Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
2.
Pharmacol Rev ; 67(1): 214-58, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25535277

RESUMO

Urotensin II (UII) is a cyclic neuropeptide that was first isolated from the urophysis of teleost fish on the basis of its ability to contract the hindgut. Subsequently, UII was characterized in tetrapods including humans. Phylogenetic studies and synteny analysis indicate that UII and its paralogous peptide urotensin II-related peptide (URP) belong to the somatostatin/cortistatin superfamily. In mammals, the UII and URP genes are primarily expressed in cholinergic neurons of the brainstem and spinal cord. UII and URP mRNAs are also present in various organs notably in the cardiovascular, renal, and endocrine systems. UII and URP activate a common G protein-coupled receptor, called UT, that exhibits relatively high sequence identity with somatostatin, opioid, and galanin receptors. The UT gene is widely expressed in the central nervous system (CNS) and in peripheral tissues including the retina, heart, vascular bed, lung, kidney, adrenal medulla, and skeletal muscle. Structure-activity relationship studies and NMR conformational analysis have led to the rational design of a number of peptidic and nonpeptidic UT agonists and antagonists. Consistent with the wide distribution of UT, UII has now been shown to exert a large array of biologic activities, in particular in the CNS, the cardiovascular system, and the kidney. Here, we review the current knowledge concerning the pleiotropic actions of UII and discusses the possible use of antagonists for future therapeutic applications.


Assuntos
Hormônios Peptídicos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Urotensinas/metabolismo , Sequência de Aminoácidos , Animais , Antagonistas de Hormônios/farmacologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Ligantes , Dados de Sequência Molecular , Hormônios Peptídicos/antagonistas & inibidores , Hormônios Peptídicos/química , Hormônios Peptídicos/genética , Conformação Proteica , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais , Relação Estrutura-Atividade , Urotensinas/antagonistas & inibidores , Urotensinas/química , Urotensinas/genética
3.
Br J Pharmacol ; 180(22): 2839-2845, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37846458

RESUMO

In a physiological context, the extracellular matrix (ECM) provides an important scaffold for organs. Dysregulation of ECM in disease conditions, characterised by excess deposition of connective tissue and extracellular matrix in response to a pathological insult, is a key driver of disease progression in multiple organs. The resultant fibrosis is predominantly an irreversible process and directly contributes to, and exacerbates, dysfunction of an affected organ. This is particularly paramount in the kidney, liver, heart and lung. A hybrid Joint Meeting of NC-IUPHAR and British Pharmacological Society was held in Paris and via a webinar in November 2020, when two successive sessions were devoted to translational advances in fibrosis as a therapeutic target. On the upsurge of response to these sessions, the concept of a special themed issue on this topic emerged, and is entitled Translational Advances in Fibrosis as a Therapeutic Target. In this special issue, we seek to provide an up-to-date account of the diverse molecular mechanisms and causal role that fibrosis plays in disease progression (contributing to, and exacerbating, dysfunction of affected organs). Recent developments in the understanding of molecular targets involved in fibrosis, and how their actions can be manipulated therapeutically, are included. LINKED ARTICLES: This article is part of a themed issue on Translational Advances in Fibrosis as a Therapeutic Target. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.22/issuetoc.


Assuntos
Matriz Extracelular , Coração , Humanos , Fibrose , Progressão da Doença
4.
Circ Res ; 105(7): 686-95, 19 p following 695, 2009 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-19696412

RESUMO

RATIONALE: Expression of the vasoactive peptide Urotensin II (UII) is elevated in a number of cardiovascular diseases. OBJECTIVE: Here, we sought to determine the effect of UII receptor (UT) gene deletion in a mouse model of atherosclerosis. METHODS AND RESULTS: UT knockout (KO) mice were crossed with ApoE KO mice to generate UT/ApoE double knockout (DKO) mice. Mice were placed on a high-fat Western-type diet for 12 weeks. We evaluated the degree of atherosclerosis and hepatic steatosis by histology. In addition, serum glucose, insulin, and lipids were determined. DKO mice exhibited significantly increased atherosclerosis compared to ApoE KO mice (P<0.05). This was associated with a significant increase in serum insulin and lipids (P<0.001) but a decrease in hepatic steatosis (P<0.001). UT gene deletion led to a significant increase in systolic pressure and pulse pressure. RT-PCR and immunoblot analyses showed significant reductions in hepatic scavenger receptors, nuclear receptors, and acyl-CoA:cholesterol acyltransferase (ACAT1) expression in DKO mice. UII induced a significant increase in intracellular cholesteryl ester formation in primary mouse hepatocytes, which was blocked by the MEK inhibitor, PD98059. Hepatocytes of UTKO mice showed a significant reduction in lipoprotein uptake compared to wild-type mice. CONCLUSIONS: We propose that UT gene deletion in an ApoE-deficient background promotes downregulation of ACAT1, which in turn attenuates hepatic lipoprotein receptor-mediated uptake and lipid transporter expression. As the liver is the main organ for uptake of lipoprotein-derived lipids, DKO leads to an increase in hyperlipidemia, with a concomitant decrease in hepatic steatosis, and consequently increased atherosclerotic lesion formation. Furthermore, the hypertension associated with UT gene deletion is likely to contribute to the increased atherosclerotic burden.


Assuntos
Aorta/metabolismo , Apolipoproteínas E/deficiência , Aterosclerose/metabolismo , Hiperlipidemias/metabolismo , Fígado/metabolismo , Receptores Acoplados a Proteínas G/deficiência , Urotensinas/metabolismo , Acetil-CoA C-Acetiltransferase/metabolismo , Animais , Aorta/efeitos dos fármacos , Aorta/patologia , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Glicemia/metabolismo , Pressão Sanguínea , Células Cultivadas , Ésteres do Colesterol/metabolismo , Gorduras na Dieta/administração & dosagem , Modelos Animais de Doenças , Fígado Gorduroso/metabolismo , Fígado Gorduroso/prevenção & controle , Genótipo , Hiperlipidemias/genética , Hiperlipidemias/patologia , Hiperlipidemias/fisiopatologia , Insulina/sangue , Lipídeos/sangue , Fígado/efeitos dos fármacos , Fígado/patologia , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fenótipo , Inibidores de Proteínas Quinases/farmacologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Depuradores/metabolismo , Fatores de Tempo
5.
Nucleic Acids Res ; 37(Database issue): D680-5, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18948278

RESUMO

The IUPHAR database (IUPHAR-DB) integrates peer-reviewed pharmacological, chemical, genetic, functional and anatomical information on the 354 nonsensory G protein-coupled receptors (GPCRs), 71 ligand-gated ion channel subunits and 141 voltage-gated-like ion channel subunits encoded by the human, rat and mouse genomes. These genes represent the targets of approximately one-third of currently approved drugs and are a major focus of drug discovery and development programs in the pharmaceutical industry. IUPHAR-DB provides a comprehensive description of the genes and their functions, with information on protein structure and interactions, ligands, expression patterns, signaling mechanisms, functional assays and biologically important receptor variants (e.g. single nucleotide polymorphisms and splice variants). In addition, the phenotypes resulting from altered gene expression (e.g. in genetically altered animals or in human genetic disorders) are described. The content of the database is peer reviewed by members of the International Union of Basic and Clinical Pharmacology Committee on Receptor Nomenclature and Drug Classification (NC-IUPHAR); the data are provided through manual curation of the primary literature by a network of over 60 subcommittees of NC-IUPHAR. Links to other bioinformatics resources, such as NCBI, Uniprot, HGNC and the rat and mouse genome databases are provided. IUPHAR-DB is freely available at http://www.iuphar-db.org.


Assuntos
Bases de Dados de Proteínas , Canais Iônicos/genética , Canais Iônicos/fisiologia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiologia , Animais , Descoberta de Drogas , Humanos , Canais Iônicos/química , Ligantes , Camundongos , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/fisiologia , Ratos , Receptores Acoplados a Proteínas G/química
6.
J Pharmacol Exp Ther ; 325(2): 466-74, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18287212

RESUMO

Peroxisome proliferator-activated receptor (PPAR)-delta is a transcription factor that belongs to the PPAR family. PPAR-delta is abundantly expressed in the heart, and its role in the heart is largely unknown. We tested whether pharmacological activation of PPAR-delta protects the heart from ischemia/reperfusion (I/R) injury in male Zucker fatty rats, a rodent model of obesity and dyslipidemia. A highly selective PPAR-delta agonist, [4-[[[2-[3-fluoro-4-(trifluoromethyl)phenyl]-4-methyl-5-thiazolyl]methyl] thio]-2-methylphenoxy]acetic acid (GW0742), was administered for 7 days at 10 mg/kg/day (p.o., once a day). Ischemic injury was produced by occlusion of the left anterior descending artery for 30 min followed by reperfusion for up to 24 h. Treatment with GW0742 reduced serum levels of cardiac troponin-I and infarct size by 63% (p < 0.01) and 32% (p < 0.01), respectively, and improved left ventricular function. Treatment with GW0742 up-regulated gene expression involved in cardiac fatty acid oxidation, increased fat use in the heart, and reduced serum levels of free fatty acids. The enhanced cardiac expression of interleukin (IL)-6, IL-8, intercellular adhesion molecule-1, and monocyte chemoattractant protein-1 induced by I/R were significantly attenuated by GW0742. Treatment with GW0742 also reduced apoptotic cardiomyocytes by 34% and cardiac caspase-3 activity by 61% (both p < 0.01 versus vehicle). GW0742 differentially regulated Bcl family members, favoring cell survival, and attenuated I/R-induced cardiac mitochondrial damage. In addition, GW0742 treatment augmented the cardiac Akt signaling pathway, as reflected by enhanced phospho-3-phosphoinositide-dependent kinase-1 and p-Akt. The results indicate that activation of PPAR-delta protected the heart from I/R injury in Zucker fatty rats, and multiple mechanisms including amelioration of lipotoxicity, anti-inflammation, and up-regulation of prosurvival signaling contribute together to the cardioprotection.


Assuntos
Cardiotônicos/uso terapêutico , Traumatismo por Reperfusão Miocárdica/prevenção & controle , PPAR delta/agonistas , Tiazóis/uso terapêutico , Animais , Pressão Sanguínea/efeitos dos fármacos , Citocinas/genética , Modelos Animais de Doenças , Ácidos Graxos/sangue , Ácidos Graxos/metabolismo , Coração/fisiopatologia , Cetonas/metabolismo , Masculino , Síndrome Metabólica/sangue , Síndrome Metabólica/fisiopatologia , Traumatismo por Reperfusão Miocárdica/sangue , Traumatismo por Reperfusão Miocárdica/fisiopatologia , PPAR delta/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Zucker , Troponina I/sangue
7.
Diabetes ; 54(2): 554-62, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15677515

RESUMO

The mechanism responsible for the enhanced myocardial susceptibility to ischemic insult in patients with type 2 diabetes is not clear. The present study examines the effect of rosiglitazone treatment on cardiac insulin sensitization and its association with cardioprotection from ischemia/reperfusion injury in an animal model of diabetes. Male Zucker diabetic fatty (ZDF) rats were treated with rosiglitazone (3 mg . kg(-1) . day(-1) orally) or vehicle for 8 days before undergoing 30 min of coronary artery ligation, followed by reperfusion for 4 h (apoptosis) or 24 h (infarction). Rosiglitazone reduced the blood levels of glucose, triglycerides, and free fatty acids; enhanced cardiac glucose oxidation; and increased Akt phosphorylation (Akt-pS473) 2.1-fold and Akt kinase activity 1.8-fold in the ischemic myocardium. The phosphorylation of two downstream targets of Akt, glycogen synthase kinase-3beta and FKHR (forkhead transcription factor), was also enhanced by 2- and 2.9-fold, respectively. In rosiglitazone-treated rats, the number of apoptotic cardiomyocytes and the myocardial infarct size were decreased by 58 and 46%, respectively, and the myocardial contractile dysfunction was improved. Blockade of the insulin-Akt signaling pathway by wortmannin in the 8-day rosiglitazone-treated ZDF rats resulted in a markedly diminished cardioprotective effect of rosiglitazone. In addition, 8-day rosiglitazone treatment in Zucker lean rats or 2-day rosiglitazone treatment in ZDF rats, both of which showed no change in whole-body insulin sensitivity, resulted in a significant reduction in cardiac infarct size, but to a lesser degree when compared with that observed in 8-day rosiglitazone-treated ZDF rats. These results suggest that chronic treatment with rosiglitazone protects the heart against ischemia/reperfusion injury in ZDF rats, and that the enhanced cardiac protection observed after rosiglitazone treatment might be attributable in part to an improvement in cardiac insulin sensitivity.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Coração/fisiopatologia , Resistência à Insulina/fisiologia , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Tiazolidinedionas/uso terapêutico , Administração Oral , Animais , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Vasos Coronários/fisiopatologia , Ácidos Graxos não Esterificados/sangue , Coração/efeitos dos fármacos , Hipoglicemiantes/uso terapêutico , Masculino , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Ratos , Ratos Zucker , Rosiglitazona , Triglicerídeos/sangue
8.
Peptides ; 27(11): 2919-26, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16919371

RESUMO

It is now well established that urotensin-II (UII) levels are increased in several cardiovascular diseases. We previously demonstrated that UII and the UII receptor (UT) protein levels are significantly increased in the hearts of both humans and rats with congestive heart failure (CHF). We have also recently demonstrated that UII blockade, with a selective UII antagonist, improves heart function in a rat model of ischemic CHF. Here, we evaluated the attenuation of cardiac remodeling associated with UII antagonism in the same rat model of ischemic CHF. Animals were administered a specific UT receptor antagonist, SB-611812 (30 mg/kg/day, gavage), or vehicle 30 min prior to coronary artery ligation followed by daily treatment for 8 weeks. Myocardial interstitial fibrosis was analyzed by Masson's trichrome and picrosirius red staining. RT-PCR analysis was utilized for mRNA expression studies. We used Western blotting to assess levels of collagen types I and III. Mitogenic activity of UII on cultured neonatal cardiac fibroblasts was also evaluated. Following coronary ligation, SB-611812 significantly attenuated both myocardial and endocardial interstitial fibrosis, and reduced collagen type I:III ratio (P<0.01). UII induced proliferation of cardiac fibroblasts and this mitogenic effect was significantly inhibited with 1 microM of SB-611218 (P<0.05). We demonstrate here that selective blockade of UT reduces diastolic dysfunction by decreasing myocardial fibrosis post-coronary ligation in vivo, and inhibits UII-mediated fibroblast proliferation in vitro.


Assuntos
Isquemia Miocárdica/tratamento farmacológico , Isquemia Miocárdica/fisiopatologia , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Sulfonamidas/farmacologia , Remodelação Ventricular/efeitos dos fármacos , Animais , Células Cultivadas , Modelos Animais de Doenças , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibrose/patologia , Fibrose/prevenção & controle , Perfilação da Expressão Gênica , Masculino , Isquemia Miocárdica/patologia , Miocárdio/patologia , Ratos , Ratos Endogâmicos Lew , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Urotensinas/farmacologia
9.
Methods Mol Biol ; 316: 1-11, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16671397

RESUMO

Gene identification followed by determination of the expression of genes in a given disease and understanding of the function of the gene products is central to the drug discovery process. The ability to associate a specific gene with a disease can be attributed primarily to the extraordinary progress that has been made in the areas of gene sequencing and information technologies. Selection and validation of novel molecular targets have become of great importance in light of the abundance of new potential therapeutic drug targets that have emerged from human gene sequencing. In response to this revolution within the pharmaceutical industry, the development of high-throughput methods in both biology and chemistry has been necessitated. Further, the successful translation of basic scientific discoveries into clinical experimental medicine and novel therapeutics is an increasing challenge. As such, a new paradigm for drug discovery has emerged. This process involves the integration of clinical, genetic, genomic, and molecular phenotype data partnered with cheminformatics. Central to this process, the data generated are managed, collated, and interpreted with the use of informatics. This review addresses the use of new technologies that have arisen to deal with this new paradigm.


Assuntos
Desenho de Fármacos , Genoma Humano , Biotecnologia , Indústria Farmacêutica , Genômica , Humanos , Farmacogenética , Proteômica
10.
Circulation ; 105(12): 1497-502, 2002 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-11914261

RESUMO

BACKGROUND: Recent evidence from cultured endothelial cell studies suggests that phosphorylation of endothelial nitric oxide synthase (eNOS) through the PI3-kinase-Akt pathway increases NO production. This study was designed to elucidate the signaling pathway involved in the antiapoptotic effect of insulin in vivo and to test the hypothesis that phosphorylation of eNOS by insulin may participate in the cardioprotective effect of insulin after myocardial ischemia and reperfusion. METHODS AND RESULTS: Male Sprague-Dawley rats were subjected to 30 minutes of myocardial ischemia and 4 hours of reperfusion. Rats were randomized to receive vehicle, insulin, insulin plus wortmannin, or insulin plus L-NAME. Treatment with insulin resulted in 2.6-fold and 4.3-fold increases in Akt and eNOS phosphorylation and a significant increase in NO production in ischemic/reperfused myocardial tissue. Phosphorylation of Akt and eNOS and increase of NO production by insulin were completely blocked by wortmannin, a PI3-kinase inhibitor. Pretreatment with L-NAME, a nonselective NOS inhibitor, had no effect on Akt and eNOS phosphorylation but significantly reduced NO production. Moreover, treatment with insulin markedly reduced myocardial apoptotic death (P<0.01 versus vehicle). Pretreatment with wortmannin abolished the antiapoptotic effect of insulin. Most importantly, pretreatment with L-NAME also significantly reduced the antiapoptotic effect of insulin (P<0.01 versus insulin). CONCLUSIONS: These results demonstrated that in vivo administration of insulin activated Akt through the PI3-kinase-dependent mechanism and reduced postischemic myocardial apoptotic death. Phosphorylation of eNOS and the concurrent increase of NO production contribute significantly to the antiapoptotic effect of insulin.


Assuntos
Apoptose , Insulina/farmacologia , Isquemia Miocárdica/metabolismo , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico/metabolismo , Proteínas Serina-Treonina Quinases , Traumatismo por Reperfusão/metabolismo , Androstadienos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Glucose/farmacologia , Masculino , Isquemia Miocárdica/tratamento farmacológico , Isquemia Miocárdica/patologia , Miocárdio/citologia , Miocárdio/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Potássio/farmacologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/prevenção & controle , Transdução de Sinais/efeitos dos fármacos , Wortmanina
11.
Circulation ; 105(20): 2429-34, 2002 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-12021232

RESUMO

BACKGROUND: Most clinical studies have shown that beta-adrenergic receptor antagonists improve long-term survival in heart failure patients. Bucindolol, a nonselective beta-receptor blocker, however, failed to reduce heart failure mortality in a recent large clinical trial. The reasons for this failure are not known. Bucindolol has partial agonist properties in rat myocardium, but whether it has agonist activity in human heart is controversial. To address this, we measured the ability of bucindolol to increase cAMP accumulation in human myocardium. METHODS AND RESULTS: Myocardial strips ( approximately 1 mm(3)) obtained from rat and nonfailing human hearts were confirmed to be viable for > or = 48 hours in normoxic tissue culture by MTT assay and histology. Freshly isolated strips were exposed to beta-adrenergic antagonists and agonists and assayed for cAMP. In both rat and human strips, the full beta-adrenergic agonist isoproterenol raised cAMP levels by >2.5-fold at 15 minutes. Carvedilol and propranolol had no effect on basal cAMP levels, whereas metoprolol reduced basal cAMP by approximately 25%. In contrast, bucindolol and xamoterol increased cAMP levels in a concentration-dependent manner in both rat and human myocardium (maximum 1.64+/-0.25-fold and 2.00+/-0.27-fold over control, respectively, P<0.01 for human tissue). CONCLUSIONS: Bucindolol exhibits approximately 60% of the beta-adrenergic agonist activity of xamoterol in normal human myocardial tissue.


Assuntos
Agonistas Adrenérgicos beta/farmacologia , Antagonistas Adrenérgicos beta/farmacologia , Coração/efeitos dos fármacos , Propanolaminas/farmacologia , Simpatomiméticos/farmacologia , Adulto , Idoso , Animais , Sobrevivência Celular , Colforsina/farmacologia , Técnicas de Cultura , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Feminino , Coração/fisiologia , Humanos , Líquido Intracelular/metabolismo , Masculino , Pessoa de Meia-Idade , Miocárdio/citologia , Miocárdio/metabolismo , Ratos , Xamoterol/farmacologia
12.
Circulation ; 108(22): 2805-11, 2003 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-14610009

RESUMO

BACKGROUND: Peroxisome proliferator-activated receptor (PPAR) signaling pathways have been reported to exert anti-inflammatory effects and attenuate atherosclerosis formation. However, the mechanisms responsible for their anti-inflammatory and antiatherosclerotic effects remain largely unknown. The present study tested the hypothesis that a PPARgamma agonist may exert significant endothelial protection by antioxidative and antinitrative effects. METHODS AND RESULTS: Male New Zealand White rabbits were randomized to receive a normal (control) or a high-cholesterol diet and treated with vehicle or rosiglitazone (a PPARgamma agonist) 3 mg x kg(-1) x d(-1) for 5 weeks beginning 3 weeks after the high-cholesterol diet. At the end of 8 weeks of a high-cholesterol diet, the rabbits were killed, and the carotid arteries were isolated. Bioactive nitric oxide was determined functionally (endothelium-dependent vasodilatation) and biochemically (the phosphorylation of vasodilator-stimulated phosphoprotein, or P-VASP). Vascular superoxide production, PPARgamma, gp91phox, and inducible nitric oxide synthase (iNOS) expression, and vascular ONOO- formation were determined. Hypercholesterolemia caused severe endothelial dysfunction and reduced P-VASP, despite a marked increase in iNOS expression and total NOx production. Treatment with rosiglitazone enhanced PPARgamma expression, improved endothelium-dependent vasodilatation, preserved P-VASP, suppressed gp91phox and iNOS expression, reduced superoxide and total NOx production, and inhibited nitrotyrosine formation. CONCLUSIONS: The PPARgamma agonist rosiglitazone exerted a significant vascular protective effect in hypercholesterolemic rabbits, most likely by attenuation of oxidative and nitrative stresses. The endothelial protective effects of PPARgamma agonists may reduce leukocyte accumulation in vascular walls and contribute to their antiatherosclerotic effect.


Assuntos
Antioxidantes/farmacologia , Vasos Sanguíneos/efeitos dos fármacos , Hipercolesterolemia/tratamento farmacológico , Nitratos/metabolismo , Receptores Citoplasmáticos e Nucleares/agonistas , Tiazolidinedionas/farmacologia , Fatores de Transcrição/agonistas , Tirosina/análogos & derivados , Animais , Antioxidantes/uso terapêutico , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , GMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Modelos Animais de Doenças , Hipercolesterolemia/patologia , Hipercolesterolemia/fisiopatologia , Técnicas In Vitro , Lipídeos/sangue , Masculino , Óxido Nítrico/metabolismo , Nitritos/metabolismo , Coelhos , Rosiglitazona , Transdução de Sinais/efeitos dos fármacos , Tiazolidinedionas/uso terapêutico , Tirosina/metabolismo , Vasodilatação/efeitos dos fármacos
13.
Circulation ; 108(19): 2393-9, 2003 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-14557369

RESUMO

BACKGROUND: Peroxisome proliferator-activated receptor-alpha (PPAR-alpha) is expressed in the heart and regulates genes involved in myocardial fatty acid oxidation (FAO). The role of PPAR-alpha in acute ischemia/reperfusion myocardial injury remains unclear. METHODS AND RESULTS: The coronary arteries of male mice were ligated for 30 minutes. After reperfusion for 24 hours, ischemic and infarct sizes were determined. A highly selective and potent PPAR-alpha agonist, GW7647, was administered by mouth for 2 days, and the third dose was given 1 hour before ischemia. GW7647 at 1 and 3 mg x kg(-1) x d(-1) reduced infarct size by 28% and 35%, respectively (P<0.01), and myocardial contractile dysfunction was also improved. Cardioprotection by GW7647 was completely abolished in PPAR-alpha-null mice. Ischemia/reperfusion downregulated mRNA expression of cardiac PPAR-alpha and FAO enzyme genes, decreased myocardial FAO enzyme activity and in vivo cardiac fat oxidation, and increased serum levels of free fatty acids. All of these changes were reversed by GW7647. Moreover, GW7647 attenuated ischemia/reperfusion-induced release of multiple proinflammatory cytokines and inhibited neutrophil accumulation and myocardial expression of matrix metalloproteinases-9 and -2. Furthermore, GW7647 inhibited nuclear factor-kappaB activation in the heart, accompanied by enhanced levels of inhibitor-kappaBalpha. CONCLUSIONS: Activation of PPAR-alpha protected the heart from reperfusion injury. This cardioprotection might be mediated through metabolic and antiinflammatory mechanisms. This novel effect of the PPAR-alpha agonist could provide an added benefit to patients treated with PPAR-alpha activators for dyslipidemia.


Assuntos
Butiratos/uso terapêutico , Isquemia Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Compostos de Fenilureia/uso terapêutico , Receptores Citoplasmáticos e Nucleares/agonistas , Fatores de Transcrição/agonistas , Animais , Butiratos/administração & dosagem , Quimiotaxia de Leucócito/efeitos dos fármacos , Citocinas/metabolismo , Regulação para Baixo/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Ácidos Graxos/sangue , Proteínas I-kappa B/biossíntese , Ligadura , Masculino , Metaloproteinases da Matriz/biossíntese , Camundongos , Camundongos Knockout , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/patologia , Isquemia Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/enzimologia , Inibidor de NF-kappaB alfa , NF-kappa B/antagonistas & inibidores , Oxirredução , Compostos de Fenilureia/administração & dosagem , Pré-Medicação , RNA Mensageiro/biossíntese , Receptores Citoplasmáticos e Nucleares/deficiência , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/fisiologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia
14.
Trends Pharmacol Sci ; 25(4): 225-33, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15063087

RESUMO

The latter half of the 20th century has been characterized by pharmacologists as the 'age of the receptor', an era in which the bioassay, that stalwart of classical pharmacology, has played a seminal role in identifying novel cardiovascular medicines. In this article, we ask what, if anything, has changed in the pharmacologist's approach to discovering novel cardiovascular drugs on this, the 25th anniversary of the inaugural publication of Trends in Pharmacological Sciences.


Assuntos
Fármacos Cardiovasculares/uso terapêutico , Farmacologia/tendências , Receptores Purinérgicos/efeitos dos fármacos , Animais , Humanos , Receptores Purinérgicos/genética , Receptores Purinérgicos/metabolismo
15.
Br J Pharmacol ; 145(5): 620-35, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15852036

RESUMO

1. SB-706375 potently inhibited [(125)I]hU-II binding to both mammalian recombinant and 'native' UT receptors (K(i) 4.7+/-1.5 to 20.7+/-3.6 nM at rodent, feline and primate recombinant UT receptors and K(i) 5.4+/-0.4 nM at the endogenous UT receptor in SJRH30 cells). 2. Prior exposure to SB-706375 (1 microM, 30 min) did not alter [(125)I]hU-II binding affinity or density in recombinant cells (K(D) 3.1+/-0.4 vs 5.8+/-0.9 nM and B(max) 3.1+/-1.0 vs 2.8+/-0.8 pmol mg(-1)) consistent with a reversible mode of action. 3. The novel, nonpeptidic radioligand [(3)H]SB-657510, a close analogue of SB-706375, bound to the monkey UT receptor (K(D) 2.6+/-0.4 nM, B(max) 0.86+/-0.12 pmol mg(-1)) in a manner that was inhibited by both U-II isopeptides and SB-706375 (K(i) 4.6+/-1.4 to 17.6+/-5.4 nM) consistent with the sulphonamides and native U-II ligands sharing a common UT receptor binding domain. 4. SB-706375 was a potent, competitive hU-II antagonist across species with pK(b) 7.29-8.00 in HEK293-UT receptor cells (inhibition of [Ca(2+)](i)-mobilization) and pK(b) 7.47 in rat isolated aorta (inhibition of contraction). SB-706375 also reversed tone established in the rat aorta by prior exposure to hU-II (K(app) approximately 20 nM). 5. SB-706375 was a selective U-II antagonist with >/=100-fold selectivity for the human UT receptor compared to 86 distinct receptors, ion channels, enzymes, transporters and nuclear hormones (K(i)/IC(50)>1 microM). Accordingly, the contractile responses induced in isolated aortae by KCl, phenylephrine, angiotensin II and endothelin-1 were unaltered by SB-706375 (1 microM). 6. In summary, SB-706375 is a high-affinity, surmountable, reversible and selective nonpeptide UT receptor antagonist with cross-species activity that will assist in delineating the pathophysiological actions of U-II in mammals.


Assuntos
Pirrolidinas/farmacologia , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/genética , Sulfonamidas/farmacologia , Algoritmos , Animais , Aorta Torácica/efeitos dos fármacos , Ligação Competitiva/efeitos dos fármacos , Gatos , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Haplorrinos , Humanos , Técnicas In Vitro , Camundongos , Contração Muscular/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Ensaio Radioligante , Ratos , Proteínas Recombinantes/metabolismo , Rabdomiossarcoma/metabolismo , Especificidade da Espécie
16.
Peptides ; 26(12): 2464-72, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16026900

RESUMO

The vasoactive peptide urotensin-II (U-II) is best known for its ability to regulate peripheral vascular and cardiac contractile function in vivo, and recent in vitro studies have suggested a role for the peptide in the control of vascular remodeling by inducing smooth muscle proliferation and fibroblast-mediated collagen deposition. Therefore, U-II may play a role in the etiology of atherosclerosis. In the present study we sought to determine the expression of U-II in coronary arteries from patients with coronary atherosclerosis and from normal control subjects, using immunohistochemistry and in situ hybridization. In normal coronary arteries, there was little expression of U-II in all types of cells. In contrast, in patients with coronary atherosclerosis, endothelial expression of U-II was significantly increased in all diseased segments (P<0.05). Greater expression of U-II was noted in endothelial cells of lesions with subendothelial inflammation or fibrofatty lesion compared with that of endothelial cells underlined by dense fibrosis or minimal intimal thickening. Myointimal cells and foam cells also expressed U-II. In most diseased segments, medial smooth muscle cells exhibited moderate expression of U-II. These findings demonstrate upregulation of U-II in endothelial, myointimal and medial smooth muscle cells of atherosclerotic human coronary arteries, and suggest a possible role for U-II in the pathogenesis of coronary atherosclerosis.


Assuntos
Doença da Artéria Coronariana/metabolismo , Vasos Coronários/metabolismo , Células Espumosas/metabolismo , Regulação da Expressão Gênica , Túnica Íntima/metabolismo , Urotensinas/biossíntese , Adulto , Doença da Artéria Coronariana/patologia , Vasos Coronários/patologia , Feminino , Células Espumosas/patologia , Humanos , Imuno-Histoquímica , Hibridização In Situ , Masculino , Pessoa de Meia-Idade , Túnica Íntima/patologia
17.
Cardiovasc Res ; 62(1): 135-44, 2004 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15023560

RESUMO

OBJECTIVES: This study examined the effects of diet-induced hypercholesterolemia on the extent of postischemic myocyte apoptosis and elucidated the potential mechanisms involved. Furthermore, the effects of rosiglitazone (RSG) on postischemic myocardial apoptosis in HC rabbits were investigated. METHODS: Male New Zealand rabbits were fed normal or high cholesterol diets for 8 weeks. Three weeks after being fed a high cholesterol diet, HC rabbits were randomized to receive vehicle or RSG during the remaining 5 weeks. Rabbits were then subjected to 60 min of coronary occlusion followed by 4 h of reperfusion. RESULTS: Compared with rabbits fed with a normal diet, HC rabbits had increased caspase-3 activity, apoptotic cell death and retarded contractile function recovery after reperfusion. HC increased iNOS expression, total NOx and nitrotyrosine contents, indicating that an increased nitrative stress occurred in HC myocardial tissue. Activity of a hypertrophic/anti-apoptotic mitogen-activated protein kinase (MAPK), ERK1/2, was significantly decreased while activation of a pro-apoptotic MAPK, p38, was increased. Treatment with RSG in HC rabbits attenuated postischemic myocardial nitrative stress, restored a beneficial balance between pro- and anti-apoptotic MAPK signaling, reduced postischemic myocardial apoptosis, and improved cardiac functional recovery. CONCLUSIONS: Our results demonstrated that HC increased postischemic myocardial apoptosis likely by increasing the production of pro-apoptotic molecules, activating pro-apoptotic signaling pathways and inhibiting anti-apoptotic signaling. In addition, our results suggest that peroxisome proliferator-activated receptor (PPAR) gamma agonists may not only attenuate the formation of atherosclerosis associated with hypercholesterolemia as previously reported, but may also protect the heart from subsequent ischemic/reperfusion-induced myocardial apoptosis.


Assuntos
Hipercolesterolemia/tratamento farmacológico , Isquemia Miocárdica/tratamento farmacológico , Receptores Citoplasmáticos e Nucleares/agonistas , Tiazolidinedionas/uso terapêutico , Fatores de Transcrição/agonistas , Animais , Apoptose , Dieta , Indução Enzimática , Hipercolesterolemia/patologia , Marcação In Situ das Extremidades Cortadas , Sistema de Sinalização das MAP Quinases , Masculino , Isquemia Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/patologia , Óxido Nítrico/metabolismo , Coelhos , Rosiglitazona
18.
Cardiovasc Res ; 53(2): 414-22, 2002 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11827692

RESUMO

BACKGROUND: In vitro evidence suggests that the p38 mitogen-activated protein kinase (p38 MAPK) plays a crucial role in PMN activation and inflammatory cytokine production. However, the effect of p38 MAPK on myocardial reperfusion injury, a pathologic condition involving a typical inflammatory response, has not been fully examined. In the present study, we investigated the effect of SB 239063, a specific p38 MAPK inhibitor, on myocardial injury in a murine ischemia/reperfusion (I/R) model and elucidated the mechanism by which p38 MAPK inhibitor may exert its protective effect against I/R injury. METHODS AND RESULTS: I/R resulted in a significant myocardial injury (myocardial infarct 45 +/- 2.9%) and marked PMN accumulation (myeloperoxidase activity 1.03 +/- 0.16 U/100 g tissue). Administration of SB 239063 significantly inhibited the myocardial inflammatory response as evidenced by reduced PMN accumulation in I/R myocardial tissue (0.62 +/- 0.008 U/100 g tissue, P<0.01 vs. vehicle), and markedly attenuated myocardial reperfusion injury (myocardial infarct size: 28 +/- 2.4%, P<0.01 vs. vehicle). Moreover, treatment with SB 239063 significantly attenuated I/R-induced P-selectin and ICAM-1 upregulation (13.8 +/- 2.7 vs. 23.9 +/- 3.1%, and 29.4 +/- 1.6 vs. 56.3 +/- 4.8%, respectively P<0.01). In addition, pre-treatment with R15.7, a monoclonal antibody against CD 18 adhesion molecule on PMN surface that virtually abolished PMN accumulation in ischemic-reperfused myocardial tissue, significantly, but not completely, blocked the cardioprotection exerted by SB 239063. CONCLUSION: These results demonstrated for the first time that p38 MAPK activation plays a significant role in adhesion molecule upregulation on ischemia-reperfused endothelial cells and is an important signaling step in the pathogenesis of PMN-mediated tissue injury.


Assuntos
Endotélio Vascular/metabolismo , Imidazóis/farmacologia , Molécula 1 de Adesão Intercelular/análise , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Pirimidinas/farmacologia , Transdução de Sinais , Análise de Variância , Animais , Masculino , Proteínas Quinases Ativadas por Mitógeno/análise , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/imunologia , Neutrófilos/fisiologia , Selectina-P/análise , Peroxidase/análise , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Proteínas Quinases p38 Ativadas por Mitógeno
19.
Cardiovasc Res ; 54(3): 549-58, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12031700

RESUMO

OBJECTIVE: Matrix metalloproteinase-9 (MMP-9) activity is up regulated in the heart subjected to ischemic insult. Whether increased MMP-9 activity contributes to acute myocardial injury after ischemia-reperfusion remains unknown. To investigate the role of MMP-9 in myocardial infarction, we utilized a MMP-9 knockout mouse. METHODS AND RESULTS: Standard homologous recombination in embryonic stem cells was used to generate a mouse lacking MMP-9. The left anterior descending coronary artery was occluded for 30 min followed by 24 h reperfusion, and the ischemic and infarct sizes were determined. Targeted deletion of MMP-9 protected the heart from no-flow ischemia-reperfusion-induced myocardial injury. The myocardial infarct size was reduced by 17.5% in MMP-9 heterozygotes (+/-) (P<0.01) and 35.4% in MMP-9 knockout (-/-) mice (P<0.01) versus the wild-type (+/+) mice, respectively. Analysis of MMP activity in myocardial extracts by zymography demonstrated that ischemia-reperfusion-induced expression of proMMP-9 and active MMP-9 was reduced by 77.8% (P<0.01) and 69.1% (P<0.001), respectively, in (+/-) mice compared to (+/+) mice, and was absent in (-/-) animals. The expression of TIMP-1, an endogenous inhibitor of MMP-9, was elevated 4.7-fold (P<0.05) and 21.4-fold (P<0.05) in the (+/-) and (-/-) mice, respectively, compared to (+/+) mice. Immunohistochemical analysis revealed that neutrophils were the primary cellular source of MMP-9, and less neutrophils were detected in the ischemic region of the heart following ischemia-reperfusion in (-/-) mice compared to (+/+) mice. Measurement of myeloperoxidase activity, a marker enzyme of neutrophils, demonstrated a 44% reduction in neutrophils infiltrated into the ischemic myocardium in the (-/-) mice compared to the (+/+) mice (P<0.05). CONCLUSION: These results suggest that MMP-9 plays an important role in ischemia-reperfusion-induced myocardial infarction and MMP-9 could be a target for prevention or treatment of acute ischemic myocardial injury.


Assuntos
Deleção de Genes , Metaloproteinase 9 da Matriz/genética , Traumatismo por Reperfusão Miocárdica/enzimologia , Miocárdio/enzimologia , Animais , Imuno-Histoquímica/métodos , Metaloproteinase 2 da Matriz/análise , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/análise , Camundongos , Camundongos Knockout , Modelos Animais , Traumatismo por Reperfusão Miocárdica/genética , Miocárdio/imunologia , Infiltração de Neutrófilos , Neutrófilos/enzimologia , Inibidor Tecidual de Metaloproteinase-1/análise , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Remodelação Ventricular
20.
Stroke ; 33(2): 578-85, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11823673

RESUMO

BACKGROUND AND PURPOSE: Although used clinically to prevent stroke, there are few examples of anticoagulant investigations in the treatment of acute thromboembolic stroke in animal models. The treatment of thromboembolic stroke in experimental models has been investigated almost exclusively around the use of tissue plasminogen activator (tPA). In this study, using a rat thromboembolic stroke model, we investigated the use of an inhibitory anti-factor IX(a) monoclonal antibody (SB 249417) for the treatment of thromboembolic stroke and compared its efficacy to that of tPA. METHODS: Stroke was initiated by delivering 6 clots into the internal carotid artery. After 2, 4, or 6 hours, rats received either intravenous vehicle, 10.0 mg/kg tPA, or 1.0, 2.0, or 3.0 mg/kg SB 249417. At 24 hours after stroke, infarct volumes and neurological deficits were assessed. RESULTS: Treatment with tPA 2, 4, or 6 hours after stroke reduced infarct volumes by 35% (P=NS), 45%, and 39%, respectively. tPA treatment did not improve neurological deficits at any time point. Treatment with SB 249417 (3.0 mg/kg) 2, 4, or 6 hours after stroke reduced infarct volumes by 44%, 50%, and 13% (P=NS), respectively. Neurological deficits were reduced by 49%, 42%, and 13% (P=NS), respectively. Neither mortality nor hemorrhage was affected by either treatment. CONCLUSIONS: The data indicate that the inhibition of factor IX(a) within 4 hours of thromboembolic stroke produced a more favorable outcome than tPA. When treatment was initiated 6 hours after stroke, the benefits of factor IX(a) inhibition were lost, whereas tPA continued to suppress lesion development, albeit without a corresponding improvement in functional deficits. This study suggests that cerebral ischemia and the resultant perfusion deficit are exacerbated by the activation of blood coagulation and that anticoagulants like SB 249417 may find utility in the treatment of ischemic stroke.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Fator IXa/antagonistas & inibidores , Fibrinolíticos/uso terapêutico , Acidente Vascular Cerebral/terapia , Tromboembolia/terapia , Doença Aguda , Animais , Comportamento Animal/efeitos dos fármacos , Modelos Animais de Doenças , Progressão da Doença , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Ativação Enzimática/efeitos dos fármacos , Exame Neurológico , Ativadores de Plasminogênio/uso terapêutico , Prosencéfalo/irrigação sanguínea , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/patologia , Ratos , Acidente Vascular Cerebral/etiologia , Acidente Vascular Cerebral/fisiopatologia , Taxa de Sobrevida , Tromboembolia/complicações , Tromboembolia/patologia , Fatores de Tempo , Ativador de Plasminogênio Tecidual/uso terapêutico , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA