Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 35(45): 5826-5838, 2016 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-27181202

RESUMO

Hyperphosphorylation of signal transducer and activator of transcription 3 (STAT3) has been found in various types of human cancers, including head and neck cancer (HNC). Although smoking is critical in the development and progression of HNC, how tobacco components activate STAT3 is unclear. We demonstrated that exposure of HNC cell lines to a tobacco extract induced a rapid Y705 phosphorylation of STAT3 and a rapid increase in the SUMO protease SENP3 that depended on a simultaneous increase in reactive oxygen species. We identified that SUMOylation at the lysine 451 site facilitated STAT3 binding to the phosphatase TC45 through an SUMO-interacting motif of TC45. SENP3 could thus enhance STAT3 phosphorylation by de-conjugating the SUMO2/3 modification of STAT3. Knocking-down of SENP3 greatly impaired basal and induced STAT3 phosphorylation by tobacco extract or interleukin 6. A correlation between SENP3 protein levels and STAT3 Y705 phosphorylation levels in human laryngeal carcinoma specimens was found, which was more significant in the specimens derived from the smoker patients and with poor clinicopathological parameters. Our data identified SUMOylation as a previously undescribed post-translational modification of STAT3 and SENP3 as a critical positive modulator of tobacco- or cytokine-induced STAT3 activation. These findings provide novel insights into the hyperphosphorylation of STAT3 in development of HNC.


Assuntos
Cisteína Endopeptidases/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Fator de Transcrição STAT3/metabolismo , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/etiologia , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Interleucina-6/farmacologia , Neoplasias Laríngeas/etiologia , Neoplasias Laríngeas/metabolismo , Neoplasias Laríngeas/patologia , Masculino , Estadiamento de Neoplasias , Nitrosaminas/farmacologia , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Fumar/efeitos adversos , Sumoilação , Transcrição Gênica , Ubiquitinas/metabolismo
2.
Clin Pharmacol Ther ; 95(1): 45-52, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24091715

RESUMO

Anthracyclines are powerful chemotherapy agents that are still widely used today. However, their clinical use is limited by the development of dose-dependent cardiotoxicity. Recently, we showed that topoisomerase 2ß (Top2ß) is required for anthracycline to induce DNA double-strand breaks and changes in the transcriptome, leading to mitochondrial dysfunction and generation of reactive oxygen species. Furthermore, deleting Top2ß from cardiomyocytes prevented the development of anthracycline-induced cardiotoxicity in mice. On the basis of this molecular insight, new strategies should be developed to prevent anthracycline-induced cardiotoxicity. First, Top2α-specific anthracyclines should be tested to determine whether they will spare the heart. Second, Top2ß should be studied as a potential biomarker to predict risk of developing cardiotoxicity before anthracycline treatment. Third, inhibiting and deleting Top2ß in the heart should also be tested as primary prevention strategies. We propose that Top2ß is a promising molecular target that can be used to design interventions to prevent anthracycline-induced cardiotoxicity.


Assuntos
Antraciclinas/toxicidade , Cardiotoxinas/toxicidade , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Marcação de Genes/métodos , Prevenção Primária/métodos , Animais , Antraciclinas/antagonistas & inibidores , Cardiotoxinas/antagonistas & inibidores , DNA Topoisomerases Tipo II/genética , Proteínas de Ligação a DNA/genética , Humanos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/enzimologia
3.
Oncogene ; 32(19): 2493-8, 2013 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-22733136

RESUMO

SUMO-specific protease 1 (SENP1) is a member of de-SUMOylation protease family and has an important role in the regulation of androgen receptor-dependent transcription and hypoxia signaling. This activity profile of SENP1 prompted us to investigate whether SENP1 is involved in the pathogenesis of prostate cancer. In previous studies, we have detected the overexpression of SENP1 in both precancerous prostate intraepithelial neoplasia (PIN) lesions and prostate cancer tissue samples from patients. Whereas our whole-animal model has demonstrated that SENP1 induction is critical for prostate cell transformation, the role of SENP1 in prostate cancer progression is still unknown. In this study, we show that SENP1 expression directly correlates with prostate cancer aggressiveness and reccurrence, by analyzing more than 150 prostate cancer specimens. Modulating SENP1 level dictates colony formation of prostate cancer cell lines, tumor growth in nude mice and also prostate cancer cell migration and invasion. Silencing SENP1 level in highly metastatic prostate cancer cells perturbs their ability to metastasize to the bone and initiates secondary tumors. Mechanistically, the expression of two critical bone remodeling proteins, matrix metalloproteinase 2 (MMP2) and MMP9, is regulated by SENP1 through the HIF1α signaling pathway. All these results show the contribution of SENP1 to the progression of prostate cancer, and suggest that SENP1 may be a prognostic marker and a therapeutic target for metastasis in prostate cancer patients.


Assuntos
Endopeptidases/biossíntese , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Animais , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Cisteína Endopeptidases , Progressão da Doença , Endopeptidases/genética , Endopeptidases/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Nus , Metástase Neoplásica , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Transfecção
4.
Oncogene ; 29(41): 5568-78, 2010 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-20661221

RESUMO

Hypoxia-inducible factor-1α (HIF1α) is a crucial regulator of the cellular response to hypoxia through its regulation of genes that control erythropoiesis, angiogenesis and anaerobic metabolism. We have previously shown that HIF1α stability is regulated by SUMOylation under the hypoxic condition. However, how HIF1α became SUMOylated during hypoxia is still unknown. In this study we identify PIASy as a specific E3 ligase for hypoxia-induced HIF1α SUMOylation. Hypoxia promotes translocation of HIF1α to the nucleus to facilitate its binding to PIASy, enabling the conjugation of HIF1α by SUMO1. We further show that PIASy negatively regulates hypoxia-induced HIF1α stability and transactivation. Knocking down PIASy increases the angiogenic activity of endothelial cells. Moreover, we show an inverse relationship between expression of PIASy and tumor angiogenesis in colon cancer. Thus, we define an important role of PIASy in hypoxia signaling through promoting HIF1α SUMOylation.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteínas Inibidoras de STAT Ativados/metabolismo , Proteína SUMO-1/metabolismo , Animais , Hipóxia Celular , Linhagem Celular , Células Cultivadas , Neoplasias do Colo/irrigação sanguínea , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Cisteína Endopeptidases , Endopeptidases/genética , Endopeptidases/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Immunoblotting , Imuno-Histoquímica , Imunoprecipitação , Camundongos , Camundongos Knockout , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Ligação Proteica , Proteínas Inibidoras de STAT Ativados/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína SUMO-1/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
5.
J Thromb Haemost ; 4(11): 2308-16, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16961584

RESUMO

Atherothrombosis is the process that links atherosclerotic lesion development with unpredictable and life-threatening ischemic vascular events such as angina, myocardial infarction, transient ischemic attack, and stroke. Atherothrombosis is triggered when an unstable atherosclerotic lesion is ruptured, leading to platelet activation and thrombus formation. Inflammatory mediators are responsible for lesion instability leading to rupture, and in recent years atherothrombosis and its underlying condition of atherosclerosis have come to be recognized as manifestations of inflammatory disease. Inflammatory mediators may therefore serve as early markers of atherothrombosis. Measurement of early markers may be used to predict future ischemic events and improve risk stratification in patients following diagnosis of atherothrombotic disease. In addition, detection of such markers may help to optimize the use of current therapies to manage atherothrombosis. Molecules that may serve as early markers of atherothrombotic disease include C-reactive protein, CD40 ligand, myeloperoxidase, pregnancy-associated plasma protein and plasminogen activator inhibitor-1. Early indications are that levels of these markers are influenced by therapies currently in use in the treatment of atherothrombotic conditions, including antiplatelet agents. Ongoing studies will provide further insight into routine assessment of inflammatory markers as a guide to the management of patients with atherothrombosis.


Assuntos
Aterosclerose/sangue , Mediadores da Inflamação/sangue , Inibidores da Agregação Plaquetária/uso terapêutico , Trombose/sangue , Aterosclerose/complicações , Aterosclerose/tratamento farmacológico , Aterosclerose/fisiopatologia , Biomarcadores/sangue , Feminino , Humanos , Inflamação/sangue , Inflamação/tratamento farmacológico , Masculino , Gravidez , Prognóstico , Trombose/tratamento farmacológico , Trombose/etiologia , Trombose/fisiopatologia
6.
J Clin Invest ; 108(10): 1513-22, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11714743

