Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 131
Filtrar
1.
J Immunol ; 180(3): 1729-36, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18209070

RESUMO

We evaluated the capacity of NK cells to influence expansion of CD4(+)CD25(+)FoxP3(+) regulatory T cells (Tregs) in response to microbial Ags, using Mycobacterium tuberculosis as a model. We previously found that Tregs expand when CD4(+) cells and monocytes are exposed to M. tuberculosis. Addition of NK cells that were activated by monokines (IL-12, IL-15, and IL-18) or by exposure to M. tuberculosis-stimulated monocytes reduced Treg expansion in response to M. tuberculosis. NK cell inhibition of Treg expansion was not mediated through IFN-gamma. Activated NK cells lysed expanded, but not freshly isolated Tregs. Although monokines increased NK cell expression of the activating receptors NKp46, NKG2D, 2B4, CD16, and DNAM-1, only anti-NKG2D and anti-NKp46 inhibited NK cell lysis of expanded Tregs. Of five NKG2D ligands, only UL16-binding protein 1 (ULBP1) was up-regulated on M. tuberculosis-expanded Tregs, and anti-ULBP1 inhibited NK cell lysis of expanded Tregs. M. tuberculosis-stimulated monocytes activated NK cells to lyse expanded Tregs, and this was also inhibited by anti-NKG2D and anti-ULBP1, confirming the physiological relevance of this effect. Our study identifies a potential new role for NK cells in maintaining the delicate balance between the regulatory and effector arms of the immune response.


Assuntos
Citotoxicidade Imunológica , Células Matadoras Naturais/imunologia , Mycobacterium tuberculosis/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/microbiologia , Antígenos CD4/análise , Células Cultivadas , Fatores de Transcrição Forkhead/análise , Proteínas Ligadas por GPI , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Interferon gama/metabolismo , Subunidade alfa de Receptor de Interleucina-2/análise , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células Matadoras Naturais/efeitos dos fármacos , Ativação Linfocitária , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Monocinas/farmacologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Receptores Imunológicos/agonistas , Receptores de Células Matadoras Naturais
2.
J Exp Med ; 204(10): 2397-405, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17875674

RESUMO

Monokines (i.e., interleukin [IL]-12, -18, and -15) induce natural killer (NK) cells to produce interferon-gamma (IFN-gamma), which is a critical factor for immune surveillance of cancer and monocyte clearance of infection. We show that SET, which is a potent inhibitor of protein phosphatase type 2A (PP2A) activity, is highly expressed in human CD56bright NK cells, which produce more IFN-gamma than CD56dim NK cells. SET was up-regulated upon monokine stimulation of primary human NK cells. Furthermore, ectopic overexpression of SET significantly enhanced IFN-gamma gene expression in monokine-stimulated NK cells. In contrast, RNAi-mediated suppression of SET expression renders NK cells inefficient in producing high levels of IFN-gamma in response to monokine costimulation. Mechanistically, suppression of PP2A activity by SET is important for IFN-gamma gene expression in NK cells. In fact, treatment of primary human NK cells with the PP2A activator 1,9-dideoxy-forskolin, as well as administration of the drug to C57BL/6 mice, significantly reduced NK-dependent IFN-gamma production in response to monokine treatment. Further, SET knockdown or pharmacologic activation of PP2A diminished extracellular signal-regulated kinase 1/2, p65RelA, signal transducer and activator of transduction 4 (STAT4), and STAT5 activity in monokine-stimulated NK cells, potentially contributing to the reduction in IFN-gamma gene expression. Thus, SET expression is essential for suppressing PP2A phosphatase activity that would otherwise limit NK cell antitumoral and/or antiinflammatory functions by impairing NK cell production of IFN-gamma.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Interferon gama/biossíntese , Células Matadoras Naturais/metabolismo , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosfoproteínas Fosfatases/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células Cultivadas , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA , Ativação Enzimática , Regulação da Expressão Gênica , Chaperonas de Histonas , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Monocinas/farmacologia , Transdução de Sinais , Fatores de Transcrição/genética
3.
Protein Expr Purif ; 55(1): 132-8, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17513126

RESUMO

MIG (monokine induced by IFN-gamma) is a CXC-chemokine (CXCL9). It plays important roles in regulation of immune activities, and knowledge of the protein in areas of allograft transplants, autoimmune diseases, and cancer therapy is evolving quickly. The non-tagged recombinant murine MIG (rMuMIG) is therefore required to facilitate the functional studies of this important chemokine. Here we present the use of a bacteria expression system to produce non-tagged rMuMIG. The coding sequence for MIG was cloned into the pET28a (+) vector that was transformed into Escherichia coli BL21 (DE3). Expression of rMuMIG was induced by IPTG. Bacteria inclusion bodies containing the protein were isolated and washed to remove contaminated bacteria proteins, and resolved in Urea buffer. Renaturation of the denatured protein was carried out in the defined protein refolding buffer, and the refolded protein was purified using S-Sepharose cation exchange chromatography. The final preparation of the rMuMIG was more than 99% pure as measured by capillary electrophoresis and SDS-PAGE analysis. The biological activity of rMuMIG was demonstrated in a murine spleen cell chemotaxis assay with ED50 30 ng/ml. Further experiments showed that rMuMIG could inhibit proliferation of mouse bone marrow cells in vivo.


Assuntos
Quimiocinas CXC/biossíntese , Monocinas/biossíntese , Proteínas Recombinantes/biossíntese , Animais , Sequência de Bases , Células da Medula Óssea/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Quimiocina CXCL9 , Quimiocinas CXC/genética , Quimiocinas CXC/farmacologia , Quimiotaxia , Eletroforese Capilar , Eletroforese em Gel de Poliacrilamida , Escherichia coli/genética , Interferon gama/farmacologia , Interferon gama/fisiologia , Camundongos , Dados de Sequência Molecular , Monocinas/genética , Monocinas/farmacologia , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia
4.
J Cell Physiol ; 211(3): 630-7, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17192849

RESUMO

Human leucine zipper protein (LZIP) associates with CC chemokine receptor 1 (CCR1) and this protein-protein interaction should play an important role in leukocyte cell mobility. LZIP is known to regulate leukotactin-1 (Lkn-1)-dependent cell migration without affecting the chemotactic activities of other CC chemokines that bind to CCR1. Since Lkn-1 is engaged in the transcriptional activation of nuclear factor kappaB (NF-kappaB) and subsequent activation of the chemoattractant ability of leukocytes, we investigated the regulatory role of LZIP in the NF-kappaB pathway that is induced by CCR1-dependent chemokines. LZIP increased NF-kappaB-dependent luciferase activity in response to Lkn-1 in HOS/CCR1 cells and THP-1 cells. However, the NF-kappaB-dependent luciferase activities induced by other CCR1-dependent chemokines were not affected by LZIP overexpression. LZIP also increased Lkn-1-induced chemotactic activity through activation of the NF-kappaB pathway, whereas LZIP did not affect either the transactivation of NF-kappaB or the chemotactic activities induced by other CCR1-dependent chemokines. Western blot analysis showed that LZIP increased the degradation of IkappaBalpha induced by Lkn-1 but not by other CCR1-dependent chemokines. Results from electrophoretic mobility shift assay (EMSA) showed that LZIP enhanced the Lkn-1-induced DNA-binding activity of NF-kappaB. These data indicate that LZIP functions as a positive regulator in the NF-kappaB activation pathway that is triggered by Lkn-1 without affecting the transcriptional activation of NF-kappaB induced by other CCR1-dependent chemokines.


Assuntos
Quimiotaxia/fisiologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , NF-kappa B/metabolismo , Receptores de Quimiocinas/metabolismo , Transdução de Sinais/fisiologia , Linhagem Celular Tumoral , Quimiocina CCL11 , Quimiocina CCL4 , Quimiocina CCL5/metabolismo , Quimiocina CCL5/farmacologia , Quimiocinas CC/metabolismo , Quimiocinas CC/farmacologia , Quimiotaxia/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Humanos , Proteínas I-kappa B/metabolismo , Luciferases/genética , Proteínas Inflamatórias de Macrófagos/metabolismo , Proteínas Inflamatórias de Macrófagos/farmacologia , Monócitos/citologia , Monócitos/metabolismo , Monocinas/metabolismo , Monocinas/farmacologia , Inibidor de NF-kappaB alfa , Osteossarcoma , RNA Interferente Pequeno , Receptores CCR1 , Transdução de Sinais/efeitos dos fármacos
5.
Blood ; 109(6): 2481-7, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17110450

RESUMO

Natural killer (NK) cells contribute to host immunity, including tumor surveillance, through the production of interferon gamma (IFN-gamma). Although there is some knowledge about molecular mechanisms that induce IFN-gamma in NK cells, considerably less is known about the mechanisms that reduce its expression. Here, we investigate the role of the Hlx transcription factor in IFN-gamma production by NK cells. Hlx expression is induced in monokine-activated NK cells, but with delayed kinetics compared to IFN-gamma. Ectopic Hlx expression decreases IFN-gamma synthesis in primary human NK cells and IFN-gamma promoter activity in an NK-like cell line. Hlx protein levels inversely correlate with those of STAT4, a requisite factor for optimal IFN-gamma transcription. Mechanistically, we provide evidence indicating that Hlx overexpression accelerates dephosphorylation and proteasome-dependent degradation of the active Y693-phosphorylated form of STAT4. Thus, Hlx expression in activated NK cells temporally controls and limits the monokine-induced production of IFN-gamma, in part through the targeted depletion of STAT4.


Assuntos
Regulação para Baixo , Proteínas de Homeodomínio/metabolismo , Interferon gama/biossíntese , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/metabolismo , Monocinas/farmacologia , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Genes Homeobox/genética , Proteínas de Homeodomínio/genética , Humanos , Interferon gama/genética , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Fator de Transcrição STAT4/metabolismo , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
6.
Inflamm Res ; 55(5): 200-6, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16830107

RESUMO

OBJECTIVE AND DESIGN: This study examined the role of neutrophil-derived reactive oxygen species (ROS) in neutrophil recruitment into ultraviolet B (UVB)-exposed skin of mice. METHODS: Mouse dorsal skin was irradiated with UVB (600 mJ/cm2). Accumulation of neutrophils within the inflammatory sites was observed histochemically. Keratinocyte-derived chemokine (KC) and macrophage inflammatory protein 2 (MIP-2) were quantified, and in vivo chemotaxis of neutrophils toward KC and MIP-2 was examined. RESULTS: UVB exposure of mice deficient in myeloperoxidase (MPO), NADPH oxidase, or both, caused skin neutrophil infiltration peaking at 60, 48, and 48 h, respectively, which was earlier than the 72-h peak in wild-type mice. MIP-2 level was higher in mutant than wild-type mice. Mutant neutrophils produced more MIP-2 in vitro. Neutrophil migration toward a localized source of KC was higher in mutant than wild type mice. NADPH oxidase deficiency had a greater effect on migration than MPO deficiency. CONCLUSIONS: These results suggest that ROS produced by neutrophils regulate expression of MIP-2 and migration of neutrophils toward KC. This may explain the earlier infiltration of mutant neutrophils in response to UVB.


Assuntos
Dermatite/imunologia , NADPH Oxidases/deficiência , Neutrófilos/efeitos da radiação , Peroxidase/deficiência , Raios Ultravioleta , Animais , Quimiocina CXCL1 , Quimiocina CXCL2 , Quimiocinas/farmacologia , Quimiocinas CXC , Quimiotaxia de Leucócito , Dermatite/metabolismo , Dermatite/patologia , Feminino , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monocinas/farmacologia , NADPH Oxidases/genética , Neutrófilos/imunologia , Neutrófilos/metabolismo , Peroxidase/genética , Pele/imunologia , Pele/patologia , Pele/efeitos da radiação
7.
Life Sci ; 78(2): 134-9, 2005 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-16112685

RESUMO

Inhibitory effects of highly purified vitamin B2 (riboflavin-5'-sodium phosphate, >97%) on the interleukin (IL)-6, macrophage inflammatory protein (MIP)-2 and nitric oxide (NO) in LPS-induced shock mice were evaluated. Vitamin B2 at 20 mg/kg (protective effect on mice mortality induced by LPS), intravenously administered 6 h after LPS injection, significantly decreased the plasma elevated levels of IL-6 and MIP-2 at 9 and 12 h. In addition, vitamin B2 lowered the tissue concentration and the mRNA expression of IL-6 in lung and those of MIP-2 in liver at 9 h. Vitamin B2 also reduced concentration of MIP-2 concentration in lung, and inhibited mRNA expression in kidney, respectively. Vitamin B2 decreased the plasma elevated NO levels in accordance with a reduction in expression of inducible NO synthase (iNOS) both at 21 and 24 h. Accordingly, the reduction in elevated plasma cytokine levels and NO based on the inhibitory effect on local cytokine mRNA expression and iNOS would be responsible for the anti-septic effect of vitamin B2.


Assuntos
Citocinas/biossíntese , Inflamação/metabolismo , Óxido Nítrico/biossíntese , Riboflavina/farmacologia , Choque Séptico/metabolismo , Vitaminas/farmacologia , Animais , Western Blotting , Quimiocina CXCL2 , Interleucina-6/farmacologia , Rim/efeitos dos fármacos , Rim/metabolismo , Lipopolissacarídeos/farmacologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Monocinas/farmacologia , Óxido Nítrico Sintase Tipo II/biossíntese , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Exp Cell Res ; 307(2): 342-53, 2005 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-15894313

RESUMO

Adhesion, migration and invasion of endothelial cells are prerequisites for the formation of blood vessels and have to be controlled on a subcellular level. We report that subconfluent human umbilical vein endothelial cells (HUVEC) are able to constitutively form podosomal adhesions that are sites of matrix metalloprotease concentration and matrix degradation. Importantly, incubation of serum-starved cells with VEGF or TNFalpha revealed the dependence of podosomes on cytokine signaling. Podosome formation was also stimulated by addition of monocytes to HUVEC. Microinjection/application of specific inhibitors or active/inactive mutants showed that regulatory pathways include Src kinase and RhoGTPase signaling, N-WASP activation and Arp2/3 complex-dependent actin nucleation. In sum, our data show that HUVEC displaying a migratory phenotype constitutively form f-actin-rich adhesions with podosomal characteristics downstream of cytokine signaling. We propose that HUVEC podosomes play an important role in endothelial cell migration and invasion.


Assuntos
Extensões da Superfície Celular/metabolismo , Citocinas/fisiologia , Células Endoteliais/citologia , Transdução de Sinais/fisiologia , Proteínas rho de Ligação ao GTP/fisiologia , Proteína 2 Relacionada a Actina , Proteína 3 Relacionada a Actina , Actinas/metabolismo , Toxinas Bacterianas/farmacologia , Plaquetas/fisiologia , Adesão Celular/fisiologia , Comunicação Celular/fisiologia , Contagem de Células , Extensões da Superfície Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Meios de Cultura Livres de Soro/farmacologia , Citocinas/farmacologia , Proteínas do Citoesqueleto/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Proteínas de Escherichia coli/farmacologia , Matriz Extracelular/metabolismo , Humanos , Metaloproteinases da Matriz/metabolismo , Microinjeções , Microscopia de Fluorescência , Monócitos/fisiologia , Monocinas/farmacologia , Mutação/genética , Proteínas do Tecido Nervoso/farmacologia , Fosfotirosina/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Proteína Neuronal da Síndrome de Wiskott-Aldrich , Proteínas rho de Ligação ao GTP/antagonistas & inibidores , Proteínas rho de Ligação ao GTP/genética , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
9.
Blood ; 105(8): 3011-8, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15604218

RESUMO

Monocyte cytokines (ie, monokines) induce natural killer (NK) cells to produce interferon-gamma (IFN-gamma), which is critical for monocyte clearance of infectious pathogens and tumor surveillance. Human CD56bright NK cells produce far more IFN-gamma in response to monokines than do CD56dim NK cells. The kinases and phosphatases involved in regulating IFN-gamma production by monokine-activated NK cells are not clearly identified. SHIP1 is a 5' inositol phosphatase that dephosphorylates the phosphatidylinositol-3 kinase (PI-3K) product PI3,4,5P3. Here, we show that constitutive expression of SHIP1 is distinctly lower in CD56bright NK cells compared with CD56dim NK cells, suggesting it could be an important negative regulator of IFN-gamma production in monokine-activated NK cells. Indeed, overexpression of SHIP1 in CD56bright NK cells followed by monokine activation substantially lowered IFN-gamma production. This effect was not seen when NK cells were infected with a SHIP1 mutant containing an inactive catalytic domain. Finally, NK cells in SHIP1-/- mice produced more IFN-gamma in response to monokines in vivo than did NK cells from wild-type mice. Collectively, these results demonstrate that SHIP1 negatively regulates monokine-induced NK cell IFN-gamma production in vitro and in vivo and provide the first molecular explanation for an important functional distinction observed between CD56bright and CD56dim human NK subsets.


Assuntos
Antígeno CD56/genética , Células Matadoras Naturais/fisiologia , Monocinas/farmacologia , Monoéster Fosfórico Hidrolases/genética , Animais , Linhagem Celular , Regulação para Baixo/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Humanos , Fosfatos de Inositol/metabolismo , Inositol Polifosfato 5-Fosfatases , Interferon gama/genética , Interferon gama/metabolismo , Células Matadoras Naturais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Monoéster Fosfórico Hidrolases/metabolismo , Fosforilação , Estimulação Química
10.
Exp Cell Res ; 297(1): 197-211, 2004 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15194436

RESUMO

Intrahippocamal injections of kainic acid (KA) significantly increase the expression of monocyte chemoattractant protein-1 (MCP-1) and macrophage inflammatory protein-2 (MIP-2) in the ipsilateral hippocampus at 2-4 h and 21-45 days post-administration, suggesting the possible involvement of these chemokines in both neurodegenerative and regenerative processes. To examine the possible role of these chemokines on neuronal cell death, hippocampal neurons were incubated with either MCP-1 or MIP-2 in vitro and examined to assess the effects on neuronal cell viability. These treatments resulted in significant neuronal apoptosis that could be abrogated by prior treatment with the caspase-1 inhibitor, Z-VAD-FMK, the caspase-3 inhibitor, Z-DEVD-FMK, the Galphai inhibitor, pertussis toxin, or the MAO-B inhibitor, (-)deprenyl. Furthermore, this chemokine apoptotic effect could also be observed in vivo as intrahippocampal injections of MCP-1 or MIP-2 resulted in the apoptosis of hippocampal neurons, thus supporting a direct role of these chemokines in neuronal death. In contrast, immunohistological analysis of kainic acid lesions on days 21-45 revealed significant expression of MCP-1 and MIP-2 associated with reactive astrocytes and macrophages, respectively, with no apoptotic populations being observed. These results suggested that these chemokines might also mediate distinct biological effects on local microenvironmental cell populations at various stages post truama and during cellular repair. To address this possibility, astrocyte were cultured in the presence or absence of these chemokines and examined by microarray analysis for effects on astrocytes gene expression. A number of genes encoding proteins associated with inflammation, cellular signaling, differentiation, and repair were directly modulated by chemokine treatment. More specifically, the RNA and protein expression of the neurotrophic factor, basic fibroblast growth factor (bFGF), was found to be significantly increased upon culture with MCP-1 and MIP-2. Conditioned media derived from chemokine-stimulated astrocytes also facilitated bFGF-dependent neuronal cell differentiation and promoted survival of H19-7 neurons in vitro, suggesting a possible role for chemokine-activated astrocytes as a source of trophic support. Taken together, these data support possible autocrine and paracrine roles for MCP-1 and MIP-2 in both the "death and life" of hippocampal neurons following CNS injury.


Assuntos
Quimiocina CCL2/metabolismo , Encefalite/metabolismo , Monocinas/metabolismo , Degeneração Neural/metabolismo , Regeneração Nervosa/fisiologia , Doenças Neurodegenerativas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Astrócitos/efeitos dos fármacos , Astrócitos/imunologia , Astrócitos/metabolismo , Linhagem Celular Transformada , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Quimiocina CCL2/farmacologia , Quimiocina CXCL2 , Meios de Cultivo Condicionados/farmacologia , Modelos Animais de Doenças , Encefalite/imunologia , Encefalite/fisiopatologia , Inibidores Enzimáticos/farmacologia , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/metabolismo , Hipocampo/imunologia , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Ácido Caínico , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Monocinas/farmacologia , Degeneração Neural/induzido quimicamente , Degeneração Neural/imunologia , Regeneração Nervosa/efeitos dos fármacos , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/fisiopatologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Neurotoxinas , Ratos , Ratos Endogâmicos F344 , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
11.
J Med Virol ; 73(2): 300-8, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15122808

RESUMO

Respiratory syncytial virus (RSV) is a major cause of respiratory tract disease in infants, aged adults, and immunosuppressed patients. The only approved medicines for RSV disease are administration of prophylatic antibodies or treatment with a synthetic nucleoside. Both approaches are expensive and the latter is not without risk and of controversial benefit. The present investigation studied whether pharmaceutical or biologic compounds based upon chemokines might be useful in preventing RSV disease. Of interest was RANTES/CCL5, which inhibits infection by HIV strains that use chemokine receptor (CCR)-5 as co-receptor. Herein, we report that prior or simultaneous treatment of HEp-2 cells with recombinant human CCL5 provides dose-dependent inhibition of infection with RSV. Other recombinant chemokines (MIP-1alpha/CCL3, MIP-1beta/CCL4, MCP-2/CCL8, eotaxin/CCL11, MIP-1delta/CCL15, stromal cell derived factor (SDF)-1alpha/CXCL12) were not inhibitory. The data suggested that CCL5 might inhibit infection by blocking fusion (F) protein-epithelial cell interactions. Infections by mutant RSV strains deleted of small hydrophobic and/or attachment proteins and only expressing F protein in the envelope were inhibited by prior treatment with CCL5 or a biologically inactive N-terminally modified met-CCL5. Inhibition was also observed when virus adsorption and treatment with CCL5 were performed at 4 degrees C. Flow cytometry further revealed that epithelial cells were positive for CCR3, but not CCR1 or CCR5. Thus, novel mimetics of CCL5 may be useful prophylatic agents to prevent respiratory tract disease caused by RSV.


Assuntos
Quimiocinas CC/farmacologia , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Antivirais/farmacologia , Linhagem Celular Tumoral , Quimiocina CCL11 , Quimiocina CCL3 , Quimiocina CCL4 , Quimiocina CCL5 , Quimiocina CCL8 , Quimiocina CXCL12 , Quimiocinas CC/metabolismo , Quimiocinas CXC/farmacologia , Células Epiteliais/virologia , Humanos , Proteínas Inflamatórias de Macrófagos/farmacologia , Proteínas Quimioatraentes de Monócitos/farmacologia , Monocinas/farmacologia , Receptores CCR1 , Receptores CCR3 , Receptores CCR5/análise , Receptores de Quimiocinas/análise , Proteínas Recombinantes/farmacologia , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/fisiologia , Proteínas Virais de Fusão/fisiologia , Replicação Viral/efeitos dos fármacos
12.
Arch Virol ; 148(5): 871-90, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12721796

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV)/ Human herpesvirus 8 encodes three chemokines, which are called viral macrophage inflammatory protein (vMIP)-I, -II, and -III. Here, we expressed the KSHV vMIP-I and vMIP-II proteins and analyzed their biological functions. Both vMIP-I and vMIP-II had an apparent molecular mass of 7.8 kDa and were localized to the cytoplasm in a body cavity-based lymphoma cell line BC-3, stimulated with phorbol ester. We next treated a human monocytic leukemia cell line, THP-1, with purified recombinant vMIP-I and vMIP-II, or vMIP-I and vMIP-II fused with alkaline phosphatase to study Ca(2+) signalling and in vitro chemotaxis in response to these proteins. Calcium mobilization was induced by both vMIP-I and vMIP-II. Furthermore, vMIP-I and vMIP-II induced Ca(2+) mobilization in K562 cells expressing the CC chemokine receptor 5 (CCR5), suggesting that both may be agonistic for CCR5. Additionally, vMIP-I induced Ca(2+) mobilization through the intermediary of CCR8. These viral MIPs were also capable of chemotactically activating the THP-1 cells. These results imply that vMIP-I and vMIP-II may play important roles in the propagation of KS and primary effusion lymphoma by inducing the chemotaxis of CCR5-expressing monocytes.


Assuntos
Quimiotaxia , Herpesvirus Humano 8/fisiologia , Proteínas Inflamatórias de Macrófagos/metabolismo , Monócitos/citologia , Monócitos/metabolismo , Monocinas/metabolismo , Transdução de Sinais , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Quimiocina CCL1 , Quimiocina CCL11 , Quimiocina CCL4 , Quimiocina CXCL2 , Quimiocinas CC/farmacologia , Quimiotaxia/efeitos dos fármacos , Relação Dose-Resposta a Droga , Expressão Gênica , Humanos , Células K562 , Proteínas Inflamatórias de Macrófagos/genética , Proteínas Inflamatórias de Macrófagos/farmacologia , Monócitos/efeitos dos fármacos , Monocinas/genética , Monocinas/farmacologia , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/metabolismo , Sarcoma de Kaposi/virologia , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas
13.
Arthritis Rheum ; 48(3): 668-74, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12632419

RESUMO

OBJECTIVE: Adenosine deaminase (ADA; EC 3.5.4.4) activity is elevated in the synovial fluid (SF) of patients with rheumatoid arthritis (RA). Since the antiinflammatory effect of methotrexate is reportedly associated with increased levels of extracellular adenosine, the present study was undertaken to clarify the role of 2 ADA isozymes, ADA1 and ADA2, in the pathogenesis of RA. METHODS: The activities of ADA1 and ADA2 were measured in SF from RA and osteoarthritis (OA) patients, in sera from RA patients, and in lysates prepared from mononuclear and polymorphonuclear cells from SF from RA patients, peripheral blood from RA patients, and fibroblast-like synoviocytes (FLS) from RA and OA patients. Also measured were the effects of proinflammatory cytokines on ADA1 activity and ADA messenger RNA (mRNA) expression in RA FLS, as determined using real-time polymerase chain reaction. The adenosine concentration in RA SF was measured by radioimmunoassay. RESULTS: The adenosine concentration in RA SF ranged from 0.027 microM to 0.508 microM (mean +/- SD 0.156 +/- 0.132 microM). At those concentrations, ADA1 would be expected to be functionally dominant due to its higher affinity for adenosine. ADA1 activity in RA SF (mean +/- SD 14.4 +/- 8.5 IU/liter) was significantly higher than that in OA SF (3.0 +/- 1.1 IU/liter) or RA sera (3.0 +/- 0.6 IU/liter); moreover, ADA1 activity in RA FLS lysate was the highest among the cell lysates tested. Proinflammatory cytokines did not affect ADA1 activity or ADA mRNA expression in RA FLS. CONCLUSION: Elevated ADA1 activity is an intrinsic characteristic of RA FLS, which likely contributes to the pathogenesis of RA by neutralizing the antirheumatic properties of endogenous adenosine.


Assuntos
Adenosina Desaminase/biossíntese , Artrite Reumatoide/enzimologia , Fibroblastos/enzimologia , Osteoartrite/enzimologia , Líquido Sinovial/enzimologia , Adenosina/metabolismo , Adenosina Desaminase/sangue , Adenosina Desaminase/genética , Artrite Reumatoide/sangue , Artrite Reumatoide/patologia , Células Cultivadas , Fibroblastos/citologia , Humanos , Isoenzimas , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/enzimologia , Monocinas/farmacologia , Neutrófilos/citologia , Neutrófilos/enzimologia , Osteoartrite/patologia , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Líquido Sinovial/citologia
14.
J Neurochem ; 84(5): 997-1005, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12603824

RESUMO

Chemokines are important mediators in immune responses and inflammatory processes of neuroimmunologic and infectious diseases. Although chemokines are expressed predominantly by cells of the immune system, neurons also express chemokines and chemokine receptors. We report herein that human neuronal cells (NT2-N) produce macrophage inflammatory protein-1alpha and -1beta (MIP-1alpha and MIP-1beta), which could be enhanced by interleukin (IL)-1beta at both mRNA and protein levels. The addition of supernatants from human peripheral blood monocyte-derived macrophage (MDM) cultures induced MIP-1beta mRNA expression in NT2-N cells. Anti-IL-1beta antibody removed most, but not all, of the MDM culture supernatant-induced MIP-1beta mRNA expression in NT2-N cells, suggesting that IL-1beta in the MDM culture supernatants is a major factor in the induction of MIP-1beta expression. Investigation of the mechanism(s) responsible for IL-1beta-induced MIP-1alpha and -1beta expression demonstrated that IL-1beta activated nuclear factor kappa B (NF-kappaB) promoter-directed luciferase activity in NT2-N cells. Caffeic acid phenethyl ester, a potent and specific inhibitor of activation of NF-kappaB, not only blocked IL-1beta-induced activation of the NF-kappaB promoter but also decreased IL-1beta-induced MIP-1alpha and -1beta expression in NT2-N cells. These data suggest that NF-kappaB is at least partially involved in the IL-1beta-mediated action on MIP-1alpha and -1beta in NT2-N cells. IL-1beta-mediated up-regulation of beta-chemokine expression may have important implications in the immunopathogenesis of inflammatory diseases in the CNS.


Assuntos
Interleucina-1/farmacologia , Proteínas Inflamatórias de Macrófagos/biossíntese , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Linhagem Celular , Quimiocina CCL3 , Quimiocina CCL4 , Meios de Cultivo Condicionados/farmacologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Humanos , Macrófagos/metabolismo , Monocinas/biossíntese , Monocinas/farmacologia , NF-kappa B/metabolismo , Neurônios/citologia , Isoformas de Proteínas/biossíntese , Fator de Necrose Tumoral alfa/farmacologia
15.
J Hepatol ; 36(6): 766-73, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12044526

RESUMO

BACKGROUND/AIMS: Recently, we have reported that macrophage inflammatory protein-2 (MIP-2) plays a pivotal role in concanavalin A (Con A)-induced liver injury. In this study, we investigated the effect of antithrombin III (AT-III) on liver damage, and production of MIP-2 and prostacyclin in this model. METHODS: Liver injury was induced by intravenous injection of Con A (15 mg/kg) and AT-III was administered (50, 250 and 500 units/kg, iv) 30 mm before Con A injection. Plasma levels of alanine aminotransferase (ALT), MIP-2 and 6-keto-prostaglandin F1alpha (6k-PG-F1alpha), stable metabolite of prostaglandin I(2) (prostacyclin), were determined. RESULTS: The elevated plasma ALT levels 8, 16, 24 h after Con A injection were inhibited by AT-III pretreatment. The elevated plasma MIP-2 levels were significantly inhibited by AT-III pretreatment compared with vehicle treatment. The inhibitory effect of AT-III on plasma ALT and MIP-2 in Con A-induced liver injury was attenuated by indomethacin (5 mg/kg, ip). Plasma concentration of 6k-PG-F1alpha at 2 h after AT-III injection was significantly elevated compared with baseline and vehicle pretreatment. CONCLUSIONS: These findings suggest that AT-III prevents Con A-induced liver injury through an inhibition of MIP-2 release and a production of prostacyclin.


Assuntos
Antitrombina III/farmacologia , Quimiocinas/metabolismo , Epoprostenol/biossíntese , Hepatopatias/tratamento farmacológico , Hepatopatias/imunologia , Inibidores de Serina Proteinase/farmacologia , Alanina Transaminase/sangue , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Antineoplásicos/farmacologia , Doença Hepática Induzida por Substâncias e Drogas , Quimiocina CXCL2 , Concanavalina A , Citocinas/biossíntese , Epoprostenol/sangue , Feminino , Indometacina/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Monocinas/farmacologia , Neutrófilos/imunologia , Proteínas Recombinantes/farmacologia , Organismos Livres de Patógenos Específicos , Fator de Necrose Tumoral alfa/farmacologia
16.
Microvasc Res ; 60(3): 249-60, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11078641

RESUMO

The leukocyte integrins LFA-1 and Mac-1 bind to endothelial intercellular adhesion molecule-1 (ICAM-1). Leukocyte adhesion induced by micropipette injection of formylmethionylleucylphenylalanine (fMLP) or macrophage inflammatory protein 2 (MIP-2) next to a venule in the exteriorized mouse cremaster muscle was almost completely blocked after intravenous injection of the ICAM-1 mAb YN-1. In contrast, after 2-h pretreatment with TNF-alpha, leukocyte adhesion induced in postcapillary venules by fMLP or MIP-2 was not blocked by the ICAM-1 mAb. Leukocyte adhesion was significantly reduced by mAb GAME-46 to CD18 even after TNF-alpha treatment. We conclude that ICAM-1 is necessary for neutrophil adhesion to unstimulated endothelium, but not for adhesion to cytokine-stimulated endothelium. Although ICAM-1 is expressed at high levels after TNF-alpha, ICAM-1 either is not functional or is redundant with other endothelial ligands for beta(2) integrins.


Assuntos
Adesão Celular/fisiologia , Molécula 1 de Adesão Intercelular/fisiologia , Leucócitos/fisiologia , Animais , Anticorpos Monoclonais/farmacologia , Antígenos CD18/fisiologia , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Quimiocina CXCL2 , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Inflamação/patologia , Inflamação/fisiopatologia , Molécula 1 de Adesão Intercelular/imunologia , Leucócitos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monocinas/farmacologia , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Vênulas/citologia , Vênulas/fisiologia
17.
Vet Surg ; 29(1): 1-7, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10653489

RESUMO

OBJECTIVE: To determine if there is a difference in in vitro growth of fibroblasts isolated from the trunk and distal aspect of the limb of horses and ponies. To determine the effects of a corticosteroid and monokine on in vitro growth of fibroblasts isolated from the trunk and distal aspect of the limb of horses and ponies. STUDY DESIGN: Growth of fibroblasts from tissues harvested from the trunk and limb were compared from horse and pony samples grown in control media and control media with triamcinolone or monokine added. ANIMALS OR SAMPLE POPULATION: Dermal and subcutaneous tissue from 22 horses and 17 ponies of various ages and breeds. METHODS: Fibroblast growth was assessed by tritiated thymidine uptake using standard cell culture techniques. The effect of a monokine or triamcinolone plus control media were compared with control media for fibroblast growth. RESULTS: Fibroblast growth from tissues isolated from the horse limb was significantly less than growth from the horse trunk and the limb and trunk of ponies. Monokine was more effective than triamcinolone in suppressing fibroblast growth from tissues isolated from the trunk and limb in both horses and ponies. CONCLUSIONS: There are growth differences in fibroblasts isolated from the limb of horses compared with those isolated from the trunk and from the limb and trunk of ponies. CLINICAL RELEVANCE: The difference in fibroblast growth from tissues isolated from the trunk and limb of horses and ponies may provide evidence for the difference reported in the healing characteristics of limb wounds in horses and ponies. Influencing fibroblast growth may provide a key to controlling the development of exuberant granulation tissue in horses and ponies.


Assuntos
Anti-Inflamatórios/farmacologia , Fibroblastos/citologia , Cavalos/fisiologia , Monocinas/farmacologia , Pele/citologia , Triancinolona/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultura/farmacologia , Fibroblastos/efeitos dos fármacos , Membro Anterior , Cavalos/cirurgia , Cicatrização/efeitos dos fármacos , Cicatrização/fisiologia
18.
FASEB J ; 13(12): 1565-74, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10463948

RESUMO

Severe acute liver injury due to accidental or intentional acetaminophen overdose presents a major clinical dilemma often requiring liver transplantation. In the present study, liver regeneration after profound liver injury in mice challenged with acetaminophen was facilitated by the exogenous addition of ELR-containing CXC chemokines such as macrophage inflammatory protein-2 (MIP-2), epithelial neutrophil-activating protein-78 (ENA-78), or interleukin 8. Intravenous administration of ELR-CXC chemokines or N-acetyl-cysteine (NAC) immediately after acetaminophen challenge in mice significantly reduced histological and biochemical markers of hepatic injury. However, when the intervention was delayed until 10 h after acetaminophen challenge, only ELR-CXC chemokines significantly reduced liver injury and mouse mortality. The delayed addition of ELR-CXC chemokines to cultured hepatocytes maintained the proliferation of these cells in a CXCR2-dependent fashion after acetaminophen challenge whereas delayed NAC treatment did not. These observations demonstrate that ELR-CXC chemokines represent novel hepatic regenerative factors that exhibit prolonged therapeutic effects after acetaminophen-induced hepatotoxicity.


Assuntos
Quimiocinas CXC/farmacologia , Interleucina-8/análogos & derivados , Interleucina-8/farmacologia , Regeneração Hepática/fisiologia , Fígado/fisiologia , Monocinas/farmacologia , Receptores de Quimiocinas/fisiologia , Receptores de Interleucina/fisiologia , Acetaminofen/toxicidade , Acetilcisteína/farmacologia , Animais , Aspartato Aminotransferases/sangue , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Quimiocina CXCL2 , Quimiocina CXCL5 , Fatores Quimiotáticos/farmacologia , Feminino , Fator de Crescimento de Hepatócito/farmacologia , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/patologia , Regeneração Hepática/efeitos dos fármacos , Camundongos , Receptores de Interleucina-8B
19.
Atherosclerosis ; 144(2): 375-80, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10407498

RESUMO

Human vascular smooth muscle cells (VSMC) are a component of blood vessels, and secrete a variety of cytokines in atherosclerotic loci. Interleukin-11 (IL-11), a member of IL-6-like cytokines, is reported to be involved in inflammation and tissue remodeling, both of which are observed in atherosclerosis. However, no information is available as to the production of IL-11 by VSMC. Therefore, the expression of IL-11 in VSMC is investigated. The amounts of IL-11 protein and mRNA were determined by enzyme-linked immunosorbent assay (ELISA) and Northern blot analysis, respectively. The expression of IL-11 in VSMC was also immunohistochemically determined. IL-1 alpha, transforming growth factor-beta (TGF beta) and, to a lesser extent, tumor necrosis factor-alpha (TNF alpha) stimulated the IL-11 production by VSMC, and the stimulatory effects of IL-1 alpha and TGF beta on IL-11 production were dose-dependent. IL-1 alpha and TNF alpha synergistically augmented TGF beta-stimulated IL-11 production by VSMC. Immunohistochemical staining also revealed the expression of IL-11 protein in VSMC. Furthermore, IL-1 alpha, TGF beta, and TNF alpha induced IL-11 gene expression in VSMC. Because IL-6-like cytokines are reported to be cytoprotective, monokine-stimulated IL-11 may have a potent protective role in atherosclerotic lesions.


Assuntos
Interleucina-11/metabolismo , Monocinas/farmacologia , Músculo Liso Vascular/imunologia , Arteriosclerose/imunologia , Células Cultivadas , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-1/farmacologia , Interleucina-11/genética , Linfotoxina-alfa/farmacologia , Proteínas Recombinantes/farmacologia , Estimulação Química , Fator de Necrose Tumoral alfa/farmacologia
20.
Biochem Biophys Res Commun ; 260(3): 626-33, 1999 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-10403817

RESUMO

Patients with cancer cachexia exhibit increased glucose flux and lactate production in skeletal muscle. The aim of this study was to examine the direct effect of cancer cell-conditioned media on glucose metabolism in L6 myoblasts. Media from PANC-1 and Colo 320 cells caused a marked time-dependent and concentration-dependent increase of 2-deoxyglucose uptake in GLUT-4 transfected L6 myoblasts. This effect was greater than maximal acute stimulation by insulin and the effect of insulin was additive. Glucose utilization and lactate production increased in parallel to glucose uptake. The effect was inhibited by the protein synthesis inhibitor, cycloheximide and the glucose transport inhibitor, cytochalasin B. The bioactive factor had a molecular weight of approximately 5,000 and the biological activity was destroyed by proteinase K digestion. Radioimmunoassay and immunoneutralization studies indicated the major factor involved is not TNFalpha, IL-1beta, insulin, IGF-I or IGF-II. Further purification and characterization are needed to reveal the identity of this novel factor or factors which may have other metabolic effects that contribute to the cancer cachexia and insulin resistance.


Assuntos
Fatores Biológicos/farmacologia , Neoplasias do Colo/metabolismo , Glucose/metabolismo , Ácido Láctico/biossíntese , Proteínas Musculares , Músculos/metabolismo , Neoplasias Pancreáticas/metabolismo , Animais , Fatores Biológicos/análise , Fatores Biológicos/antagonistas & inibidores , Linhagem Celular , Meios de Cultivo Condicionados/química , Meios de Cultivo Condicionados/farmacologia , Cicloeximida/farmacologia , Citocalasina B/farmacologia , Desoxiglucose/metabolismo , Relação Dose-Resposta a Droga , Endopeptidase K/metabolismo , Transportador de Glucose Tipo 4 , Humanos , Insulina/análise , Insulina/farmacologia , Peso Molecular , Monocinas/análise , Monocinas/farmacologia , Proteínas de Transporte de Monossacarídeos/genética , Proteínas de Transporte de Monossacarídeos/metabolismo , Músculos/citologia , Músculos/efeitos dos fármacos , Ratos , Somatomedinas/análise , Somatomedinas/farmacologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA