Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Free Radic Biol Med ; 32(5): 406-13, 2002 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11864780

RESUMO

Vascular injury leads to the production of reactive oxygen species (ROS), but the mechanisms by which ROS contribute to vascular pathology are not completely understood. We hypothesized that ROS increase endothelin converting enzyme (ECE-1) expression. We found that glucose oxidase (GO) increases ECE-1 mRNA, protein, and activity in bovine aortic endothelial cells. Catalase abolishes this effect. Glucose oxidase treatment of endothelial cells transactivates the ECE-1 promoter. The ECE-1 promoter element that mediates this response to GO is located between -444 and -216 bp. This region contains a STAT response element, and GO activates STAT-3 binding to this STAT response element. Our data suggest that STAT3 mediates hydrogen peroxide induction of ECE-1 expression.


Assuntos
Antioxidantes/farmacologia , Ácido Aspártico Endopeptidases/metabolismo , Endotélio Vascular/enzimologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Glucose Oxidase/farmacologia , Peróxido de Hidrogênio/farmacologia , Regiões Promotoras Genéticas/genética , Espécies Reativas de Oxigênio/metabolismo , Animais , Aorta/metabolismo , Ácido Aspártico Endopeptidases/genética , Western Blotting , Catalase/metabolismo , Bovinos , Núcleo Celular , Células Cultivadas , Citosol , Primers do DNA/química , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Endotelina-1/metabolismo , Enzimas Conversoras de Endotelina , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Células HeLa , Humanos , Luciferases/metabolismo , Metaloendopeptidases , Reação em Cadeia da Polimerase , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Fator de Transcrição STAT3 , Deleção de Sequência , Transativadores/genética , Transativadores/metabolismo , Transfecção
2.
Circulation ; 104(19): 2369-75, 2001 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-11696480

RESUMO

BACKGROUND: Inducible nitric oxide synthase (iNOS, or NOS2) reduces the severity of accelerated graft arteriosclerosis (AGA) in transplanted organs, although the precise mechanism is unclear. METHODS AND RESULTS: We transplanted wild-type murine hearts into either wild-type or NOS2-null recipient mice; we then measured cardiac allograft survival and analyzed tissue sections by immunohistochemistry. We have confirmed that NOS2 increases cardiac allograft survival. We now show that there is less inflammation of cardiac allografts in wild-type hosts than in NOS2-null hosts. Furthermore, staining for von Willebrand factor reveals that the presence of NOS2 is correlated with the presence of Weibel-Palade bodies inside endothelial cells, whereas the absence of NOS2 is correlated with the release of Weibel-Palade bodies. CONCLUSIONS: Weibel-Palade bodies contain mediators that promote thrombosis and inflammation. Therefore, nitric oxide (NO) may stabilize the vessel wall and prevent endothelial activation in part by inhibiting the release of the contents of Weibel-Palade bodies. Prevention of Weibel-Palade body release might be a mechanism by which NO protects the vessel wall from inflammatory disorders such as atherosclerosis or graft arteriosclerosis.


Assuntos
Rejeição de Enxerto/patologia , Transplante de Coração , Óxido Nítrico Sintase/metabolismo , Transplante Homólogo/patologia , Corpos de Weibel-Palade/patologia , Animais , Progressão da Doença , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Endotélio Vascular/ultraestrutura , Imunofluorescência , Rejeição de Enxerto/genética , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/genética , Sobrevivência de Enxerto/imunologia , Transplante de Coração/imunologia , Imuno-Histoquímica , Inflamação/enzimologia , Inflamação/genética , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Microscopia Eletrônica , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Transplante Homólogo/imunologia , Corpos de Weibel-Palade/metabolismo , Corpos de Weibel-Palade/ultraestrutura , Fator de von Willebrand/biossíntese
3.
Cancer Res ; 61(9): 3682-8, 2001 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-11325839

RESUMO

Activated macrophages play a central role in antitumor immunity. However, the stimuli that activate macrophages to kill tumor cells are not completely understood. Because the center of solid tumors can be hypoxic, we hypothesized that hypoxia may be an important signal in activating macrophages to kill tumor cells. Hypoxia stimulates IFN-primed macrophages to express the inducible nitric oxide synthase (NOS2) and to synthesize nitric oxide (NO). We show that this synergy between IFN and hypoxia is mediated by the direct interaction of the hypoxia inducible factor-1 (HIF-1) and IFN regulatory factor-1 (IRF-1), which are both required for the hypoxic transcription of NOS2. This interaction between HIF-1 and IRF-1 may explain the mechanism by which macrophages infiltrating into tumors are activated to express NOS2 and to produce NO, a mediator of tumor apoptosis.


Assuntos
Apoptose/fisiologia , Proteínas de Ligação a DNA/fisiologia , Interferon gama/fisiologia , Macrófagos/imunologia , Óxido Nítrico/fisiologia , Proteínas Nucleares/fisiologia , Fosfoproteínas/fisiologia , Transdução de Sinais/fisiologia , Animais , Sítios de Ligação , Hipóxia Celular/fisiologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Indução Enzimática , Feminino , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Fator Regulador 1 de Interferon , Interferon gama/farmacologia , Ativação de Macrófagos/fisiologia , Macrófagos/enzimologia , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico Sintase/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Saccharomyces cerevisiae/genética , Fatores de Transcrição/fisiologia , Ativação Transcricional , Transfecção
5.
J Mol Cell Cardiol ; 33(1): 109-19, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11133227

RESUMO

Although redox-sensitive transcription factors, including nuclear factor kappa B (NF kappa B) and activator protein-1 (AP-1), have been shown to induce intercellular adhesion molecule-1 (ICAM-1) gene transcription in isolated cells, little is known about their involvement in the regulation of the ICAM-1 gene in vivo during ischemia-reperfusion. Anesthetized closed-chest dogs underwent 90 min coronary artery occlusion, followed by reperfusion for 0, 15, 30, 60, 180, or 360 min. Blood flow (fluorescent or radioactive microspheres), ICAM-1 protein expression (immunohistochemistry), ICAM-1 gene activation (Northern blotting), and nuclear DNA-binding activity of NF kappa B and AP-1 (electrophoretic mobility shift assays) were assessed in myocardial tissue samples. ICAM-1 protein was expressed constitutively on vascular endothelium, but expression levels decreased markedly during ischemia. Within 15 min reperfusion, endothelial ICAM-1 protein increased, associated with a rapid appearance of ICAM-1 mRNA. Activation of both NF kappa B and AP-1 occurred following ischemia-reperfusion, but did not coincide temporally with early post-reperfusion ICAM-1 gene induction. NF kappa B was activated during ischemia, when ICAM-1 mRNA was undetectable, and did not increase further until 60 min reperfusion, well after the increase in ICAM-1 mRNA had begun. Similarly, AP-1 did not increase until 60 min reperfusion. In non-ischemic myocardium, NF kappa B and AP-1 were both activated, but ICAM-1 mRNA did not appear until 6 h later. By immunohistology, NF kappa B (p65 subunit) and the c-Fos subunit of AP-1 were localized primarily in vascular endothelium. Reperfusion of ischemic myocardium is associated with very rapid ICAM-1 gene induction in the context of prior NF kappa B activation, without new activation of NF kappa B. In non-ischemic myocardium, ICAM-1 transcription begins hours after NF kappa B is activated. These findings support a role for NF kappa B in ICAM-1 induction in vivo, but suggest that other processes, such as oxygen-radical generation, may combine with NF kappa B to trigger an accelerated transcription of ICAM-1 following ischemia-reperfusion.


Assuntos
Regulação da Expressão Gênica , Molécula 1 de Adesão Intercelular/biossíntese , Isquemia Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/genética , NF-kappa B/metabolismo , Animais , Arteríolas/metabolismo , Arteríolas/patologia , Northern Blotting , Capilares/metabolismo , Capilares/patologia , Núcleo Celular/metabolismo , Cães , Endotélio Vascular/metabolismo , Feminino , Inflamação , Molécula 1 de Adesão Intercelular/genética , Masculino , Isquemia Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Especificidade de Órgãos , Oxirredução , Subunidades Proteicas , RNA Mensageiro/biossíntese , Fatores de Tempo , Fator de Transcrição AP-1/metabolismo , Ativação Transcricional
6.
J Infect Dis ; 183(2): 232-238, 2001 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11120929

RESUMO

Chlamydia pneumoniae infection can exacerbate atherosclerosis in animals. To test the hypothesis that antibiotic therapy inhibits the atherogenic effects of C. pneumoniae infection, 10-week-old apolipoprotein E (ApoE) null mice were infected with C. pneumoniae or placebo, were treated for 2 weeks after infection with azithromycin or placebo, and were killed at 20 weeks of age. Infection did not affect the size of the aortic lesion, and antibiotic treatment had no effect. Another group of mice, 12-week-old ApoE mice, were infected with C. pneumoniae or placebo, were treated for 2 weeks after infection with azithromycin or placebo, and were killed at 26 weeks of age. C. pneumoniae infection increased the size of the lesion in infected mice, but azithromycin did not reduce the size of the aortic lesion in infected mice. Therefore, immediate therapy of acute infection may be necessary to prevent the proatherogenic effects of C. pneumoniae infection.


Assuntos
Antibacterianos/uso terapêutico , Arteriosclerose/tratamento farmacológico , Azitromicina/uso terapêutico , Infecções por Chlamydophila/tratamento farmacológico , Chlamydophila pneumoniae/patogenicidade , Animais , Anticorpos Antibacterianos/sangue , Aorta/patologia , Apolipoproteínas E/deficiência , Arteriosclerose/microbiologia , Arteriosclerose/patologia , Infecções por Chlamydophila/complicações , Infecções por Chlamydophila/microbiologia , Infecções por Chlamydophila/patologia , Chlamydophila pneumoniae/imunologia , Chlamydophila pneumoniae/isolamento & purificação , Feminino , Lipídeos/análise , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase
7.
Am J Physiol Lung Cell Mol Physiol ; 279(4): L707-15, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11000131

RESUMO

Pulmonary inflammation and fibrosis are characterized by increased turnover and production of the extracellular matrix as well as an impairment of lung fibrinolytic activity. Although fragments of the extracellular matrix component hyaluronan induce macrophage production of inflammatory mediators, the effect of hyaluronan on the fibrinolytic mediators plasminogen activator inhibitor (PAI)-1 and urokinase-type plasminogen activator (uPA) is unknown. This study demonstrates that hyaluronan fragments augment steady-state mRNA, protein, and inhibitory activity of PAI-1 as well as diminish the baseline levels of uPA mRNA and inhibit uPA activity in an alveolar macrophage cell line. Hyaluronan fragments alter macrophage expression of PAI-1 and uPA at the level of gene transcription. Similarly, hyaluronan fragments augment PAI-1 and diminish uPA mRNA levels in freshly isolated inflammatory alveolar macrophages from bleomycin-treated rats. These data suggest that hyaluronan fragments influence alveolar macrophage expression of PAI-1 and uPA and may be a mechanism for regulating fibrinolytic activity during lung inflammation.


Assuntos
Regulação da Expressão Gênica/fisiologia , Ácido Hialurônico/farmacologia , Macrófagos Alveolares/fisiologia , Macrófagos/fisiologia , Inibidor 1 de Ativador de Plasminogênio/genética , Ativador de Plasminogênio Tipo Uroquinase/genética , Amilorida/farmacologia , Animais , Células da Medula Óssea/citologia , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos Alveolares/efeitos dos fármacos , Camundongos , Biossíntese de Proteínas/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Transcrição Gênica/efeitos dos fármacos
8.
J Biol Chem ; 275(34): 26423-7, 2000 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-10833511

RESUMO

Reactive oxygen species (ROS) act as signaling molecules in the cardiovascular system, regulating cellular proliferation and migration. However, an excess of ROS can damage cells and alter endothelial cell function. We hypothesized that endogenous mechanisms protect the vasculature from excess levels of ROS. We now show that superoxide can inhibit endothelin-converting enzyme activity (ECE) and decrease endothelin-1 synthesis. Superoxide inhibits ECE but hydrogen peroxide and nitric oxide do not. Superoxide inhibits ECE by ejecting zinc from the enzyme, and the addition of exogenous zinc restores enzymatic activity. Superoxide may inhibit other zinc metalloproteinases by a similar mechanism and may thus play an important role in regulating the biology of blood vessels.


Assuntos
Ácido Aspártico Endopeptidases/metabolismo , Superóxidos/metabolismo , Animais , Bovinos , Células Cultivadas , Endotelina-1/metabolismo , Enzimas Conversoras de Endotelina , Endotélio Vascular/metabolismo , Peróxido de Hidrogênio/farmacologia , Metaloendopeptidases , Óxido Nítrico/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Xantina/farmacologia , Xantina Oxidase/metabolismo , Zinco/metabolismo
9.
J Biol Chem ; 275(11): 8183-9, 2000 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-10713142

RESUMO

Nitric oxide (NO) plays an important role in airway function, and endothelial NO synthase (eNOS) is expressed in airway epithelium. To determine the basis of cell-specific eNOS expression in airway epithelium, studies were performed in NCI-H441 human bronchiolar epithelial cells transfected with the human eNOS promoter fused to luciferase. Transfection with 1624 base pairs of sequence 5' to the initiation ATG (position -1624) yielded a 19-fold increase in promoter activity versus vector alone. No activity was found in lung fibroblasts, which do not express eNOS. 5' deletions from -1624 to -279 had modest effects on promoter activity in H441 cells. Further deletion to -248 reduced activity by 65%, and activity was lost with deletion to -79. Point mutations revealed that the GATA binding motif at -254 is mandatory for promoter activity and that the positive regulatory element between -248 and -79 is the Sp1 binding motif at -125. Electrophoretic mobility shift assays yielded two complexes with the GATA site and three with the Sp1 site. Immunodepletion with antiserum to GATA-2 prevented formation of the slowest migrating GATA complex, and antiserum to Sp1 supershifted the slowest migrating Sp1 complex. An electrophoretic mobility shift assay with H441 versus fibroblast nuclei revealed that the slowest migrating GATA complex is unique to airway epithelium. Thus, cell-specific eNOS expression in airway epithelium is dependent on the interaction of GATA-2 with the core eNOS promoter, and the proximal Sp1 binding site is also an important positive regulatory element.


Assuntos
Óxido Nítrico Sintase/genética , Mucosa Respiratória/enzimologia , Sítios de Ligação , Brônquios/enzimologia , Proteínas de Ligação a DNA/metabolismo , Endotélio Vascular/enzimologia , Fatores de Ligação de DNA Eritroide Específicos , Fibroblastos/enzimologia , Fator de Transcrição GATA2 , Regulação Enzimológica da Expressão Gênica , Genes Reporter , Humanos , Luciferases/biossíntese , Luciferases/genética , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico Sintase Tipo III , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Distribuição Tecidual , Fatores de Transcrição/metabolismo
10.
FASEB J ; 14(2): 418-29, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10657998

RESUMO

Reperfusion of ischemic tissue results in the generation of reactive oxygen species that contribute to tissue injury. The sources of reactive oxygen species in reperfused tissue are not fully characterized. We hypothesized that the small GTPase Rac1 mediates the oxidative burst in reperfused tissue and thereby contributes to reperfusion injury. In an in vivo model of mouse hepatic ischemia/reperfusion injury, recombinant adenoviral expression of a dominant negative Rac1 (Rac1N17) completely suppressed the ischemia/reperfusion-induced production of reactive oxygen species and lipid peroxides, activation of nuclear factor-kappa B, and resulted in a significant reduction of acute liver necrosis. Expression of Rac1N17 also suppressed ischemia/reperfusion-induced acute apoptosis. The protection offered by Rac1N17 was also evident in knockout mice deficient for the gp91phox component of the phagocyte NADPH oxidase. This work demonstrates the crucial role of a Rac1-regulated oxidase in mediating the production of injurious reactive oxygen species, which contribute to acute necrotic and apoptotic cell death induced by ischemia/reperfusion in vivo. Targeted inhibition of this oxidase, which is distinct from the phagocyte NADPH oxidase, should provide a new avenue for in vivo therapy aimed at protecting organs at risk from ischemia/reperfusion injury.-Ozaki, M., Deshpande, S. S., Angkeow, P., Bellan, J., Lowenstein, C. J., Dinauer, M. C., Goldschmidt-Clermont, P. J., Irani, K. Inhibition of the Rac1 GTPase protects against nonlethal ischemia/reperfusion-induced necrosis and apoptosis in vivo.


Assuntos
Fígado/irrigação sanguínea , NADPH Oxidases/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Peroxidação de Lipídeos , Fígado/patologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , NADPH Oxidase 2 , NADPH Oxidases/genética , NF-kappa B/metabolismo , Necrose , Fagócitos/enzimologia , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes , Proteínas rac1 de Ligação ao GTP/genética
11.
J Immunol ; 163(10): 5497-504, 1999 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-10553076

RESUMO

Coxsackievirus infection causes myocarditis and pancreatitis in humans. In certain strains of mice, Coxsackievirus causes a severe pancreatitis. We explored the role of NO in the host immune response to viral pancreatitis. Coxsackievirus replicates to higher titers in mice lacking NO synthase 2 (NOS2) than in wild-type mice, with particularly high viral titers and viral RNA levels in the pancreas. Mice lacking NOS have a severe, necrotizing pancreatitis, with elevated pancreatic enzymes in the blood and necrotic acinar cells. Lack of NOS2 leads to a rapid increase in the mortality of infected mice. Thus, NOS2 is a critical component in the immune response to Coxsackievirus infection.


Assuntos
Infecções por Coxsackievirus/enzimologia , Infecções por Coxsackievirus/prevenção & controle , Óxido Nítrico Sintase/fisiologia , Pancreatite/enzimologia , Pancreatite/prevenção & controle , Doença Aguda , Animais , Infecções por Coxsackievirus/genética , Infecções por Coxsackievirus/mortalidade , Encefalite Viral/etiologia , Enterovirus Humano B , Macrófagos/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocardite/etiologia , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico Sintase/deficiência , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Pâncreas/enzimologia , Pâncreas/virologia , Pancreatite/genética , Pancreatite/mortalidade
12.
J Biol Chem ; 274(42): 30250-7, 1999 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-10514518

RESUMO

A variety of transcriptional and post-transcriptional mechanisms regulate the expression of the inducible nitric-oxide synthase (iNOS, or NOS2). Although neurons and endothelial cells express proteins that interact with and inhibit neuronal NOS and endothelial NOS, macrophage proteins that inhibit NOS2 have not been identified. We show that murine macrophages express a 110-kDa protein that interacts with NOS2, which we call NOS-associated protein-110 kDa (NAP110). NAP110 directly interacts with the amino terminus of NOS2, and inhibits NOS catalytic activity by preventing formation of NOS2 homodimers. Expression of NAP110 may be a mechanism by which macrophages expressing NOS2 protect themselves from cytotoxic levels of nitric oxide.


Assuntos
Óxido Nítrico Sintase/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Linhagem Celular , Dimerização , Camundongos , Dados de Sequência Molecular , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Isoformas de Proteínas/metabolismo , Especificidade por Substrato , Tunicamicina/farmacologia , Técnicas do Sistema de Duplo-Híbrido
14.
J Mol Biol ; 289(3): 459-71, 1999 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-10356322

RESUMO

We investigated the molecular mechanism for the synergistic induction of inducible nitric oxide synthase transcription by TNF-alpha and IFN-gamma. Since TNF-alpha and IFN-gamma stimulate cells in part by activating NF-kappaB and IRF-1, we hypothesized that these two transcription factors interact with each other. IRF-1 and NF-kappaB co-localize in the nucleus of stimulated macrophages. Co-immunoprecipitation experiments show that IRF-1 and NF-kappaB interact in stimulated but not resting cells. Super-shift experiments show that IRF-1 and NF-kappaB interact while binding to their respective DNA binding sites. These results demonstrate the existence of a physical interaction between IRF-1 and NF-kappaB proteins in vivo. We next suggested that this interaction between IRF-1 and NF-kappaB bends the DNA of the iNOS promoter region. Using a cyclization assay, we demonstrate that nuclear extracts from stimulated cells accelerate the rate of conversion of a linear to circular DNA, compared to extracts from resting cells. However, stimulated nuclear extracts cannot affect the rate of cyclization of a promoter with a mutant IRE or kappaB site. Furthermore, stimulated nuclear extracts depleted of IRF-1 and NF-kappaB cannot induce cyclization. We conclude that IRF-1 and NF-kappaB interact in vivo, and that this interaction physically bends the indicible nitric oxide synthase promoter DNA. This interaction may explain the mechanism by which IFN-gamma synergistically augments inducible nitric oxide synthase transcription.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Interferon gama/metabolismo , NF-kappa B/metabolismo , Óxido Nítrico Sintase/genética , Fosfoproteínas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Sítios de Ligação , Linhagem Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , DNA/química , DNA/metabolismo , Fator Regulador 1 de Interferon , Interferon gama/farmacologia , Camundongos , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Testes de Precipitina , Regiões Promotoras Genéticas , Fatores de Transcrição/metabolismo , Transcrição Gênica , Fator de Necrose Tumoral alfa/farmacologia
15.
J Immunol ; 162(7): 4171-6, 1999 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-10201943

RESUMO

Although the metalloproteinase murine metalloelastase (MME) has been implicated in lung disorders such as emphysema and pulmonary fibrosis, the mechanisms regulating MME expression are unclear. Low m.w. fragments of the extracellular matrix component hyaluronan (HA) that accumulate at sites of lung inflammation are capable of inducing inflammatory gene expression in macrophages (Mphi). The purpose of this study was to examine the effect of HA fragments on the expression of MME in alveolar Mphi. The mouse alveolar Mphi cell line MH-S was stimulated with HA fragments over time, total RNA was isolated, and Northern blot analysis was performed. HA fragments induced MME mRNA in a time-dependent fashion, with maximal levels at 6 h. HA fragments also induced MME protein expression as well as enzyme activity. The induction of MME gene expression was specific for low m.w. HA fragments and dependent upon new protein synthesis; it occurred at the level of gene transcription. We also examined the effect of HA fragments on MME expression in inflammatory alveolar Mphi from bleomycin-injured rat lungs. Although normal rat alveolar Mphi did not express MME mRNA in response to HA fragments, alveolar Mphi from the bleomycin-treated rats responded to HA fragment stimulation by increasing MME mRNA levels. Furthermore, baseline and HA fragment-induced MME gene expression in alveolar Mphi from bleomycin-treated rats was inhibited by IFN-gamma. These data suggest that HA fragments may be an important mechanism for the expression of MME by Mphi in inflammatory lung disorders.


Assuntos
Ácido Hialurônico/farmacologia , Macrófagos Alveolares/enzimologia , Metaloendopeptidases/biossíntese , Animais , Bleomicina/farmacologia , Linhagem Celular , Modelos Animais de Doenças , Relação Dose-Resposta Imunológica , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/imunologia , Indução Enzimática/efeitos dos fármacos , Indução Enzimática/imunologia , Feminino , Humanos , Macrófagos Alveolares/efeitos dos fármacos , Masculino , Metaloproteinase 12 da Matriz , Metaloendopeptidases/genética , Metaloendopeptidases/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Peso Molecular , Biossíntese de Proteínas , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley
16.
Ann Thorac Surg ; 67(1): 65-71, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10086526

RESUMO

BACKGROUND: Prolonged hypothermic circulatory arrest (HCA) causes neurologic injury. However, the mechanism of this injury is unknown. We hypothesized that HCA causes nitric oxide production to result in neuronal necrosis. This study was undertaken to determine whether the neuronal nitric oxide synthase inhibitor 17477AR reduces necrosis after HCA. METHODS: Thirty-two dogs underwent 2 hours of HCA at 18 degrees C. Nitric oxide synthase catalytic assay and intracerebral microdialysis for nitric oxide production were performed in acute nonsurvival experiments (n = 16). Sixteen animals survived for 72 hours after HCA: Group 1 (n = 9) was treated with 17477AR (Astra Arcus), and group 2 (n = 7) received vehicle only. Animals were scored from 0 (normal) to 500 (coma) for neurologic function and from 0 (normal) to 100 (severe) for neuronal necrosis. RESULTS: Administration of 17477AR reduced nitric oxide production in the striatum by 94% (HCA alone), 3.65+/-2.42 micromol/L; HCA and 17477AR, 0.20+/-0.14 micromol/L citrulline). Dogs treated with 17477AR after HCA had superior neurologic function (62.22+/-29.82 for group 1 versus 141.86+/-61.53 for group 2, p = 0.019) and significantly reduced neuronal necrosis (9.33+/-4.67 for group 1 versus 38.14+/-2.23 for group 2, p<0.00001) compared with untreated HCA dogs. CONCLUSIONS: Our results provide evidence that neuronal nitric oxide synthase mediates neuronal necrosis after HCA and plays a significant role in HCA-induced neurotoxicity. Pharmacologic strategies to inhibit neuronal nitric oxide synthase after the ischemic period of HCA may be clinically beneficial.


Assuntos
Lesões Encefálicas/metabolismo , Parada Cardíaca Induzida , Neurônios/patologia , Óxido Nítrico Sintase/fisiologia , Amidinas/farmacologia , Animais , Lesões Encefálicas/patologia , Cães , Inibidores Enzimáticos/farmacologia , Hipotermia Induzida , Masculino , Microdiálise , Necrose , Óxido Nítrico Sintase/antagonistas & inibidores
17.
Immunity ; 10(1): 21-8, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10023767

RESUMO

Although nitric oxide (NO) kills or inhibits the replication of a variety of intracellular pathogens, the antimicrobial mechanisms of NO are unknown. Here, we identify a viral protease as a target of NO. The life cycle of many viruses depends upon viral proteases that cleave viral polyproteins into individual polypeptides. NO inactivates the Coxsackievirus protease 3C, an enzyme necessary for the replication of Coxsackievirus. NO S-nitrosylates the cysteine residue in the active site of protease 3C, inhibiting protease activity and interrupting the viral life cycle. Substituting a serine residue for the active site cysteine renders protease 3C resistant to NO inhibition. Since cysteine proteases are critical for virulence or replication of many viruses, bacteria, and parasites, S-nitrosylation of pathogen cysteine proteases may be a general mechanism of antimicrobial host defenses.


Assuntos
Antivirais/farmacologia , Cisteína Endopeptidases/metabolismo , Inibidores de Cisteína Proteinase/fisiologia , Enterovirus Humano B/enzimologia , Óxido Nítrico/fisiologia , Proteases Virais 3C , Substituição de Aminoácidos/genética , Sítios de Ligação , Cisteína/genética , Cisteína/metabolismo , Cisteína Endopeptidases/genética , Inibidores de Cisteína Proteinase/farmacologia , Enterovirus Humano B/efeitos dos fármacos , Células HeLa , Humanos , Hidrólise/efeitos dos fármacos , Mutagênese Sítio-Dirigida , Óxido Nítrico/farmacologia , Compostos Nitrosos/metabolismo , Serina/genética , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo
18.
J Biol Chem ; 274(2): 993-9, 1999 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-9873042

RESUMO

Nitric oxide (NO) acts as a neurotransmitter. However, excess NO produced from neuronal NO synthase (nNOS) or inducible NOS (iNOS) during inflammation of the central nervous system can be neurotoxic, disrupting neurotransmitter and hormone production and killing neurons. A screen of a hippocampal cDNA library showed that a unique region of the iNOS protein interacts with Kalirin, previously identified as an interactor with a secretory granule peptide biosynthetic enzyme. Kalirin associates with iNOS in vitro and in vivo and inhibits iNOS activity by preventing the formation of iNOS homodimers. Expression of exogenous Kalirin in pituitary cells dramatically reduces iNOS inhibition of ACTH secretion. Thus Kalirin may play a neuroprotective role during inflammation of the central nervous system by inhibiting iNOS activity.


Assuntos
Proteínas de Transporte , Inibidores Enzimáticos/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Fatores de Troca do Nucleotídeo Guanina , Óxido Nítrico Sintase/antagonistas & inibidores , Hormônio Adrenocorticotrópico/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Dimerização , Camundongos , Dados de Sequência Molecular , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Proteínas Recombinantes/metabolismo , Células Tumorais Cultivadas
19.
J Biol Chem ; 273(40): 25922-8, 1998 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-9748268

RESUMO

We have constructed a recombinant adenovirus (Ad.Id1) that allows for efficient expression of the helix-loop-helix protein Id1. After infection with Ad.Id1, neonatal cardiac myocytes display a significant reduction in viability, which was proportional to the level of Id1 expression. A similar effect was observed in adult myocytes. Morphological and biochemical assays demonstrated that Id1 expression resulted in myocyte apoptosis. In contrast, expression of Id1 in endothelial cells, vascular smooth muscle cells, or fibroblasts did not affect the viability of these cells. Along with the induction of apoptosis, the expression of Id1 in neonatal cardiac myocytes resulted in an increase in the level of intracellular reactive oxygen species. The source of these reactive oxygen species appears to be the mitochondria. Reducing the ambient oxygen concentration or treatment with a cell-permeant H2O2 scavenger prevented Id1-stimulated apoptosis in cardiac myocytes. These results suggest that the expression of Id1 leads to the induction of apoptosis in cardiac myocytes through a redox-dependent mechanism.


Assuntos
Apoptose/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Sequências Hélice-Alça-Hélice/genética , Miocárdio/metabolismo , Proteínas Repressoras , Fatores de Transcrição/fisiologia , Adenoviridae/genética , Animais , Sobrevivência Celular/genética , Células Cultivadas , Fragmentação do DNA/fisiologia , Imunofluorescência , Sequestradores de Radicais Livres/metabolismo , Vetores Genéticos/genética , Imuno-Histoquímica , Proteína 1 Inibidora de Diferenciação , Microscopia Eletrônica , Mitocôndrias Cardíacas/metabolismo , Miocárdio/citologia , Cadeias Pesadas de Miosina/análise , Oxirredução , Oxigênio/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
20.
Surgery ; 124(2): 298-306, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9706152

RESUMO

BACKGROUND: Prolonged hypothermic circulatory arrest (HCA) causes clinical neurologic injury. This injury involves neuronal apoptosis, or programmed cell death. We have previously demonstrated that HCA causes glutamate excitotoxicity, increased nitric oxide (NO) production, and NO-mediated apoptosis. We hypothesized that monosialoganglioside GM1 inhibits NO synthase. The purpose of this study was to determine whether GM1 inhibits NO production and neuronal apoptosis after HCA. METHODS: Fourteen dogs underwent intracerebral microdialysis to measure excitatory amino acids, glutamate, aspartate, and citrulline, an equal coproduct of NO. They underwent 2 hours of HCA at 18 degrees C and were sacrificed 8 hours after HCA. Group 1 (n = 6) was pretreated with GM1, 30 mg/kg intravenously every day for 3 days, as well as before and after HCA. Group 2 control dogs (n = 8) received vehicle only. Apoptosis was scored from 0 (normal) to 100 (severe injury). RESULTS: Excitatory amino acids, aspartate and glutamate, coagonist glycine, and citrulline levels increased significantly over baseline during HCA and after HCA. GM1 pretreatment did not appreciably alter levels of glutamate, aspartate, and glycine; however, it substantially decreased citrulline and therefore NO production throughout the experiment. GM1 significantly inhibited apoptosis (group 1 vs group 2: 15.56 +/- 13.60 vs 62.92 +/- 6.17; P < .001). CONCLUSIONS: Our results provide the first direct evidence that GM1 inhibits NO synthase to reduce NO production and HCA-induced neuronal apoptosis. GM1 did not affect excitatory glutamate or aspartate levels. GM1 has been used in clinical trials of spinal cord injury and may be efficacious in reducing neurologic injury after HCA.


Assuntos
Ponte Cardiopulmonar , Giro Denteado/fisiopatologia , Gangliosídeo G(M1)/farmacologia , Parada Cardíaca/fisiopatologia , Hipotermia Induzida , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Giro Denteado/citologia , Giro Denteado/enzimologia , Cães , Parada Cardíaca/metabolismo , Injeções Intraventriculares , Masculino , Microdiálise , Microscopia Eletrônica , Neurônios/citologia , Neurônios/enzimologia , Neurônios/ultraestrutura , Fármacos Neuroprotetores/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo I
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...