RESUMO

NADPH oxidase is upregulated in smooth muscle cells (SMCs) in response to growth factor stimulation, concomitant with increased reactive oxygen species (ROS) production. We investigated the role of ROS production by NADPH oxidase in SMC responses to growth factors and in atherosclerotic lesion formation in ApoE(-/-) mice. SMCs from wild-type, p47phox(-/-), and gp91phox(-/-) mice differed markedly with respect to growth factor responsiveness and ROS generation. p47phox(-/-) SMCs had diminished superoxide production and a decreased proliferative response to growth factors compared with wild-type cells, whereas the response of gp91phox(-/-) SMCs was indistinguishable from that of wild-type SMCs. The relevance of these in vitro observations was tested by measuring atherosclerotic lesion formation in genetically modified (wild-type, p47phox(-/-), ApoE(-/-), and ApoE(-/-)/p47phox(-/-)) mice. ApoE(-/-)/p47phox(-/-) mice had less total lesion area than ApoE(-/-) mice, regardless of whether mice were fed standard chow or a high-fat diet. Together, these studies provide convincing support for the hypothesis that superoxide generation in general, and NADPH oxidase in particular, have a requisite role in atherosclerotic lesion formation, and they provide a rationale for further studies to dissect the contributions of ROS to vascular lesion formation.


Assuntos
Apolipoproteínas E/fisiologia , Arteriosclerose/fisiopatologia , Fosfoproteínas/fisiologia , Animais , Apolipoproteínas E/genética , Arteriosclerose/genética , Arteriosclerose/metabolismo , Sequência de Bases , Divisão Celular/fisiologia , Primers do DNA , Progressão da Doença , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/patologia , NADPH Oxidases , Espécies Reativas de Oxigênio
7.
J Biol Chem ; 276(49): 46655-60, 2001 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-11585840

RESUMO

NEDD8 is a ubiquitin-like protein that controls vital biological events through its conjugation to cullin family members. Recently, we identified a negative regulator of the NEDD8 conjugation system, NUB1, which interacts with NEDD8 and down-regulates NEDD8 expression post-transcriptionally (Kito, K., Yeh, E. T. H., and Kamitani, T. (2001) J. Biol. Chem. 276, 20603-20609). Here, we show that NUB1 possesses a ubiquitin-like domain at the N-terminal region and binds to S5a of the 19 S proteasome activator (PA700). A GST pull-down assay revealed that the overexpression of NUB1 leads to a greater precipitation of NEDD8 conjugates with GST-S5a, suggesting that NUB1 might have an adaptor function between S5a and NEDD8. Furthermore, proteasome inhibitors completely block NUB1-mediated down-regulation of NEDD8 expression. These results suggest that NUB1 recruits NEDD8 and its conjugates to the proteasome for degradation, providing a direct functional link between the NEDD8 conjugation system and the proteasomal degradation pathway.


Assuntos
Cisteína Endopeptidases/metabolismo , Complexos Multienzimáticos/metabolismo , Fatores de Transcrição/metabolismo , Ubiquitinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Western Blotting , Células COS , Glutationa Transferase/metabolismo , Humanos , Hidrólise , Proteína NEDD8 , Complexo de Endopeptidases do Proteassoma , Ligação Proteica , Fatores de Transcrição/isolamento & purificação , Células Tumorais Cultivadas , Ubiquitinas/isolamento & purificação
9.
Circulation ; 103(21): 2531-4, 2001 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-11382718

RESUMO

BACKGROUND: C-reactive protein (CRP) induces adhesion molecule expression by endothelial cells. However, the effects of CRP on chemokine expression by endothelial cells are not known. METHODS AND RESULTS: We tested the effects of CRP on the production of the chemokines monocyte chemoattractant protein-1 (MCP-1) and RANTES in cultured human umbilical vein endothelial cells. The secretion of chemokines was assessed by ELISA. Incubation with 100 microgram/mL recombinant human CRP induced a 7-fold increase in MCP-1 but no change in RANTES secretion. We showed that the effect of CRP on MCP-1 was present even at 5 microgram/mL CRP, with stepwise increases as the CRP concentration was increased to 10, 50, and 100 microgram/mL. The effect of CRP on MCP-1 induction was not influenced by aspirin (at concentrations up to 1 mmol/L), but it was significantly inhibited by 5 micromol/L simvastatin. The peroxisome proliferator-activated receptor-alpha activators fenofibrate (100 micromol/L) and Wy-14649 (100 micromol/L) almost completely abolished the induction of MCP-1, but the peroxisome proliferator-activated receptor-gamma activator ciglitazone had only a moderate effect. CONCLUSIONS: These results further strengthen the role of CRP in the pathogenesis of vascular inflammation and, likely, atherosclerosis and provide a crucial insight into a novel mechanism of action of anti-atherosclerosis drugs such as simvastatin and fenofibrate.


Assuntos
Proteína C-Reativa/farmacologia , Quimiocina CCL2/metabolismo , Endotélio Vascular/efeitos dos fármacos , Hipolipemiantes/farmacologia , Células Cultivadas , Quimiocina CCL5/metabolismo , Relação Dose-Resposta a Droga , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Fenofibrato/farmacologia , Humanos , Receptores Citoplasmáticos e Nucleares/metabolismo , Sinvastatina/farmacologia , Fatores de Tempo , Fatores de Transcrição/metabolismo
10.
J Biol Chem ; 276(23): 20603-9, 2001 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-11259415

RESUMO

NEDD8, a ubiquitin-like protein, covalently conjugates to cullin family members. It appears to control vital biological events through its conjugation to cullins. To study how this conjugation pathway is regulated, we performed yeast two-hybrid screening by using NEDD8 as a bait and isolated a cDNA fragment encoding a potent down-regulator of the NEDD8 expression. Here, we report this novel regulator, NUB1 (NEDD8 Ultimate Buster-1). NUB1 is composed of 601 residues with a calculated 69.1-kDa molecular mass. It is an interferon-inducible protein and predominantly localized in the nucleus. The NUB1 message is specifically expressed in adult human testis, ovary, heart, and skeletal muscle tissues and is developmentally down-regulated in mouse embryos. In biochemical analysis, we found that NUB1 overexpression leads to severe reduction of NEDD8 monomer and NEDD8 conjugates in cells. This reduction is not due to down-regulation of NEDD8 transcription, but due to post-transcriptional mechanism. As expected from this activity, overexpression of NUB1 had a profound growth-inhibitory effect on U2OS cells. Thus, NUB1 is a strong down-regulator of the NEDD8 expression and appears to play critical roles in regulating biological events, including cell growth.


Assuntos
Regulação para Baixo , Interferons/fisiologia , Fatores de Transcrição/biossíntese , Ubiquitinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting , Células COS , Divisão Celular , DNA Complementar , Humanos , Dados de Sequência Molecular , Proteína NEDD8 , Ligação Proteica , Frações Subcelulares/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Células Tumorais Cultivadas , Técnicas do Sistema de Duplo-Híbrido
11.
Circulation ; 102(18): 2165-8, 2000 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-11056086

RESUMO

BACKGROUND: The acute-phase reactant C-reactive protein (CRP) is an important risk factor for coronary heart disease. However, the possible effects of CRP on vascular cells are not known. METHODS AND RESULTS: We tested the effects of CRP on expression of adhesion molecules in both human umbilical vein and coronary artery endothelial cells. Expression of vascular cell adhesion molecule (VCAM-1), intercellular adhesion molecule (ICAM-1), and E-selectin was assessed by flow cytometry. Incubation with recombinant human CRP (10 microg/mL) for 24 hours induced an approximately 10-fold increase in expression of ICAM-1 and a significant expression of VCAM-1, whereas a 6-hour incubation induced significant E-selectin expression. Adhesion molecule induction was similar to that observed in endothelial cells activated with interleukin-1beta. In coronary artery endothelial cells, induction of ICAM-1 and VCAM-1 was already present at 5 microg/mL and reached a maximum at 50 microg/mL, at which point a substantial increase in expression of E-selectin was also evident. The CRP effect was dependent on presence of human serum in the culture medium, because no effect was seen in cells cultured with serum-free medium. In contrast, interleukin-1beta was able to induce adhesion molecule expression in the absence of human serum. CONCLUSIONS: CRP induces adhesion molecule expression in human endothelial cells in the presence of serum. These findings support the hypothesis that CRP may play a direct role in promoting the inflammatory component of atherosclerosis and present a potential target for the treatment of atherosclerosis.


Assuntos
Proteína C-Reativa/metabolismo , Endotélio Vascular/metabolismo , Mediadores da Inflamação/metabolismo , Vasculite/metabolismo , Arteriosclerose/etiologia , Arteriosclerose/imunologia , Proteína C-Reativa/imunologia , Proteína C-Reativa/farmacologia , Moléculas de Adesão Celular/biossíntese , Células Cultivadas , Vasos Coronários , Meios de Cultura Livres de Soro/farmacologia , Selectina E/biossíntese , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Citometria de Fluxo , Humanos , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/farmacologia , Molécula 1 de Adesão Intercelular/biossíntese , Interleucina-1/metabolismo , Interleucina-1/farmacologia , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Veias Umbilicais , Molécula 1 de Adesão de Célula Vascular/biossíntese , Vasculite/etiologia
12.
Am J Physiol Heart Circ Physiol ; 279(6): H3065-75, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11087265

RESUMO

We tested the hypothesis that selectin inhibition with blocking antibodies or a small-molecular-weight inhibitor of L-, P-, and E-selectin, methoxybenzoylpropionic acid (MBPA), prevents thrombus formation in a canine coronary Folts' model. Cyclic flow variations (CFVs) were induced by crush injury and constriction of the left anterior descending coronary artery in dogs. Systemic infusion of antibodies to P- and L-selectin abolished CFVs, respectively, in 50% and 17% of treated dogs [P = not significant (NS)]. The combination of P- and L-selectin antibodies suppressed CFVs in 60% of treated dogs (P = NS). In contrast, systemic selectin blockade by intravenous infusion or local adventitial application of MBPA markedly reduced CFVs and, in addition, reduced myocardial myeloperoxidase (MPO) activity. We conclude that inhibition of L-, P-, and E-selectin binding by a small-molecular-weight, noncarbohydrate compound markedly reduces arterial thrombosis, whereas systemic administration of antibodies to L- and P-selectin fail to reproduce this antithrombotic effect. These results underscore the role of selectins in the pathogenesis of arterial thrombosis under high shear stress and suggest that inhibition of P- and L- selectin may not suffice to prevent thrombus formation in this model. The role of E-selectin in thrombus formation in this model awaits further testing.


Assuntos
Trombose Coronária/tratamento farmacológico , Trombose Coronária/imunologia , Propionatos/farmacologia , Selectinas/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Contagem de Células Sanguíneas , Trombose Coronária/prevenção & controle , Vasos Coronários/lesões , Vasos Coronários/fisiopatologia , Modelos Animais de Doenças , Cães , Selectina E/imunologia , Feminino , Injeções Intravenosas , Selectina L/imunologia , Leucócitos/imunologia , Masculino , Testes de Neutralização , Neutrófilos/imunologia , Selectina-P/imunologia , Éteres Fenílicos , Agregação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/fisiologia , Vasculite/tratamento farmacológico , Vasculite/imunologia , Vasculite/prevenção & controle
13.
Biochim Biophys Acta ; 1494(1-2): 185-8, 2000 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11072084

RESUMO

CATCH 22 syndromes, which include DiGeorge syndrome and Velocardiofacial syndrome, are the most common cause of congenital heart disease which involve microdeletion of 22q11. Using a strategy including EST searching, PCR amplification and 5'-RACE, we have cloned a 1487 bp cDNA fragment from human heart cDNA library. The cloned GNB1L cDNA encodes a G-protein beta-subunit-like polypeptide, and the GNB1L gene is located in the critical region for DiGeorge syndrome. A comparison of GNB1L cDNA sequence with corresponding genomic DNA sequence revealed that this gene consists of seven exons and spans an approximately 60 kb genomic region. Northern blot analysis revealed GNB1L is highly expressed in the heart.


Assuntos
Deleção Cromossômica , Cromossomos Humanos Par 22/genética , Síndrome de DiGeorge/genética , Proteínas Heterotriméricas de Ligação ao GTP/genética , Sequência de Aminoácidos , Sequência de Bases , Clonagem Molecular , Éxons/genética , Etiquetas de Sequências Expressas , Perfilação da Expressão Gênica , Proteínas Heterotriméricas de Ligação ao GTP/química , Humanos , Dados de Sequência Molecular , Miocárdio/metabolismo , Mapeamento Físico do Cromossomo , Subunidades Proteicas , RNA Mensageiro/análise , RNA Mensageiro/genética , Alinhamento de Sequência
14.
J Biol Chem ; 275(50): 39458-65, 2000 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-10978339

RESUMO

MCL1 (ML1 myeloid cell leukemia 1), a Bcl-2 (B- cell lymphoma-leukemia 2) homologue, is known to function as an anti-apoptotic protein. Here we show in vitro and in vivo that MCL1 interacts with the cell cycle regulator, proliferating cell nuclear antigen (PCNA). This finding prompted us to investigate whether MCL1, in addition to its anti-apoptotic function, has an effect on cell cycle progression. A bromodeoxyuridine uptake assay showed that the overexpression of MCL1 significantly inhibited the cell cycle progression through the S-phase. The S-phase of the cell cycle is also known to be regulated by PCNA. A mutant of MCL1 that lacks PCNA binding (MCL1(Delta)(4A)) could not inhibit cell cycle progression as effectively as wild type MCL1. In contrast, MCL1(Delta)(4A) retained its anti-apoptotic function in HeLa cells when challenged by Etoposide. In addition, the intracellular localization of MCL1(Delta)(4A) was identical to that of wild type MCL1. An in vitro pull-down assay suggested that MCL1 is the only Bcl-2 family protein to interact with PCNA. In fact, MCL1, not other Bcl-2 family proteins, contained the PCNA-binding motif described previously. Taken together, MCL1 is a regulator of both apoptosis and cell cycle progression, and the cell cycle regulatory function of MCL1 is mediated through its interaction with PCNA.


Assuntos
Apoptose , Ciclo Celular/fisiologia , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/fisiologia , Sequência de Aminoácidos , Animais , Bromodesoxiuridina/farmacocinética , Linhagem Celular , Clonagem Molecular , Etoposídeo/farmacologia , Técnica Indireta de Fluorescência para Anticorpo , Biblioteca Gênica , Células HeLa , Humanos , Camundongos , Microscopia Confocal , Dados de Sequência Molecular , Mutação , Proteína de Sequência 1 de Leucemia de Células Mieloides , Testes de Precipitina , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ligação Proteica , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Homologia de Sequência de Aminoácidos , Células Tumorais Cultivadas , Técnicas do Sistema de Duplo-Híbrido
15.
J Biol Chem ; 275(19): 14212-6, 2000 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-10799498

RESUMO

Covalent conjugation of proteins by ubiquitin or ubiquitin-like molecules is an important form of post-translational modification and plays a critical role in many cellular processes. Similar to the concept of phosphorylation and dephosphorylation, these conjugates are regulated by a large number of deconjugating enzymes. Here, we report the cloning of a 2,141-base pair DNA fragment from human placenta cDNA library by a strategy that involves expressed sequence tag data base searching, polymerase chain reaction, and rapid amplification of cDNA ends. Nucleotide sequence analysis revealed that the cloned cDNA contains an open reading frame of 1,143 base pairs encoding a novel protease, USP21, which is composed of 381 residues with a calculated molecular mass of 43 kDa. The human USP21 gene is located on chromosome 1q21 and encodes a member of the ubiquitin-specific protease family with highly conserved Cys and His domains. The activity and specificity of USP21 were determined by using a COS cell expression system in vivo. We showed that USP21 is capable of removing ubiquitin from ubiquitinated proteins as expected. Furthermore, USP21 is capable of removing NEDD8 from NEDD8 conjugates but has no effect on Sentrin-1 conjugates. As expected from its biochemical activity, overexpression of USP21 has a profound growth inhibitory effect on U2OS cells. Thus, USP21 is the first ubiquitin-specific protease shown to have dual specificity for both ubiquitin and NEDD8 and may play an important role in the regulation of cell growth.


Assuntos
Carbono-Nitrogênio Liases/metabolismo , Ubiquitinas/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Células COS , Carbono-Nitrogênio Liases/química , Carbono-Nitrogênio Liases/genética , Mapeamento Cromossômico , Clonagem Molecular , Etiquetas de Sequências Expressas , Humanos , Dados de Sequência Molecular , Proteína NEDD8 , Homologia de Sequência de Aminoácidos , Especificidade por Substrato , Ubiquitina Tiolesterase
16.
J Biol Chem ; 275(22): 17008-15, 2000 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-10828074

RESUMO

NEDD8, a novel ubiquitin-like protein, has been shown to conjugate to proteins in a manner analogous to ubiquitination and sentrinization. Recently, human UBC12 was identified as a putative NEDD8 conjugation enzyme (E2). While investigating the in vivo function of UBC12, we found that the point mutant, UBC12(C111S), showed a dominant-negative effect on NEDD8 conjugation. This mutant, with a single Cys-to-Ser substitution at the conserved Cys residue in the E2 family, could specifically inhibit NEDD8 conjugation. We observed the dominant-negative effect on NEDD8 conjugation to substrates, including the C-terminal fragment of cullin-2 (Cul-2-DeltaN), full-length cullin-1, and also other uncharacterized target proteins. Interestingly, UBC12(C111S) formed a heterodimeric conjugate with NEDD8. This conjugate was stable under stringent conditions, including 6 m guanidine HCl, 8 m urea, 2% SDS, or 5% beta-mercaptoethanol. Our results are consistent with the hypothesis that UBC12(C111S) sequesters the NEDD8 monomer by forming a UBC12(C111S)-NEDD8 conjugate and, in turn, inhibits the subsequent transfer of NEDD8 to its targets. To examine the biological role of NEDD8 conjugation, this dominant-negative form of UBC12 was applied to a cell growth assay. Overexpression of UBC12(C111S) led to inhibition of growth in U2OS and HEK293 cells. Thus, this dominant-negative form of UBC12 could be useful in defining the role of NEDD8 modification in other biological systems.


Assuntos
Proteínas Culina , Genes Dominantes , Mutação Puntual , Proteínas de Saccharomyces cerevisiae , Ubiquitinas/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Humanos , Proteína NEDD8 , Ubiquitinas/genética
17.
Gene ; 248(1-2): 1-14, 2000 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-10806345

RESUMO

Ubiquitin is a small polypeptide that covalently modifies other cellular proteins and targets them to the proteasome for degradation. In recent years, ubiquitin-dependent proteolysis has been demonstrated to play a critical role in the regulation of many cellular processes, such as cell cycle progression, cell signaling, and immune recognition. The recent discovery of three new ubiquitin-like proteins, NEDD8, Sentrin/SUMO, and Apg12, has further broadened the horizon of this type of post-translational protein modification. This review will focus on the biology and biochemistry of the Sentrin/SUMO and NEDD8 modification pathways, which are clearly distinct from the ubiquitination pathway and have unique biological functions.


Assuntos
Peptídeo Hidrolases , Ubiquitinas/metabolismo , Sequência de Aminoácidos , Cisteína Endopeptidases , Endopeptidases/metabolismo , Humanos , Dados de Sequência Molecular , Proteína NEDD8 , Processamento de Proteína Pós-Traducional , Proteína SUMO-1 , Homologia de Sequência de Aminoácidos , Ubiquitinas/genética
18.
Exp Cell Res ; 255(2): 135-43, 2000 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-10694430

RESUMO

The basic helix-loop-helix/leucine zipper (bHLH/ZIP) microphthalmia-associated transcription factor (MITF) regulates transcription of genes encoding enzymes essential for melanin biosynthesis in melanocytes and retinal pigmented epithelial cells. To determine how MITF activity is regulated, we used the yeast two-hybrid system to identify proteins expressed by human melanoma cells that interact with MITF. The majority of clones that showed positive interaction with a 158-amino-acid region of MITF containing the bHLH/ZIP domain (aa 168-325) encoded the ubiquitin conjugating enzyme hUBC9. The association of MITF with hUBC9 was further confirmed by an in vitro GST pull-down assay. Although hUBC9 is known to interact preferentially with SENTRIN/SUMO1, in vitro transcription/translation analysis demonstrated greater association of MITF with ubiquitin than with SENTRIN. Importantly, cotransfection of MITF and hUBC9 expression vectors resulted in MITF protein degradation. MITF protein was stabilized by the proteasome inhibitor MG132, indicating the role of the ubiquitin-proteasome system in MITF degradation. Serine 73, which is located in a region rich in proline, glutamic acid, serine, and threonine (PEST), regulates MITF protein stability, since a serine to alanine mutation prevented hUBC9-mediated MITF (S73A) degradation. Furthermore, we identified lysine 201 as a potential ubiquitination site. A lysine to arginine mutation abolished MITF (K201R) degradation by hUBC9 in vivo. Our experiments indicate that by targeting MITF for proteasome degradation, hUBC9 is a critical regulator of melanocyte differentiation.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica , Ligases/fisiologia , Melanócitos/fisiologia , Epitélio Pigmentado Ocular/fisiologia , Enzimas de Conjugação de Ubiquitina , Sequências Hélice-Alça-Hélice/fisiologia , Humanos , Fator de Transcrição Associado à Microftalmia , Fatores de Transcrição/fisiologia , Células Tumorais Cultivadas , Ubiquitinas/fisiologia
19.
J Biol Chem ; 275(5): 3355-9, 2000 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-10652325

RESUMO

Sentrin-1, also called SUMO-1, is a protein of 101 residues that is distantly related to ubiquitin and another ubiquitin-like protein, NEDD8. Here we report the cloning of a novel sentrin-specific protease, SENP1, which has no homology to the known de-ubiquitinating enzymes or ubiquitin C-terminal hydrolases. However, SENP1 is distantly related to the yeast Smt3-specific protease, Ulp1. A COS cell expression system was used to demonstrate the activity of SENP1 in vivo. When HA-tagged sentrin-1 was co-expressed with SENP1, the higher molecular weight sentrin-1 conjugates were completely removed. Surprisingly, the major sentrinized band at 90 kDa remained intact. The disappearance of the high molecular weight sentrin-1 conjugates also coincided with an increase in free sentrin-1 monomers. SENP1 is also active against proteins modified by sentrin-2, but not those modified by ubiquitin or NEDD8. In addition, sentrinized PML, a tumor suppressor protein that resides in the nucleus, was selectively affected by SENP1, whereas sentrinized RanGAP1, which is associated with the cytoplasmic fibrils of the nuclear pore complex, remained intact. The inability of SENP1 to process sentrinized RanGAP1 in vivo is most likely due to its nuclear localization because SENP1 is active against sentrinized RanGAP1 in vitro. The identification of a nuclear-localized, sentrin-specific protease will provide a unique tool to study the role of sentrinization in the biological function of PML and in the pathogenesis of acute promyelocytic leukemia.


Assuntos
Endopeptidases/genética , Endopeptidases/metabolismo , Ubiquitinas/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Clonagem Molecular , Cisteína Endopeptidases , DNA Complementar/genética , DNA Complementar/isolamento & purificação , Dados de Sequência Molecular , Proteínas Repressoras , Proteína SUMO-1 , Alinhamento de Sequência , Especificidade por Substrato
20.
Circulation ; 101(3): 235-8, 2000 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-10645917

RESUMO

BACKGROUND: Peroxisome proliferator-activated receptor-gamma (PPARgamma) is expressed in atherosclerotic plaques and in endothelial cells. The possible effects of PPARgamma activators on endothelial activation and inflammatory response within the plaque are currently unknown. METHODS AND RESULTS: We tested the hypothesis that PPARgamma activators inhibit vascular cell adhesion molecule (VCAM-1) and intercellular adhesion molecule (ICAM-1) expression in cultured endothelial cells (evaluated by flow cytometry) and homing of monocyte/macrophages to atherosclerotic plaques in vivo. In endothelial cells, the PPARgamma agonists troglitazone at 100 micromol/L and 15-deoxy-(Delta12,14)-prostaglandin J(2) (15d-PGJ2) at 20 micromol/L markedly attenuated the tumor necrosis factor-induced expression of VCAM-1 and ICAM-1. A significant inhibition of VCAM-1 expression was also evident at 5 and 10 micromol/L 15d-PGJ2 and 20 micromol/L troglitazone. Expression of E-selectin and PECAM-1 was not altered. To confirm the biological relevance of these results, we assessed the effects of troglitazone on monocyte/macrophage homing to atherosclerotic plaques in apoE-deficient mice. A 7-day treatment with troglitazone (400 mg/kg) significantly reduced monocyte/macrophage homing to atherosclerotic plaques (236+/-77 versus 177+/-43 macrophages, P=0.03); an even more striking inhibition was found at 3200 mg/kg troglitazone (344+/-76 versus 172+/-83 macrophages, P=0.005). CONCLUSIONS: PPARgamma activators inhibit expression of VCAM-1 and ICAM-1 in activated endothelial cells and significantly reduce monocyte/macrophage homing to atherosclerotic plaques. These findings suggest that PPARgamma activators, currently used in treatment of type II diabetes, may have beneficial effects in modulating inflammatory response in atherosclerosis.


Assuntos
Arteriosclerose/tratamento farmacológico , Molécula 1 de Adesão Intercelular/fisiologia , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Tiazolidinedionas , Fatores de Transcrição/efeitos dos fármacos , Molécula 1 de Adesão de Célula Vascular/fisiologia , Vasculite/tratamento farmacológico , Animais , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Cromanos/uso terapêutico , Endotélio Vascular/efeitos dos fármacos , Humanos , Molécula 1 de Adesão Intercelular/análise , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Camundongos , Monócitos/efeitos dos fármacos , Monócitos/fisiologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Tiazóis/uso terapêutico , Troglitazona , Molécula 1 de Adesão de Célula Vascular/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...