Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biol Pharm Bull ; 46(11): 1498-1505, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37914352

RESUMO

Adiponectin is an abundant adipocytokine secreted by adipocytes. It exists in the plasma in its trimeric, hexameric, high-molecular-weight (HMW), and globular (a proteolytic product) isoforms. Adiponectin's anti-inflammatory effects on macrophages remain controversial. We have previously reported a simple and effective method for purifying native HMW adiponectin from human plasma. Here, we investigated whether native HMW adiponectin from human plasma has anti-inflammatory effects on macrophages. Pretreatment with human native HMW adiponectin inhibited lipopolysaccharide (LPS)-induced interleukin-1ß (IL-1ß) gene expression, but not tumor necrosis factor (TNF)-α expression. However, simultaneous treatment with HMW adiponectin and LPS did not inhibit IL-1ß expression. Further, HMW adiponectin pretreatment decreases glycogen synthase kinase-3ß (GSK-3ß) inactivation by abrogating LPS-induced Akt (Ser473) phosphorylation, which subsequently suppresses LPS-induced CCAAT/enhancer binding protein ß (C/EBPß) protein translation and nuclear translocation. However, HMW adiponectin pretreatment did not affect LPS-induced nuclear factor-kappaB (NF-κB) activation. These results suggest that HMW adiponectin mediates potent anti-inflammatory activities in macrophages by inhibiting its Akt-C/EBPß signaling pathway, thereby suppressing IL-1ß gene expression.


Assuntos
Adiponectina , Lipopolissacarídeos , Humanos , Adiponectina/genética , Adiponectina/metabolismo , Anti-Inflamatórios/farmacologia , Expressão Gênica , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
2.
J Immunol ; 210(12): 1867-1881, 2023 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-37186262

RESUMO

Recent studies have highlighted the pathogenic roles of IL-17-producing CD8+ T cells (T-cytotoxic 17 [Tc17]) in psoriasis. However, the underlying mechanisms of Tc17 induction remain unclear. In this study, we focused on the pathogenic subsets of Th17 and their mechanism of promotion of Tc17 responses. We determined that the pathogenic Th17-enriched fraction expressed melanoma cell adhesion molecule (MCAM) and CCR6, but not CD161, because this subset produced IL-17A abundantly and the presence of these cells in the peripheral blood of patients has been correlated with the severity of psoriasis. Intriguingly, the serial analysis of gene expression revealed that CCR6+MCAM+CD161-CD4+ T cells displayed the gene profile for adaptive immune responses, including CD83, which is an activator for CD8+ T cells. Coculture assay with or without intercellular contact between CD4+ and CD8+ T cells showed that CCR6+MCAM+CD161-CD4+ T cells induced the proliferation of CD8+ T cells in a CD83-dependent manner. However, the production of IL-17A by CD8+ T cells required exogenous IL-17A, suggesting that intercellular contact via CD83 and the production of IL-17A from activated CD4+ T cells elicit Tc17 responses. Intriguingly, the CD83 expression was enhanced in the presence of IL-15, and CD83+ cells stimulated with IL-1ß, IL-23, IL-15, and IL-15Rα did not express FOXP3. Furthermore, CCR6+MCAM+CD161-CD4+ T cells expressing CD83 were increased in the peripheral blood of patients, and the CD83+ Th17-type cells accumulated in the lesional skin of psoriasis. In conclusion, pathogenic MCAM+CD161- Th17 cells may be involved in the Tc17 responses via IL-17A and CD83 in psoriasis.

3.
FEBS Lett ; 585(19): 3058-64, 2011 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-21888904

RESUMO

Nitric oxide (NO) has been implicated in pancreatic ß-cell death in the development of diabetes. The mechanisms underlying NO-induced ß-cell death have not been clearly defined. Recently, receptor-interacting protein-1 (RIP1)-dependent necrosis, which is inhibited by necrostatin-1, an inhibitor of RIP1, has emerged as a form of regulated necrosis. Here, we show that NO donor-induced ß-cell death was inhibited by necrostatin-1. Unexpectedly, however, RIP1 knockdown neither inhibited cell death nor altered the protective effects of necrostatin-1 in NO donor-treated ß-cells. These results indicate that NO donor induces necrostatin-1-inhibitable necrotic ß-cell death independent of RIP1. Our findings raise the possibility that NO-mediated ß-cell necrosis may be a novel form of signal-regulated necrosis, which play a role in the progression of diabetes.


Assuntos
Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Imidazóis/metabolismo , Indóis/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/patologia , Doadores de Óxido Nítrico/farmacologia , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Linhagem Celular , Ciclofilina A/metabolismo , Técnicas de Silenciamento de Genes , Proteína HMGB1/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , S-Nitrosoglutationa/farmacologia , Transdução de Sinais/efeitos dos fármacos
4.
J Biol Chem ; 286(33): 29388-29396, 2011 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-21700708

RESUMO

Insulin receptor substrate-2 (IRS-2) plays a critical role in the survival and function of pancreatic ß-cells. Gene disruption of IRS-2 results in failure of the ß-cell compensatory mechanism and diabetes. Nonetheless, the regulation of IRS-2 protein expression in ß-cells remains largely unknown. Inducible nitric-oxide synthase (iNOS), a major mediator of inflammation, has been implicated in ß-cell damage in type 1 and type 2 diabetes. The effects of iNOS on IRS-2 expression have not yet been investigated in ß-cells. Here, we show that iNOS and NO donor decreased IRS-2 protein expression in INS-1/832 insulinoma cells and mouse islets, whereas IRS-2 mRNA levels were not altered. Interleukin-1ß (IL-1ß), alone or in combination with interferon-γ (IFN-γ), reduced IRS-2 protein expression in an iNOS-dependent manner without altering IRS-2 mRNA levels. Proteasome inhibitors, MG132 and lactacystin, blocked the NO donor-induced reduction in IRS-2 protein expression. Treatment with NO donor led to activation of glycogen synthase kinase-3ß (GSK-3ß) and c-Jun N-terminal kinase (JNK/SAPK) in ß-cells. Inhibition of GSK-3ß by pharmacological inhibitors or siRNA-mediated knockdown significantly prevented NO donor-induced reduction in IRS-2 expression in ß-cells. In contrast, a JNK inhibitor, SP600125, did not effectively block reduced IRS-2 expression in NO donor-treated ß-cells. These data indicate that iNOS-derived NO reduces IRS-2 expression by promoting protein degradation, at least in part, through a GSK-3ß-dependent mechanism. Our findings suggest that iNOS-mediated decreased IRS-2 expression may contribute to the progression and/or exacerbation of ß-cell failure in diabetes.


Assuntos
Regulação da Expressão Gênica/fisiologia , Quinase 3 da Glicogênio Sintase/metabolismo , Proteínas Substratos do Receptor de Insulina/biossíntese , Células Secretoras de Insulina/metabolismo , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase Tipo II/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Animais , Antracenos/farmacologia , Linhagem Celular Tumoral , Inibidores de Cisteína Proteinase/farmacologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Humanos , Proteínas Substratos do Receptor de Insulina/genética , Células Secretoras de Insulina/citologia , Interferon gama/genética , Interferon gama/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Leupeptinas/farmacologia , Camundongos , Óxido Nítrico Sintase Tipo II/genética , Complexo de Endopeptidases do Proteassoma/genética , Inibidores de Proteassoma , Ratos
5.
J Neurochem ; 106(6): 2375-84, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18643870

RESUMO

Math2 (NEX-1/NeuroD6) is a member of the basic helix-loop-helix transcription factor family and is involved in neuronal differentiation and maturation. In this study, we identified the genes targeted by Math2 using DNA microarrays and cultured rat cortical cells transfected with Math2. Of the genes regulated by Math2, we focused on plasticity-related gene 1 (Prg1). Prg1 expression induced by Math2 was confirmed in cultured rat cortical cells and PC12 cells analyzed by real-time quantitative PCR. In the promoter region of rat Prg1, we identified four E-boxes [designated -E1 to -E4 (CANNTG)] recognized by the basic helix-loop-helix transcription factor. Using chromatin immunoprecipitation assays, we found that Math2 directly bound to at least one of these E-boxes. The Prg1 reporter assay showed that -E1 was critical for the regulation of Math2-mediated Prg1 expression. Investigation of the functional roles of Math2 and Prg1 in PC12 cells revealed that 72 h after transfection with either Math2 or Prg1, neurite length and number were significantly induced. Co-transfection with Prg1-siRNA completely inhibited Math2-mediated morphological changes. Our results suggest that Math2 directly regulates Prg1 expression and that the Math2-Prg1 cascade plays an important role in neurite outgrowth in PC12 cells.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Encéfalo/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Neurônios/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Encéfalo/citologia , Encéfalo/embriologia , Proteínas de Ligação a Calmodulina , Diferenciação Celular/genética , Células Cultivadas , Elementos E-Box/genética , Sequências Hélice-Alça-Hélice/genética , Neuritos/metabolismo , Neuritos/ultraestrutura , Células PC12 , Monoéster Fosfórico Hidrolases/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica/genética , Ratos , Elementos Reguladores de Transcrição/genética , Fatores de Transcrição/genética
6.
Biochim Biophys Acta ; 1633(2): 96-105, 2003 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-12880869

RESUMO

Distinct functional coupling between cyclooxygenases (COXs) and specific terminal prostanoid synthases leads to phase-specific production of particular prostaglandins (PGs). In this study, we examined the coupling between COX isozymes and PGF synthase (PGFS). Co-transfection of COXs with PGFS-I belonging to the aldo-keto reductase family into HEK293 cells resulted in increased production of PGF(2alpha) only when a high concentration of exogenous arachidonic acid (AA) was supplied. However, this enzyme failed to produce PGF(2alpha) from endogenous AA, even though significant increase in PGF(2alpha) production occurred in cells transfected with COX-2 alone. This poor COX/PGFS-I coupling was likely to arise from their distinct subcellular localization. Measurement of PGF(2alpha)-synthetic enzyme activity in homogenates of several cells revealed another type of PGFS activity that was membrane-bound, glutathione (GSH)-activated, and stimulus-inducible. In vivo, membrane-bound PGFS activity was elevated in the lung of lipopolysaccharide-treated mice. Taken together, our results suggest the presence of a novel, membrane-associated form of PGFS that is stimulus-inducible and is likely to be preferentially coupled with COX-2.


Assuntos
Dinoprosta/biossíntese , Hidroxiprostaglandina Desidrogenases/química , Isoenzimas/química , Prostaglandina-Endoperóxido Sintases/química , Animais , Ácido Araquidônico/farmacologia , Linhagem Celular , Membrana Celular/enzimologia , Ciclo-Oxigenase 1 , Ciclo-Oxigenase 2 , Glutationa/farmacologia , Glutationa Transferase/análise , Humanos , Hidroxiprostaglandina Desidrogenases/biossíntese , Hidroxiprostaglandina Desidrogenases/metabolismo , Oxirredutases Intramoleculares/análise , Isoenzimas/genética , Lipopolissacarídeos/administração & dosagem , Pulmão/enzimologia , Macrófagos Peritoneais/enzimologia , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Prostaglandina-E Sintases , Prostaglandina-Endoperóxido Sintases/genética , Transfecção , Células Tumorais Cultivadas
7.
J Biol Chem ; 278(34): 32275-83, 2003 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-12799365

RESUMO

In this study we report the cloning and characterization of a novel human aminopeptidase, which we designate leukocyte-derived arginine aminopeptidase (L-RAP). The sequence encodes a 960-amino acid protein with significant homology to placental leucine aminopeptidase and adipocyte-derived leucine aminopeptidase. The predicted L-RAP contains the HEXXH(X)18E zinc-binding motif, which is characteristic of the M1 family of zinc metallopeptidases. Phylogenetic analysis indicates that L-RAP forms a distinct subfamily with placental leucine aminopeptidase and adipocyte-derived leucine aminopeptidase in the M1 family. Immunocytochemical analysis indicates that L-RAP is located in the lumenal side of the endoplasmic reticulum. Among various synthetic substrates tested, L-RAP revealed a preference for arginine, establishing that the enzyme is a novel arginine aminopeptidase with restricted substrate specificity. In addition to natural hormones such as angiotensin III and kallidin, L-RAP cleaved various N-terminal extended precursors to major histocompatibility complex class I-presented antigenic peptides. Like other proteins involved in antigen presentation, L-RAP is induced by interferon-gamma. These results indicate that L-RAP is a novel aminopeptidase that can trim the N-terminal extended precursors to antigenic peptides in the endoplasmic reticulum.


Assuntos
Aminopeptidases/metabolismo , Cistinil Aminopeptidase/metabolismo , Leucócitos/enzimologia , Sequência de Aminoácidos , Aminopeptidases/química , Aminopeptidases/genética , Sequência de Bases , Clonagem Molecular , Cistinil Aminopeptidase/química , Cistinil Aminopeptidase/genética , DNA Complementar , Dados de Sequência Molecular , Filogenia , Homologia de Sequência de Aminoácidos
8.
Prostaglandins Other Lipid Mediat ; 68-69: 383-99, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12432931

RESUMO

Prostaglandin E synthase (PGES), which converts cyclooxygenase (COX)-derived prostaglandin (PG)H2 to PGE2, occurs in multiple forms with distinct enzymatic properties, modes of expression, cellular and subcellular localizations and intracellular functions. Cytosolic PGES (cPGES) is a cytosolic protein that is constitutively expressed in a wide variety of cells and tissues and is associated with heat shock protein 90 (Hsp90). Membrane-associated PGES (mPGES), the expression of which is stimulus-inducible and is downregulated by anti-inflammatory glucocorticoids, is a perinuclear protein belonging to the microsomal glutathione S-transferase (GST) family. These two PGESs display distinct functional coupling with upstream COXs in cells; cPGES is predominantly coupled with the constitutive COX-1, whereas mPGES is preferentially linked with the inducible COX-2. Several cytosolic GSTs also have the capacity to convert PGH2 to PGE2 in vitro. Accumulating evidence has suggested that mPGES participates in various pathophysiological states in which COX-2 is involved, implying that mPGES represents a potential novel target for drug development.


Assuntos
Oxirredutases Intramoleculares/metabolismo , Isoenzimas/metabolismo , Doença de Alzheimer/metabolismo , Sequência de Aminoácidos , Animais , Ácido Araquidônico/metabolismo , Osso e Ossos/metabolismo , Ciclo-Oxigenase 2 , Febre/metabolismo , Glutationa Transferase/metabolismo , Humanos , Inflamação/metabolismo , Oxirredutases Intramoleculares/química , Oxirredutases Intramoleculares/genética , Isoenzimas/genética , Proteínas de Membrana , Dados de Sequência Molecular , Neoplasias/metabolismo , Dor/metabolismo , Prostaglandina-E Sintases , Prostaglandina-Endoperóxido Sintases/metabolismo , Reprodução/fisiologia , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA