strong phosphorylation of STAT-3, together with an increased synthesis of IL-6, suppressor of cytokine signaling 3 (SOCS-3) and inhibitor of activated STAT-3 (PIAS-3), which could explain the partial blockage of ACTH production. In conclusion, our data reveal that the HPA axis is altered during acute T. cruzi infection, suggesting direct and indirect influences of the parasite in the endocrine homeostasis.


Assuntos
Doença de Chagas/fisiopatologia , Sistema Hipotálamo-Hipofisário/microbiologia , Sistema Hipófise-Suprarrenal/microbiologia , Glândulas Suprarrenais/microbiologia , Glândulas Suprarrenais/fisiologia , Hormônio Adrenocorticotrópico/análise , Hormônio Adrenocorticotrópico/metabolismo , Animais , Corticosterona/análise , Corticosterona/metabolismo , Hormônio Liberador da Corticotropina/análise , Hormônio Liberador da Corticotropina/metabolismo , Sistema Hipotálamo-Hipofisário/fisiologia , Hipotálamo/microbiologia , Hipotálamo/fisiologia , Immunoblotting , Imuno-Histoquímica , Interleucina-6/análise , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Hipófise/microbiologia , Hipófise/fisiologia , Sistema Hipófise-Suprarrenal/metabolismo , Proteínas Inibidoras de STAT Ativados/análise , Proteínas Inibidoras de STAT Ativados/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/análise , Fator de Transcrição STAT3/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/análise , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Trypanosoma cruzi
8.
J Biol Chem ; 280(46): 38153-9, 2005 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-16144832

RESUMO

Group III presynaptic metabotropic glutamate receptors (mGluRs) play a central role in regulating presynaptic activity through G-protein effects on ion channels and signal transducing enzymes. Like all Class C G-protein-coupled receptors, mGluR8 has an extended intracellular C-terminal domain (CTD) presumed to allow for modulation of downstream signaling. In a yeast two-hybrid screen of an adult rat brain cDNA library with the CTDs of mGluR8a and 8b (mGluR8-C) as baits, we identified sumo1 and four different components of the sumoylation cascade (ube2a, Pias1, Piasgamma, Piasxbeta) as interacting proteins. Binding assays using recombinant GST fusion proteins confirmed that Pias1 interacts not only with mGluR8-C but also with all group III mGluR CTDs. Pias1 binding to mGluR8-C required a region N-terminal to a consensus sumoylation motif and was not affected by arginine substitution of the conserved lysine 882 within this motif. Co-transfection of fluorescently tagged mGluR8a-C, sumo1, and enzymes of the sumoylation cascade into HEK293 cells showed that mGluR8a-C can be sumoylated in vivo. Arginine substitution of lysine 882 within the consensus sumoylation motif, but not other conserved lysines within the CTD, abolished in vivo sumoylation. Our results are consistent with post-translational sumoylation providing a novel mechanism of group III mGluR regulation.


Assuntos
Proteínas Inibidoras de STAT Ativados/fisiologia , Receptores de Glutamato Metabotrópico/química , Receptores de Glutamato Metabotrópico/fisiologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/fisiologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Arginina/química , Sítios de Ligação , Encéfalo/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , DNA Complementar/metabolismo , Eletroforese em Gel de Poliacrilamida , Glutationa/química , Glutationa/metabolismo , Glutationa Transferase/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Canais Iônicos/metabolismo , Lisina/química , Dados de Sequência Molecular , Ligação Proteica , Proteínas Inibidoras de STAT Ativados/química , Proteínas Inibidoras de STAT Ativados/metabolismo , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Ratos , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Sefarose/química , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Transfecção , Técnicas do Sistema de Duplo-Híbrido , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
9.
Cell Cycle ; 4(2): 242-5, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15655377

RESUMO

Multiple myeloma (MM) remains largely incurable despite conventional and high-dose therapies. Therefore, novel biologically based treatment approaches are urgently required. Particularly, STAT3 activated by IL-6 has a key role in preventing apoptosis and stimulating growth of multiple myeloma cells. Nuclear receptors, a distinct class of ligand-activated transcriptional factors, can interact and modify the function of transcriptional factors intrinsic to the cytokine signal transduction pathways. We have investigated regulation of two nuclear receptors, peroxisome proliferator-activated receptor gamma (PPARgamma) and estrogen receptor (ER), and their crosstalk with STAT3 in multiple myeloma. These results indicate that ligand-activated nuclear receptors can function as negative modulators of STAT3 through direct mechanisms, or in turn, by facilitating coregulators such as PIAS or SMRT. Therefore, different classes of nuclear receptors affect suppression of STAT3 functions through diverse mechanisms resulting in downregulating IL-6-mediated cell growth and gene expression. Given the importance of IL-6 in multiple myeloma, the estrogen receptor-STAT3 or PPARgamma-STAT3 interaction may have significant therapeutic implications in multiple myeloma.


Assuntos
Mieloma Múltiplo/fisiopatologia , PPAR gama/fisiologia , Receptores de Estrogênio/fisiologia , Fator de Transcrição STAT3/fisiologia , Animais , Apoptose/genética , Apoptose/fisiologia , Proliferação de Células , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Regulação Neoplásica da Expressão Gênica , Humanos , Interleucina-6/fisiologia , Mieloma Múltiplo/genética , Correpressor 2 de Receptor Nuclear , PPAR gama/genética , Proteínas Inibidoras de STAT Ativados/genética , Proteínas Inibidoras de STAT Ativados/fisiologia , Receptor Cross-Talk , Receptores de Estrogênio/genética , Proteínas Repressoras/genética , Proteínas Repressoras/fisiologia , Fator de Transcrição STAT3/genética , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia
10.
J Biol Chem ; 280(6): 5004-12, 2005 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-15561718

RESUMO

Small ubiquitin-like modifier (SUMO) regulates diverse cellular processes through its reversible, covalent attachment to target proteins. Many SUMO substrates are involved in transcription and chromatin structure. Sumoylation appears to regulate the functions of target proteins by changing their subcellular localization, increasing their stability, and/or mediating their binding to other proteins. Using an in vitro expression cloning approach, we have identified 40 human SUMO1 substrates. The spectrum of human SUMO1 substrates identified in our screen suggests general roles of sumoylation in transcription, chromosome structure, and RNA processing. We have validated the sumoylation of 24 substrates in living cells. Analysis of this panel of SUMO substrates leads to the following observations. 1) Sumoylation is more efficient in vitro than in living cells. Polysumoylation occurs on several substrates in vitro. 2) SUMO isopeptidases have little substrate specificity. 3) The SUMO ligases, PIAS1 and PIASxbeta, have broader substrate specificities than does PIASy. 4) Although SUMO1 and SUMO2 are equally efficiently conjugated to a given substrate in vitro, SUMO1 conjugation is more efficient in vivo. 5) Most SUMO substrates localize to the nucleus, and sumoylation does not generally affect their subcellular localization. Therefore, sumoylation appears to regulate the functions of its substrates through multiple, context-dependent mechanisms.


Assuntos
Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , DNA/química , DNA Complementar/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Fatores de Transcrição Kruppel-Like , Lisina/química , Microscopia de Fluorescência , Plasmídeos/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose , Reação em Cadeia da Polimerase , Ligação Proteica , Proteínas Inibidoras de STAT Ativados , Proteínas/química , RNA/química , Proteína SUMO-1 , Especificidade por Substrato , Fatores de Tempo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Transfecção
11.
J Biol Chem ; 278(30): 27853-63, 2003 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-12740389

RESUMO

Transforming growth factor-beta (TGF-beta) and TGF-beta-related factors regulate cell growth, differentiation, and apoptosis, and play key roles in normal development and tumorigenesis. TGF-beta family-induced changes in gene expression are mediated by serine/threonine kinase receptors at the cell surface and Smads as intracellular effectors. Receptor-activated Smads combine with a common Smad4 to translocate into the nucleus where they cooperate with other transcription factors to activate or repress transcription. The activities of the receptor-activated Smads are controlled by post-translational modifications such as phosphorylation and ubiquitylation. Here we show that Smad4 is modified by sumoylation. Sumoylation of Smad4 was enhanced by the conjugating enzyme Ubc9 and members of the PIAS family of SUMO ligases. A major sumoylation site in Smad4 was localized to Lys-159 in its linker segment with an additional site at Lys-113 in the MH-1 domain. Increased sumoylation in the presence of the PIASy E3 ligase correlated with targeting of Smad4 to subnuclear speckles that contain SUMO-1 and PIASy. Replacement of lysines 159 and 113 by arginines or increased sumoylation enhanced the stability of Smad4, and transcription in mammalian cells and Xenopus embryos. These observations suggest a role for Smad4 sumoylation in the regulation of TGF-beta signaling through Smads.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Proteína SUMO-1/metabolismo , Transdução de Sinais , Transativadores/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Enzimas de Conjugação de Ubiquitina , Proteínas de Xenopus , Transporte Ativo do Núcleo Celular , Animais , Arginina/química , Western Blotting , Células COS , Proteínas de Transporte/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células HeLa , Humanos , Ligases/metabolismo , Lisina/química , Microscopia de Fluorescência , Fatores de Crescimento Neural , Plasmídeos/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose , Testes de Precipitina , Proteínas Inibidoras de STAT Ativados , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , RNA Complementar/metabolismo , Proteínas Smad , Proteína Smad4 , Transativadores/metabolismo , Transcrição Gênica , Ativação Transcricional , Transfecção , Células Tumorais Cultivadas , Ubiquitina-Proteína Ligases , Xenopus
12.
J Biol Chem ; 277(51): 50131-6, 2002 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-12393906

RESUMO

Mdm2, a ubiquitin ligase that acts on p53, is regulated by sumoylation. In the current study, we identify the enzymes responsible for the sumoylation of Mdm2. When mammalian cells are co-transfected with cDNAs encoding Mdm2 and PIAS1 or PIASxbeta (protein inhibitor of activated STAT) as sumoylation enzymes, Mdm2 is highly sumoylated. Mdm2 is also sumoylated in an in vitro system containing PIASxbeta, PIAS1, and RanBP2. When several lysine residues of Mdm2 were sequentially mutated to arginine, the K182R mutant was not sumoylated in intact cells; however, in the in vitro system this mutant was sumoylated by PIAS1, PIASxbeta, and RanBP2 as efficiently as the wild-type Mdm2 protein. Lysine residues 182 and 185 map within the nuclear localization signal of Mdm2. A K185R mutant of Mdm2 is sumoylated in intact cells, whereas a K182R protein is not. Only a Mdm2 protein bearing the K182R mutation is localized exclusively in the cytoplasm. Because RanBP2 is a nuclear pore protein and PIAS proteins are localized within the nucleus, our data suggest that Mdm2 is sumoylated during nuclear translocation by RanBP2 and then further sumoylated once in the nucleus by PIASxbeta and PIAS1.


Assuntos
Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Proteínas Nucleares , Proteínas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transporte Ativo do Núcleo Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , DNA Complementar/metabolismo , Humanos , Lisina/química , Lisina/metabolismo , Microscopia de Fluorescência , Chaperonas Moleculares , Mutação , Plasmídeos/metabolismo , Ligação Proteica , Proteínas Inibidoras de STAT Ativados , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-mdm2 , Proteínas Recombinantes/metabolismo , Transfecção , Células Tumorais Cultivadas
13.
J Biol Chem ; 277(10): 8255-9, 2002 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-11788578

RESUMO

The tumor suppressor protein p53 functions as a transcriptional factor that activates genes controlling cell cycle arrest and apoptosis. Here, we report that protein inhibitor of activated Stat1 (PIAS1) interacts with the tetramerization and C-terminal regulatory domains of p53 in yeast two-hybrid analyses. Endogenous PIAS1 is also associated with endogenous p53 in mammalian cells. Ectopic expression of PIAS1 activates p53-mediated expression in mouse embryonic fibroblast cells (p53(-/-)) as well as a variety of other cell lines. Furthermore, ectopic expression of PIAS1 induces p53-mediated expression of cyclin-dependent kinase inhibitor p21 and G(1) arrest of the cell cycle in H1299 cells. In addition, a PIAS1 mutant without the RING-finger domain required for sumoylation could still activate p53-mediated gene expression, indicating that activation of p53 by PIAS1 does not require the RING-finger domain. Taken together, our results suggest that PIAS1 is a novel activator of p53.


Assuntos
Proteínas/química , Proteínas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Western Blotting , Ciclo Celular , Linhagem Celular , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , DNA Complementar/metabolismo , Fibroblastos/metabolismo , Fase G1 , Genes Reporter , Genes p53/genética , Glucose-6-Fosfato/metabolismo , Células HeLa , Humanos , Luciferases/metabolismo , Camundongos , Plasmídeos/metabolismo , Testes de Precipitina , Ligação Proteica , Proteínas Inibidoras de STAT Ativados , Estrutura Terciária de Proteína , Transcrição Gênica , Transfecção , Células Tumorais Cultivadas , Técnicas do Sistema de Duplo-Híbrido
14.
Mol Pharmacol ; 60(5): 972-80, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11641425

RESUMO

The antithyroid drug, methimazole (MMI) is used to treat patients with Graves' hyperthyroidism. The major action of MMI is to inhibit synthesis of thyroid hormone in the thyroid gland. However, MMI also has antioxidant and immunomodulatory effects on thyrocytes and/or immune cells. This study identifies novel antioxidant and immunomodulatory effects of MMI involving the interferon-gamma (IFN-gamma) signaling pathway in thyroid cells. MMI inhibits transcription of the intercellular adhesion molecule-1 (ICAM-1) gene by modulating the function of transcription factor STAT1 (signal transducer and activator of transcription 1), which binds to the IFN-gamma activated site of the ICAM-1 promoter. Furthermore, MMI rapidly eliminates H(2)O(2) produced by IFN-gamma treatment in thyroid cells and thus inhibits the H(2)O(2)-mediated phosphorylation of tyrosine 701 in STAT1. MMI also eliminates H(2)O(2) in vitro. MMI facilitates electron transfer from NADPH to H(2)O(2) using thioredoxin or glutathione, fulfilling a role similar to peroxiredoxin or glutathione peroxidase, respectively. MMI prevents the IFN-gamma and H(2)O(2)-mediated reversible inactivation of phosphatases. These effects inhibit full activation of the IFN-gamma-induced Janus kinase(JAK)/STAT signaling pathway in FRTL-5 thyroid cells. These results may in part explain the antioxidant and immunomodulatory effects of MMI in thyroid cells of Graves' disease patients.


Assuntos
Antioxidantes/farmacologia , Expressão Gênica/efeitos dos fármacos , Peróxido de Hidrogênio/metabolismo , Interferon gama/farmacologia , Metimazol/farmacologia , Proteínas , Proteínas Proto-Oncogênicas , Proteínas Repressoras , Glândula Tireoide/efeitos dos fármacos , Fatores de Transcrição , Adjuvantes Imunológicos/farmacologia , Animais , Proteínas de Transporte/biossíntese , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Interações Medicamentosas , Transporte de Elétrons/efeitos dos fármacos , Peróxido de Hidrogênio/farmacologia , Molécula 1 de Adesão Intercelular/biossíntese , Molécula 1 de Adesão Intercelular/genética , Interferon gama/antagonistas & inibidores , Janus Quinase 1 , Janus Quinase 2 , Cinética , Peroxidases/farmacologia , Peroxirredoxinas , Fosforilação/efeitos dos fármacos , Biossíntese de Proteínas , Proteínas Inibidoras de STAT Ativados , Proteínas Tirosina Quinases/metabolismo , Ratos , Fator de Transcrição STAT1 , Fator de Transcrição STAT3 , Transdução de Sinais/fisiologia , Proteína 1 Supressora da Sinalização de Citocina , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina , Glândula Tireoide/citologia , Glândula Tireoide/metabolismo , Transativadores/metabolismo , Tirosina/metabolismo
15.
Proc Natl Acad Sci U S A ; 95(18): 10626-31, 1998 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9724754

RESUMO

STAT (signal transducer and activator of transcription) proteins are latent cytoplasmic transcription factors that become activated by tyrosine phosphorylation in response to cytokine stimulation. Tyrosine phosphorylated STATs dimerize and translocate into the nucleus to activate specific genes. Different members of the STAT protein family have distinct functions in cytokine signaling. Biochemical and genetic analysis has demonstrated that Stat1 is essential for gene activation in response to interferon stimulation. Although progress has been made toward understanding STAT activation, little is known about how STAT signals are down-regulated. We report here the isolation of a family of PIAS (protein inhibitor of activated STAT) proteins. PIAS1, but not other PIAS proteins, blocked the DNA binding activity of Stat1 and inhibited Stat1-mediated gene activation in response to interferon. Coimmunoprecipitation analysis showed that PIAS1 was associated with Stat1 but not Stat2 or Stat3 after ligand stimulation. The in vivo PIAS1-Stat1 interaction requires phosphorylation of Stat1 on Tyr-701. These results identify PIAS1 as a specific inhibitor of Stat1-mediated gene activation and suggest that there may exist a specific PIAS inhibitor in every STAT signaling pathway.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica/fisiologia , Proteínas/fisiologia , Transativadores/fisiologia , Sequência de Aminoácidos , Animais , Linhagem Celular , DNA Complementar , Proteínas de Ligação a DNA/antagonistas & inibidores , Humanos , Dados de Sequência Molecular , Fosforilação , Proteínas Inibidoras de STAT Ativados , Fator de Transcrição STAT1 , Homologia de Sequência de Aminoácidos , Transativadores/antagonistas & inibidores , Ativação Transcricional , Tirosina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA