Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Circulation ; 102(20): 2522-7, 2000 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-11076827

RESUMO

BACKGROUND: Interleukin-6 (IL-6) is a key molecule in chronic inflammation and has been implicated in the progression of atherosclerosis. Serotonin (5-hydroxytryptamine; 5-HT) causes vascular contraction and proliferation, but its role in atherogenesis has not been clarified. We investigated the effects of 5-HT on IL-6 synthesis in human vascular smooth muscle cells (VSMCs). METHODS AND RESULTS: IL-6 levels in the culture medium of VSMCs were determined by ELISA. IL-6 mRNA accumulation was determined by use of a Quantikine mRNA colorimetric quantification kit. NF-kappaB activation was tested by gel retardation assay. 5-HT induced IL-6 production by VSMCs in a time- and dose-dependent manner, with increased IL-6 mRNA accumulation and nuclear factor-kappaB activation. The effect of 5-HT on IL-6 production was significantly inhibited by the 5-HT(2) receptor antagonist ketanserin and the selective 5-HT(2A) receptor antagonist sarpogrelate. Conversely, the 5-HT(2) receptor agonist alpha-methyl-5-HT increased IL-6 production. The protein kinase C (PKC) inhibitor calphostin C, but not the protein kinase A inhibitor KT5720, suppressed 5-HT-induced IL-6 production. The effect of 5-HT was also abolished in PKC-depleted VSMCs after pretreatment with phorbol 12-myristate 13-acetate for 24 hours. CONCLUSIONS: 5-HT acts on 5-HT(2A) receptors and increases IL-6 synthesis in human VSMCs at least partially through a PKC-dependent pathway. These results suggested that 5-HT may contribute to inflammatory activation of the vessels during atherogenesis.


Assuntos
Interleucina-6/biossíntese , Músculo Liso Vascular/efeitos dos fármacos , Serotonina/farmacologia , Células Cultivadas , Meios de Cultivo Condicionados/química , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Relação Dose-Resposta a Droga , Humanos , Interleucina-6/análise , Interleucina-6/genética , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , NF-kappa B/biossíntese , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , RNA Mensageiro/biossíntese , Receptor 5-HT2A de Serotonina , Receptores de Serotonina/metabolismo , Antagonistas da Serotonina/farmacologia , Agonistas do Receptor de Serotonina/farmacologia , Acetato de Tetradecanoilforbol/farmacologia
2.
Cardiovasc Res ; 38(1): 192-7, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9683921

RESUMO

OBJECTIVE: The purpose of this study was to investigate the effects of vesnarinone on nitric oxide (NO) synthesis in cardiac myocytes. METHODS: We measured the accumulation of nitrite, a stable oxidation product of NO synthase (iNOS) protein in cultured neonatal rat cardiac myocytes. RESULTS: Incubation of the cultures with interleukin-1 beta (IL-1 beta; 10 ng/ml) and tumor necrosis factor alpha (TNF alpha; 10 ng/ml) caused a marked increase in nitrite production. Although vesnarinone by itself showed no effect on nitrite accumulation, it enhanced cytokine-induced nitrite production by cardiac myocytes in a dose-dependent manner. The effect of vesnarinone was completely abolished in the presence of NG-monomethyl-L-arginine or actinomycin D. The vesnarinone-induced nitrite production was accompanied by increased iNOS protein expression. In the presence of dibutyryl-cAMP, cytokine-induced nitrite accumulation was further increased, but the stimulatory effect on vesnarinone on nitrite accumulation was diminished. The effect of vesnarinone was also inhibited by Rp-8-Br-cAMPS, a competitive inhibitor of protein kinase A, in a dose-dependent manner. CONCLUSIONS: These findings indicate that vesnarinone increases NO synthesis in cytokine-stimulated cardiac myocytes, at least partially through a cAMP-dependent pathway.


Assuntos
Cardiotônicos/farmacologia , Miocárdio/metabolismo , Óxido Nítrico/biossíntese , Quinolinas/farmacologia , 8-Bromo Monofosfato de Adenosina Cíclica/análogos & derivados , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Análise de Variância , Animais , Animais Recém-Nascidos , Bucladesina/farmacologia , Células Cultivadas , Meios de Cultivo Condicionados/química , Proteína Quinase Tipo II Dependente de AMP Cíclico , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Dactinomicina/farmacologia , Relação Dose-Resposta a Droga , Interleucina-1/farmacologia , Óxido Nítrico Sintase/análise , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II , Nitritos/análise , Inibidores da Síntese de Proteínas/farmacologia , Pirazinas , Ratos , Ratos Sprague-Dawley , Tionucleotídeos/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , ômega-N-Metilarginina/farmacologia
3.
Cardiovasc Res ; 32(2): 422-9, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8796130

RESUMO

OBJECTIVE: The adhesive interaction of monocytes and endothelial cells has been implicated as a regulatory signal in the cell activation that is involved in the pathogenesis of atherosclerosis. We investigated the effect of monocyte-endothelial cell interaction on the expression of adhesion molecules, intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1), in human umbilical cord vein endothelial cells (HUVECs). METHODS: ICAM-1 and VCAM-1 protein and mRNA expression were determined by cellular ELISA and Northern blot analysis, respectively. RESULTS: The addition of unstimulated human monocytes, as well as interleukin-1 beta (IL-1 beta: 25 U/ml) and tumor necrosis factor-alpha (TNF: 100 U/ml), to HUVECs rapidly induced the expression of ICAM-1 and VCAM-1 protein and mRNA in HUVECs, whereas the addition of polymorphonuclear leukocytes (PMNs) had no significant effect on their expression. The induction of ICAM-1 and VCAM-1 by the co-culture of HUVECs and monocytes was significantly, but partially, inhibited by the combination of anti-IL-1 alpha, anti-IL-1 beta and anti-TNF Abs. Actinomycin D and genistein, but not calphostin C, also significantly inhibited the co-culture-induced adhesion molecule expression. CONCLUSIONS: These results suggest that the monocyte-endothelial cell interaction induces the expression of ICAM-1 and VCAM-1 in endothelial cells partially through the production of IL-1 and TNF. These findings also suggest that the monocyte-endothelial interaction further augments their interaction through the up-regulation of endothelial adhesion molecules, as a positive feedback mechanism.


Assuntos
Moléculas de Adesão Celular/metabolismo , Comunicação Celular/fisiologia , Endotélio Vascular/fisiologia , Monócitos/fisiologia , Arteriosclerose/metabolismo , Arteriosclerose/patologia , Northern Blotting , Moléculas de Adesão Celular/análise , Moléculas de Adesão Celular/genética , Células Cultivadas , Técnicas de Cocultura , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Molécula 1 de Adesão Intercelular/análise , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , RNA Mensageiro/análise , Cordão Umbilical/citologia , Molécula 1 de Adesão de Célula Vascular/análise , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo
4.
Cardiovasc Res ; 52(3): 438-45, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11738060

RESUMO

OBJECTIVE: Macrophage migration inhibitory factor (MIF), which plays a pivotal role in the control of inflammatory responses, was first characterized as a T-cell cytokine, but later was also found as a pituitary peptide released in response to infection and stress. However, MIF's role and expression in the myocardium has never been reported. The goal of this study is to examine MIF in the myocardium. METHODS AND RESULTS: MIF protein and mRNA levels were assayed using enzyme-linked immunosorbent assay (ELISA) and reverse transcription-polymerase chain reaction (RT-PCR), respectively. Increased MIF concentrations were detected in the sera of patients with acute myocardial infarction (AMI). In cultured rat cardiac myocytes, significant amounts of MIF were produced in response to hypoxia and hydrogen peroxide (H(2)O(2)), but not to angiotensin II, endothelin-1, interleukin-1beta (IL-1beta) or tumor necrosis factor alpha (TNFalpha). H(2)O(2)-induced MIF production increased in a time- and dose-dependent manner and was completely abolished in the presence of catalase. H(2)O(2) also induced MIF mRNA expression. The H(2)O(2)-induced MIF production was completely inhibited by the protein kinase C (PKC) inhibitor GF109203X, partially inhibited by the tyrosine kinase inhibitor herbimycin A, and uninhibited by calcium chelation or phorbol ester-sensitive PKC down-regulation. This suggests that H(2)O(2)-induced MIF production is mediated by an atypical PKC isoform. DNA microarray analysis revealed that 52 genes were preferentially expressed in response to MIF. Of these, the MIF-induced expression of both glutathione S-transferase (GST) and lipopolysaccharide-induced CXC chemokine (LIX) mRNAs was confirmed using RT-PCR analysis. CONCLUSION: The present results suggest that MIF is expressed by the myocardium in response to redox stress and may play a role in the pathogenesis of myocardial ischemia.


Assuntos
Fatores Inibidores da Migração de Macrófagos/fisiologia , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Análise de Variância , Animais , Benzoquinonas , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Feminino , Regulação da Expressão Gênica , Humanos , Peróxido de Hidrogênio/farmacologia , Indóis/farmacologia , Lactamas Macrocíclicas , Fatores Inibidores da Migração de Macrófagos/análise , Fatores Inibidores da Migração de Macrófagos/genética , Masculino , Maleimidas/farmacologia , Pessoa de Meia-Idade , Infarto do Miocárdio/sangue , Infarto do Miocárdio/genética , Análise de Sequência com Séries de Oligonucleotídeos , Oxirredução , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Quinonas/farmacologia , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rifabutina/análogos & derivados
5.
Hypertension ; 35(6): 1232-6, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10856269

RESUMO

Peroxisome proliferator-activated receptor-gamma (PPARgamma) is a key player in glucose metabolism. If PPARgamma ligands modulate nitric oxide (NO) synthesis in the vascular tissue, they may affect the process of plaque formation and postangioplasty restenosis. We investigated the effects of PPARgamma ligands on NO synthesis in vascular smooth muscle cells. Incubation of cultures with interleukin-1beta (10 ng/mL) for 24 hours caused a significant increase in the production of nitrite, a stable metabolite of NO, in cultured rat vascular smooth muscle cells. The PPARgamma agonists troglitazone and 15-deoxy-triangle up(12,14)-prostaglandin J(2) (15d-PG J(2)) dose-dependently inhibited nitrite production by interleukin-1beta-stimulated vascular smooth muscle cells. Decreased interleukin-1beta-induced nitrite production by the PPARgamma agonists was accompanied by decreased inducible NO synthase mRNA and protein accumulation. Interleukin-1beta induced nuclear factor-kappaB activation in vascular smooth muscle cells, and both troglitazone and 15d-PG J(2) markedly suppressed this nuclear factor-kappaB activation. PPARgamma ligands inhibit NO synthesis in cytokine-stimulated vascular smooth muscle cells, suggesting that these agonists may act directly on the vascular smooth muscle and influence the process of atherosclerosis and restenosis.


Assuntos
Músculo Liso Vascular/metabolismo , Óxido Nítrico/antagonistas & inibidores , Prostaglandina D2/análogos & derivados , Receptores Citoplasmáticos e Nucleares/metabolismo , Tiazolidinedionas , Fatores de Transcrição/metabolismo , Animais , Células Cultivadas , Cromanos/farmacologia , Humanos , Interleucina-1/farmacologia , Ligantes , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , NF-kappa B/fisiologia , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Nitritos/metabolismo , Prostaglandina D2/farmacologia , RNA Mensageiro/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Receptores Citoplasmáticos e Nucleares/agonistas , Proteínas Recombinantes/farmacologia , Tiazóis/farmacologia , Fatores de Transcrição/agonistas , Troglitazona
6.
Hypertension ; 29(1 Pt 1): 65-9, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9039082

RESUMO

We investigated the effects of endothelin-1 on nitric oxide synthesis in vascular smooth muscle cells. We measured the production of nitrite, a stable metabolite of nitric oxide, and the expression of inducible nitric oxide synthase mRNA and protein in cultured rat vascular smooth muscle cells. Incubation of the cultures with interleukin-1 beta (10 ng/mL) for 24 hours caused a significant increase in nitrite production. Endothelin-1 significantly decreased the interleukin-1 beta-induced nitrite production by vascular smooth muscle cells in a dose-dependent manner (10(-11) to 10(-8) mol/L). Incubation with interleukin-1 beta for 24 hours induced expression of inducible nitric oxide synthase mRNA and protein in vascular smooth muscle cells, whereas endothelin-1 showed a suppressive effect on their expressions. Addition of the endothelin type A receptor antagonist BQ-485, but not the endothelin type B receptor antagonist BQ-788, dose-dependently inhibited the effect of endothelin-1. After protein kinase C activity was functionally depleted by treatment of cells with phorbol 12-myristate 13-acetate for 24 hours, the effect of endothelin-1 was abolished. These results indicate that endothelin-1 acts on endothelin type A receptors and inhibits nitric oxide synthesis in interleukin-1 beta-stimulated vascular smooth muscle cells at least partially through a protein kinase C-dependent pathway.


Assuntos
Endotelina-1/farmacologia , Músculo Liso Vascular/metabolismo , Óxido Nítrico/biossíntese , Animais , Azepinas/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Regulação para Baixo , Antagonistas dos Receptores de Endotelina , Regulação Enzimológica da Expressão Gênica , Interleucina-1/farmacologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Oligopeptídeos/farmacologia , Piperidinas/farmacologia , Proteína Quinase C/metabolismo , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Receptores de Endotelina/metabolismo , Fatores de Tempo
7.
J Hypertens ; 18(11): 1597-604, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11081772

RESUMO

OBJECTIVE: If calcium channel blockers affect nitric oxide synthesis in the vascular tissue, they could influence disease progression in coronary arteries. We investigated the effects of the calcium channel blocker amlodipine on nitric oxide synthesis by measuring the production of nitrite, a stable metabolite of nitric oxide, in vascular smooth muscle cells. METHODS: We measured the production of nitrate in cultured rat vascular smooth muscle cells with the Griess reagent Inducible nitric oxide synthase protein and mRNA expression were assayed by Western blotting and reverse transcription-polymerase chain reaction, respectively. The levels of NF-kappaB proteins in nuclear extracts were analyzed by gel retardation assay. RESULTS: Incubation of cultures with interleukin-1 , (10 ng/ ml) for 24 h caused a significant increase in nitrite generation. Interleukin-1 l-induced nitrite production by vascular smooth muscle cells was significantly increased by amlodipine in a dose-dependent manner. This augmentative effect of amlodipine was completely abolished in the presence of N(G)-monomethyl-L-arginine or actinomycin D. Amlodipine-induced nitrite production was accompanied by increased inducible nitric oxide synthase mRNA and protein accumulation. Interleukin-1 , induced NF-kappaB activation in vascular smooth muscle cells, and addition of amlodipine further increased this NF-kappaB activation. The effect of amlodipine on nitrite production was maintained in the presence of the calcium channel agonist Bay K 8644. CONCLUSION: Amlodipine enhances nitric oxide synthesis in cytokine-stimulated cultured vascular smooth muscle cells by L-type calcium channel-independent mechanisms.


Assuntos
Anlodipino/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Interleucina-1/farmacologia , Músculo Liso Vascular/enzimologia , Óxido Nítrico Sintase/metabolismo , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/farmacologia , Animais , Aorta Torácica/citologia , Agonistas dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/metabolismo , Células Cultivadas , Diltiazem/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , NF-kappa B/metabolismo , Nifedipino/farmacologia , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Nitritos/metabolismo , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley
8.
Hypertens Res ; 23(2): 187-92, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10770267

RESUMO

Statins are lipid-lowering agents which act by inhibition of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase. This enzyme is responsible for the conversion of HMG-CoA to mevalonate. Products of mevalonate metabolism are critical for several cellular processes of eukaryotic cells, and inhibition of the mevalonate pathway by statins has pleiotropic effects. It has been reported that statins inhibit the migration and proliferation of vascular smooth cells (VSMCs) and macrophages, decrease interleukin-6 and inducible nitric oxide synthase expression in VSMCs, improve endothelial function and up-regulate endothelial nitric oxide synthase expression. The above effects of statins are independent of plasma cholesterol levels, and are completely blocked by exogenous mevalonate and some isoprenoids. These findings suggest that, in addition to their effects on plasma lipids, statins exert direct antiatherosclerotic effects on the cells primarily involved in atherosclerosis.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Macrófagos/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Animais , Arteriosclerose/fisiopatologia , Endotélio Vascular/citologia , Humanos , Macrófagos/fisiologia , Músculo Liso Vascular/citologia
9.
Eur J Pharmacol ; 338(1): 97-104, 1997 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-9408009

RESUMO

We investigated the effects of serotonin (5-hydroxytryptamine; 5-HT) on nitric oxide (NO) synthesis in vascular smooth muscle cells. We measured the production of nitrite, a stable metabolite of NO, and the expression of inducible NO synthase protein in cultured rat vascular smooth muscle cells. Incubation of the cultures with interleukin-1beta (10 ng/ml) caused a significant increase in nitrite production. 5-HT inhibited nitrite production by interleukin-1beta -stimulated vascular smooth muscle cells in a concentration-dependent manner (10(-8)-10(-5) M). 5-HT-induced inhibition of nitrite production was accompanied by decreased inducible NO synthase protein accumulation in vascular smooth muscle cells. Addition of the 5-HT2 receptor antagonist ketanserin, but not the 5-HT1A receptor antagonist spiroxatrine, inhibited the effect of 5-HT. On the other hand, the 5-HT2 receptor agonist alpha-methyl-5-HT, but not the 5-HT1A receptor agonist (+/-)-8-hydroxy-2-(di-n-propylamino) tetralin, decreased interleukin-1beta-induced nitrite production by vascular smooth muscle cells. 5-HT significantly increased protein kinase C activity in vascular smooth muscle cells, and the protein kinase C inhibitor calphostin C dose-dependently abolished the effect of 5-HT on nitrite production. After protein kinase C activity was functionally depleted by treatment of cells with phorbol 12-myristate 13-acetate for 24 h, the effect of 5-HT was abolished. These results indicate that 5-HT acts on 5-HT2 receptors and inhibits NO synthesis in interleukin-1beta-stimulated vascular smooth muscle cells at least partially through a protein kinase C-dependent pathway.


Assuntos
Interleucina-1/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Óxido Nítrico/biossíntese , Antagonistas da Serotonina/farmacologia , Serotonina/farmacologia , Animais , Células Cultivadas , Interações Medicamentosas , Músculo Liso Vascular/metabolismo , Proteína Quinase C/metabolismo , Ratos , Ratos Sprague-Dawley
11.
Am J Physiol ; 273(1 Pt 2): H59-65, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9249475

RESUMO

We investigated the effects of adenosine on nitric oxide (NO) synthesis by measuring the production of nitrite, a stable metabolite of NO, in cultured neonatal rat cardiac myocytes. Incubation of cultures with interleukin-1 beta (10 ng/ml) for 24 h caused a significant increase in nitrite production. The interleukin-1 beta-induced nitrite production by cardiac myocytes was significantly increased by adenosine or its stable analog 2-chloroadenosine in a dose-dependent manner (10(-7)-10(-4) M). The adenosine A2-receptor antagonist KF-17837 (10(-6) M), but not the A1-receptor antagonist 8-cyclopentyl-1, 3-dipropylxanthine (10(-6) M), significantly inhibited 2-chloroadenosine-mediated nitrite production. The 2-chloroadenosine-induced nitrite production by interleukin-1 beta-stimulated cells was accompanied by inducible NO synthase mRNA and protein accumulation. In the presence of N6, 2'-O-dibutyryladenosine 3',5'-cyclic monophosphate (cAMP) (10(-3) M) or isoproterenol (10(-5) M), interleukin-1 beta-induced nitrite accumulation was further increased, but the effect of 2-chloroadenosine was not additive or synergistic. The protein kinase C inhibitor calphostin C did not inhibit the effect of 2-chloroadenosine. These results indicate that adenosine acts on A2 receptors and augments NO synthesis in interleukin-1 beta-stimulated cardiac myocytes, at least partially through a cAMP-dependent pathway.


Assuntos
Adenosina/farmacologia , Miocárdio/metabolismo , Óxido Nítrico/biossíntese , ômega-N-Metilarginina/farmacologia , 2-Cloroadenosina/farmacologia , Animais , Animais Recém-Nascidos , Bucladesina/farmacologia , Células Cultivadas , Dactinomicina/farmacologia , Inibidores Enzimáticos/farmacologia , Coração/efeitos dos fármacos , Humanos , Interleucina-1/farmacologia , Isoproterenol/farmacologia , Miocárdio/citologia , Naftalenos/farmacologia , Nitritos/análise , Proteína Quinase C/antagonistas & inibidores , Antagonistas de Receptores Purinérgicos P1 , Antagonistas do Receptor Purinérgico P2 , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/farmacologia , Estereoisomerismo , Teofilina/análogos & derivados , Teofilina/farmacologia , Xantinas/farmacologia
12.
Hypertension ; 35(5): 1085-91, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10818069

RESUMO

The purpose of this study was to investigate the effects of aspirin-like drugs on nitric oxide (NO) synthesis in rat vascular smooth muscle cells (VSMCs). We measured the accumulation of nitrite, a stable oxidation product of NO, and the expression of inducible NO synthase (iNOS) mRNA and protein in rat cultured VSMCs. Sodium salicylate, aspirin, and indomethacin dose-dependently enhanced nitrite production by interleukin (IL)-1beta-stimulated VSMCs at therapeutic plasma concentration ranges. Increased nitrite production by aspirin-like drugs was accompanied by increased iNOS mRNA and protein accumulation in VSMCs. Addition of IL-1beta activated nuclear factor kappaB (NF-kappaB) in VSMCs, but sodium salicylate did not affect IL-1beta-induced NF-kappaB activation. The nonselective lipoxygenase (LO) inhibitor nordihydroguaiaretic acid inhibited sodium salicylate-induced nitrite production, whereas the selective 5-LO inhibitor caffeic acid did not influence production of nitrite. The 12-LO product 12-HETE dose-dependently enhanced nitrite production by IL-1beta-stimulated VSMCs, whereas the 15-LO product 15-HETE did not. Our study demonstrates that aspirin and the aspirin-like drugs, sodium salicylate and indomethacin, increase NO synthesis in IL-1beta-stimulated VSMCs by upregulation of iNOS transcription via a 12-LO pathway. These effects were independent of NF-kappaB activation. In addition to the direct inhibition of platelet function, aspirin-like drugs may contribute to the reduction of atherothrombotic risk in myocardial ischemia via enhancing NO production by VSMCs.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Aspirina/farmacologia , Indometacina/farmacologia , Músculo Liso Vascular/metabolismo , Óxido Nítrico/biossíntese , Salicilato de Sódio/farmacologia , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/farmacologia , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Ácidos Hidroxieicosatetraenoicos/farmacologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Ratos , Ratos Sprague-Dawley
13.
Hypertension ; 36(6): 923-8, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11116101

RESUMO

Nitric oxide (NO) production by inducible NO synthase (iNOS) may play an important role in the pathogenesis of atherosclerosis. Although fluvastatin has been shown to reduce progression of atherosclerosis, it is not known whether it regulates iNOS expression. We investigated the effects of fluvastatin on iNOS expression and subsequent NO synthesis in vascular smooth muscle cells (VSMCs) and the mechanism by which fluvastatin exerts its effects. Fluvastatin significantly increased interleukin-1ss (IL-1ss)-induced nitrite production by VSMCs in a time-dependent (0 to 24 hours) and dose-dependent (10(-)(8) to 10(-)(5) mol/L) manner. Increased nitrite production by fluvastatin was accompanied by increased iNOS mRNA and protein accumulation. IL-1ss induced nuclear factor-kappaB activation in VSMCs, which was not affected by fluvastatin. Exogenous mevalonate significantly prevented the stimulatory effect of fluvastatin on nitrite production. Cotreatment with geranylgeranyl-pyrophosphate also reversed the effect of fluvastatin. Furthermore, both Rho inhibitor C3 exoenzyme and Rho kinase inhibitor Y-27632 significantly increased IL-1ss-induced nitrite accumulation in VSMCs. These results demonstrated that fluvastatin upregulates iNOS expression and subsequent NO formation in rat VSMCs through inhibition of Rho.


Assuntos
Ácidos Graxos Monoinsaturados/farmacologia , Indóis/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Óxido Nítrico Sintase/biossíntese , Animais , Anticolesterolemiantes/farmacologia , Arteriosclerose/etiologia , Arteriosclerose/metabolismo , Células Cultivadas , Citocinas/metabolismo , Interações Medicamentosas , Fluvastatina , Expressão Gênica/efeitos dos fármacos , Ácido Mevalônico/farmacologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/enzimologia , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Regulação para Cima/efeitos dos fármacos , Proteínas rho de Ligação ao GTP/antagonistas & inibidores
14.
J Cardiovasc Pharmacol ; 36(4): 498-502, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11026652

RESUMO

Vascular endothelial growth factor (VEGF)-induced angiogenesis is involved in the etiology of some cardiovascular diseases. The soluble form of VEGF receptor, FLT-1 (sFLT-1), is a potent antagonist of VEGF. Therefore, we investigated whether transfection with the sFLT-1 gene could inhibit VEGF-induced angiogenesis. Human embryonic kidney (HEK)-293 cells were transfected with plasmids containing VEGF and sFLT-1 (pCMV-VEGF and pCMV-sFLT-1) by the calcium-phosphate co-precipitation method. VEGF- and/or sFLT-1-transfected HEK-293 cells were incubated for 24 h, and then conditioned medium was collected. The effects of conditioned medium on angiogenesis were tested by incorporation of [3H]thymidine into human umbilical vein endothelial cells (HUVECs). Expression of VEGF protein was determined by Western blotting. The conditioned medium from sFLT-1 gene-transfected HEK-293 cells significantly inhibited recombinant VEGF-induced increase in [3H]thymidine incorporation by HUVECs. VEGF gene-transfected HEK-293 cells secreted VEGF protein into conditioned medium. This conditioned medium increased [3H]thymidine incorporation by HUVECs, which was significantly inhibited by co-transfection of sFLT-1 gene with VEGF gene. These observations suggested that sFLT-1 gene transfer could inhibit VEGF-induced DNA synthesis of vascular endothelial cells.


Assuntos
Fatores de Crescimento Endotelial/fisiologia , Linfocinas/fisiologia , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Receptores de Fatores de Crescimento/genética , Transfecção , Western Blotting , Linhagem Celular , Meios de Cultivo Condicionados , DNA/biossíntese , DNA/genética , Fatores de Crescimento Endotelial/antagonistas & inibidores , Fatores de Crescimento Endotelial/genética , Humanos , Fígado/metabolismo , Linfocinas/antagonistas & inibidores , Linfocinas/genética , Plasmídeos/genética , Fator A de Crescimento do Endotélio Vascular , Receptor 1 de Fatores de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
15.
Hypertension ; 36(3): 325-9, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10988259

RESUMO

Matrix metalloproteinase-1 (MMP-1), also called interstitial collagenase, may play an important role in the pathogenesis of atherosclerosis and atherosclerotic plaque rupture. We investigated the effects of fluvastatin on MMP-1 expression in human vascular endothelial cells (ECs). The addition of fluvastatin decreased the basal MMP-1 levels in the culture media of ECs in a time-dependent (0 to 48 hours) and dose-dependent (10(-)(8) to 10(-)(5) mol/L) manner. On the other hand, fluvastatin did not affect tissue inhibitor of metalloproteinase-1 levels. Collagenolytic activity in conditioned media of ECs was also dose-dependently reduced by fluvastatin. The effect of fluvastatin on MMP-1 expression was completely reversed in the presence of mevalonate or geranylgeranyl-pyrophosphate, but not in the presence of squalene. Inhibition of Rho by C3 exoenzyme also significantly decreased MMP-1 expression in ECs. Our findings revealed that fluvastatin decreases MMP-1 expression in human vascular ECs through inhibition of Rho.


Assuntos
Anticolesterolemiantes/farmacologia , Endotélio Vascular/efeitos dos fármacos , Ácidos Graxos Monoinsaturados/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Indóis/farmacologia , Metaloproteinase 1 da Matriz/efeitos dos fármacos , Análise de Variância , Relação Dose-Resposta a Droga , Endotélio Vascular/metabolismo , Fluvastatina , Humanos , Lovastatina/farmacologia , Metaloproteinase 1 da Matriz/metabolismo , Ácido Mevalônico/farmacologia , Fosfatos de Poli-Isoprenil/farmacologia , Pravastatina/farmacologia , Sesquiterpenos , Esqualeno/farmacologia , Fatores de Tempo , Inibidor Tecidual de Metaloproteinase-1/metabolismo
16.
J Cardiovasc Pharmacol ; 38(1): 69-77, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11444504

RESUMO

Nitric oxide production by inducible nitric oxide synthase (iNOS) may play an important role in the pathogenesis of cardiovascular dysfunction. We investigated the effects of statins on iNOS expression and subsequent nitric oxide synthesis in cardiac myocytes and the mechanism by which statins exert their effects. We measured the production of nitrite, a stable metabolite of nitric oxide, in cultured neonatal rat cardiac myocytes with the Griess reagent. iNOS mRNA and protein expression were assayed by reverse transcription polymerase chain reaction and Western blotting, respectively. The lipophilic statins fluvastatin and lovastatin significantly increased interleukin-1beta-induced nitrite production by cardiac myocytes, whereas hydrophilic pravastatin did not. Increased nitrite production by fluvastatin was accompanied by increased iNOS mRNA and protein accumulation. Exogenous mevalonate, but not squalene, significantly blocked the stimulatory effect of fluvastatin on nitrite production. Cotreatment with geranylgeranyl-pyrophosphate also reversed the effect of fluvastatin. Furthermore, both Rho inhibitor C3 exoenzyme and Rho kinase inhibitor Y-27632 significantly increased interleukin-1beta-induced nitrite accumulation in cardiac myocytes. These results demonstrated that lipophilic statins upregulate iNOS expression and subsequent nitric oxide formation in cardiac myocytes via inhibition of Rho.


Assuntos
Anticolesterolemiantes/farmacologia , Citocinas/farmacologia , Ácidos Graxos Monoinsaturados/farmacologia , Indóis/farmacologia , Lovastatina/farmacologia , Miocárdio/enzimologia , Óxido Nítrico Sintase/biossíntese , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Fluvastatina , Miocárdio/citologia , Óxido Nítrico Sintase Tipo II , Nitritos/metabolismo , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley
17.
Exp Clin Cardiol ; 6(1): 50-5, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-20428445

RESUMO

BACKGROUND: Adeno-associated virus (AAV) has a number of attractive features for gene therapy including the ability to transduce nondividing cells and long term transgene expression. OBJECTIVE: To investigate whether the endothelial constitutive nitric oxide synthase (ecNOS) gene can be efficiently introduced into rat aortic segments using AAV vectors and thereby modulate the vasoconstrictive response. ANIMALS AND METHODS: Excised rat aortas were incubated with medium containing ecNOS-expressing AAV vectors (AAV-ecNOS). Expression of ecNOS in the aortic segments was evaluated by immunohistochemical staining. The isometric tension of the aortic segments transduced with AAV-ecNOS was measured. RESULTS: Adventitial cells in rat aortic segments were efficiently transduced with AAV-ecNOS. The vasoconstrictive response induced by 30 mmol/L K(+) was enhanced in endothelium-denuded aortic segments compared with intact aortic segments. However, in endothelium-denuded aortic segments transduced with AAV-ecNOS, the enhancement of the vasoconstrictive response disappeared. This effect induced by ecNOS gene transfer was abolished in the presence of the nitric oxide synthase inhibitor N(G)-monomethyl-l-arginine acetate. CONCLUSIONS: These results show that ecNOS gene transfer using AAV vectors abolishes the pathological enhancement of the vasoconstrictive response in endothelium-denuded aortic segments.

18.
J Cardiovasc Pharmacol ; 36(4): 438-43, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11026643

RESUMO

Vascular endothelial growth factor (VEGF) is an angiogenic growth factor that stimulates endothelial cell proliferation, increases endothelial permeability, and promotes collateral vessel formation. We transferred human VEGF gene into rat cardiac myocytes using adeno-associated virus (AAV) vectors and investigated whether VEGF secreted from the transduced cardiac myocytes promoted proliferation of endothelial cells. We produced VEGF-expressing AAV vectors (AAV-VEGF) by the adenovirus-free method. Immunoblotting revealed VEGF protein expression in AAV-VEGF-transduced rat cardiac myocytes. More than 60% of cardiac myocytes were stained positively on immunohistochemical staining using anti-VEGF antibody. Concentration of VEGF in the culture medium of AAV-VEGF-transduced myocytes was increased in a vector dose-dependent manner, and VEGF secretion from the transduced myocytes persisted for > or = 14 days. Thymidine incorporation into human vascular endothelial cells was significantly increased by incubation with the conditioned medium from AAV-VEGF-transduced myocytes. This increased thymidine uptake was significantly inhibited by anti-VEGF antibody. We demonstrated here that AAV-mediated VEGF gene transfer into cardiac myocytes induces the secretion of functional VEGF.


Assuntos
Dependovirus/genética , Fatores de Crescimento Endotelial/genética , Vetores Genéticos/genética , Linfocinas/genética , Miocárdio/citologia , Animais , Western Blotting , Divisão Celular/genética , Células Cultivadas , Meios de Cultura , Endotélio Vascular/citologia , Imuno-Histoquímica , Ratos , Ratos Sprague-Dawley , Timidina/metabolismo , Transdução Genética , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
19.
Am J Nephrol ; 20(3): 242-7, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10878409

RESUMO

Adeno-associated virus (AAV) vectors have a number of attractive features, including lack of cytotoxicity, ability to transduce nondividing cells, and long-term transgene expression. We investigated whether rat renal cells could be efficiently transduced with AAV vectors. Rat glomerular mesangial cells were transduced with AAV-lacZ vector containing beta-galactosidase gene in vitro, and the expression of beta-galactosidase was evaluated by X-gal staining and ELISA. For ex vivo experiments, sections of rat kidneys were incubated with AAV-lacZ, and then evaluated by X-gal histochemical staining. The level of beta-galactosidase expression in cultured rat mesangial cells increased in a dose-dependent manner (ranging from 1 x 10(5) to 5 x 10(6) particles/cell). When transduced with 5 x 10(6) vector particles/cell of AAV-lacZ, about 50% of mesangial cells were stained positively with X-gal, and the level of beta-galactosidase expression reached 9.9 +/- 1.5 ng/mg protein. Expression was detectable during the culture period for at least 7 days. X-gal histochemical examination of the ex vivo transduced renal tissue revealed tubular cell and interstitial tissue staining. However, gene transfer was not clearly observed in glomeruli. These findings suggest that AAV vectors have the potential for gene therapy of renal diseases.


Assuntos
Dependovirus , Técnicas de Transferência de Genes , Vetores Genéticos , Mesângio Glomerular/citologia , Animais , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Masculino , Ratos , Ratos Sprague-Dawley , Transdução Genética , beta-Galactosidase/genética
20.
J Pharmacol Exp Ther ; 292(1): 387-93, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10604975

RESUMO

We investigated whether endothelial nitrite oxide synthase (NOS) gene transfer inhibited cellular proliferation. Endothelial NOS and endothelin type A receptor genes were transferred into 293 cells, a human embryonic kidney cell line, by calcium-phosphate coprecipitation. The cytosolic free Ca(2+) levels ([Ca(2+)](i)) of transfected cells were estimated with fura-2 fluorescence. Thymidine incorporation was increased by endothelin-1 in type A receptor-transfected cells. The endothelial NOS gene transfer did not affect endothelin-1-induced increase in [Ca(2+)](i) of type A receptor-transfected cells, but markedly inhibited mitogen-activated protein kinase and c-fos promoter activities. The endothelial NOS gene transfer also inhibited thymidine incorporation into type A receptor-transfected cells in response to endothelin-1, which was abolished in the presence of the NOS inhibitor N(G)-monomethyl-L-arginine acetate. The endothelin-1-induced increase in cell number was significantly suppressed by endothelial NOS gene transfer as well as by the mitogen-activated protein kinase inhibitor PD98059. These results indicate that endothelial NOS gene transfer inhibits cellular proliferation via inhibition of the mitogen-activated protein kinase cascade.


Assuntos
Divisão Celular/fisiologia , Endotélio/fisiologia , Óxido Nítrico Sintase/genética , Óxido Nítrico/fisiologia , Receptores de Endotelina/genética , Cálcio/metabolismo , Células Cultivadas , Embrião de Mamíferos , Endotelina-1/farmacologia , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Fura-2 , Humanos , Técnicas In Vitro , Rim/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/biossíntese , Fosfatos/química , Proteínas Proto-Oncogênicas c-fos/metabolismo , Timidina/farmacocinética , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA