Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 185
Filtrar
Mais filtros

País/Região como assunto
Intervalo de ano de publicação
1.
Diabetes Obes Metab ; 19(6): 901-905, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28105731

RESUMO

We assessed the effects of liraglutide treatment on five cardiovascular risk biomarkers, reflecting different pathophysiology: tumour necrosis factor (TNF)-α; soluble urokinase plasminogen activator receptor (suPAR); mid-regional pro-adrenomedullin (MR-proADM); mid-regional pro-atrial natriuretic peptide (MR-proANP); and copeptin, in people with type 2 diabetes with albuminuria. In a randomized, double-blind, placebo-controlled, crossover trial we enrolled people with type 2 diabetes and persistent albuminuria (urinary albumin-to-creatinine ratio [UACR] >30 mg/g) and estimated glomerular filtration rate (eGFR) ≥30 mL/min/1.73 m2 . Participants received liraglutide (1.8 mg/d) and matched placebo for 12 weeks, in random order. The primary endpoint was change in albuminuria; this was a prespecified sub-study. A total of 32 participants were randomized, of whom 27 completed the study. TNF-α level was 12% (95% confidence interval [CI] 3; 20) lower after liraglutide treatment compared with placebo (P = .012); MR-proADM level was 4% (95% CI 0; 8) lower after liraglutide treatment compared with placebo (P = .038), and MR-proANP level was 13% (95% CI 4; 21) lower after liraglutide treatment compared with placebo (P = .006). In the present study, we showed anti-inflammatory effects of liraglutide treatment, reflected in reductions in levels of TNF-α and MR-proADM, while the reduction in MR-proANP levels may represent a clinically relevant benefit with regard to heart failure.


Assuntos
Albuminúria/sangue , Doenças Cardiovasculares/etiologia , Diabetes Mellitus Tipo 2/sangue , Hipoglicemiantes/uso terapêutico , Liraglutida/uso terapêutico , Adrenomedulina/sangue , Adrenomedulina/efeitos dos fármacos , Idoso , Albuminúria/tratamento farmacológico , Albuminúria/etiologia , Fator Natriurético Atrial/sangue , Fator Natriurético Atrial/efeitos dos fármacos , Biomarcadores/sangue , Estudos Cross-Over , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/tratamento farmacológico , Método Duplo-Cego , Feminino , Glicopeptídeos/sangue , Glicopeptídeos/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Fragmentos de Peptídeos/sangue , Fragmentos de Peptídeos/efeitos dos fármacos , Precursores de Proteínas/sangue , Precursores de Proteínas/efeitos dos fármacos , Receptores de Ativador de Plasminogênio Tipo Uroquinase/sangue , Receptores de Ativador de Plasminogênio Tipo Uroquinase/efeitos dos fármacos , Fatores de Risco , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/efeitos dos fármacos
2.
Am J Physiol Heart Circ Physiol ; 311(3): H604-12, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27342880

RESUMO

BIBF1000 is a small molecule tyrosine kinase inhibitor targeting vascular endothelial growth factor receptor (VEGFR), fibroblast growth factor receptor (FGFR), and platelet-derived growth factor receptor (PDGFR) and is a powerful inhibitor of fibrogenesis. BIBF1000 is very similar to BIBF1120 (nintedanib), a drug recently approved for the treatment of idiopathic pulmonary fibrosis (IPF). A safety concern pertaining to VEGFR, FGFR, and PDGFR inhibition is the possible interference with right ventricular (RV) responses to an increased afterload, which could adversely affect clinical outcome in patients with IPF who developed pulmonary hypertension. We tested the effect of BIBF1000 on the adaptation of the RV in rats subjected to mechanical pressure overload. BIBF1000 was administered for 35 days in pulmonary artery-banded (PAB) rats. RV adaptation was assessed by echocardiography, pressure volume loop analysis, histology, and determination of atrial natriuretic peptide (ANP) expression. BIBF1000 treatment resulted in growth attenuation but had no effects on RV function after PAB, given absence of changes in cardiac index, end-systolic elastance, connective tissue disposition, and capillary density. We conclude that, in this experimental model of increased afterload, combined VEGFR, FGFR, and PDGFR inhibition does not hamper RV adaptation to pressure overload.


Assuntos
Adaptação Fisiológica/efeitos dos fármacos , Ventrículos do Coração/efeitos dos fármacos , Indóis/farmacologia , Pressão , Inibidores de Proteínas Quinases/farmacologia , Função Ventricular Direita/efeitos dos fármacos , Animais , Fator Natriurético Atrial/efeitos dos fármacos , Fator Natriurético Atrial/metabolismo , Western Blotting , Modelos Animais de Doenças , Ecocardiografia , Hipertensão Pulmonar Primária Familiar/diagnóstico por imagem , Hipertensão Pulmonar Primária Familiar/metabolismo , Hipertensão Pulmonar Primária Familiar/patologia , Ventrículos do Coração/diagnóstico por imagem , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Masculino , Proteína Quinase 1 Ativada por Mitógeno/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/efeitos dos fármacos , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Artéria Pulmonar/cirurgia , Ratos , Ratos Sprague-Dawley , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores
3.
Climacteric ; 18(1): 86-93, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25017924

RESUMO

OBJECTIVE: To assess the effects of oral low-dose and non-oral hormone therapy (HT) on ultra-sensitive C-reactive protein (CRP), atrial natriuretic peptide (ANP), and cardiovascular risk factors in postmenopause. METHODS: In this randomized, cross-over study, 44 recently postmenopausal women, with no clinical evidence of cardiovascular disease, received oral low-dose HT (estradiol 1 mg + drospirenone 2 mg/day) for 3 months. Forty-two patients received non-oral, conventional HT (1.5 mg/day percutaneous 17ß-estradiol gel or equivalent for nasal route) for 3 months followed by 200 mg/day micronized progesterone by the vaginal route (14 days during each menstrual period). After 3 months, patients were crossed over without washout. Post-HT vs. pre-HT measures were determined: lipids, glucose, body mass index, waist circumference, fibrinogen, CRP-stratified levels, and ANP levels. The study was registered at clinical trials.gov (NCT01432028). RESULTS: The mean age was 51 ± 3 years and the mean time since the menopause was 22 ± 10 months. CRP-stratified high levels decreased in a higher number of non-oral HT patients, who moved to intermediate and low levels (p = 0.02). No effect of HT was observed on ANP levels (baseline 67.4 (18.4-104.5), low-dose oral 43.5 (14.4-95.9), non-oral 39.8 (15.5-67.5) pg/ml). Markers of endothelial function did not worsen with either low-dose oral or non-oral HT: von Willebrand factor (baseline 118 ± 37%, low-dose oral 119 ± 38%, non-oral 108 ± 3%, p < 0.01), fibrinogen (baseline 356 ± 58 mg/dl; low-dose oral 343 ± 77 mg/dl; non-oral 326 ± 71 mg/dl, p < 0.01). CONCLUSIONS: Low-dose oral and non-oral HT for 6 months had neutral or beneficial effects in recently postmenopausal women with no clinical evidence of cardiovascular disease.


Assuntos
Androstenos/administração & dosagem , Fator Natriurético Atrial/efeitos dos fármacos , Proteína C-Reativa/efeitos dos fármacos , Estradiol/administração & dosagem , Terapia de Reposição de Estrogênios/métodos , Pós-Menopausa/efeitos dos fármacos , Administração Cutânea , Administração Intranasal , Administração Oral , Fator Natriurético Atrial/metabolismo , Proteína C-Reativa/metabolismo , Doenças Cardiovasculares , Estudos Cross-Over , Combinação de Medicamentos , Feminino , Géis , Humanos , Pessoa de Meia-Idade , Fatores de Risco
4.
Clin Exp Hypertens ; 37(5): 353-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25496380

RESUMO

Bifunctional angiotensin II (Ang II) type 1 (AT1) receptor blockers (ARBs) that can block the activation of not only AT1 receptor, but also neprilysin, which metabolizes vasoactive peptides including atrial natriuretic peptide (ANP), are currently being developed. However, the usefulness of the inactivation of ANP in addition to the AT1 receptor with regard to aldosterone (Ald) synthesis is not yet clear. We evaluated the inhibitory effects of various ARBs combined with or without ANP on Ang II-induced adrenal Ald synthesis using a human adrenocortical cell line (NCI-H295R). Ang II increased Ald synthesis in a dose- and time-dependent manner. Ald synthesis induced by Ang II was completely blocked by azilsartan, but not PD123319 (AT2 receptor antagonist). CGP42112 AT2 receptor agonist did not affect Ald synthesis. While most ARBs block Ang II-induced Ald synthesis to different extents, azilsartan and olmesartan have similar blocking effects on Ald synthesis. The different effects of ARBs were particularly observed at 10(-7) and 10(-8 )M. ANP attenuated Ang II-induced Ald synthesis, and ANP-mediated attenuation of Ang II-induced Ald synthesis were blocked by inhibitors of G-protein signaling subtype 4 and protein kinase G. ANP (10(-8) and 10(-7 )M) without ARBs inhibited Ald synthesis, and the combination of ANP (10(-7 )M) and ARB (10(-8 )M) had an additive effect with respect to the inhibition of Ald synthesis. In conclusions, ARBs had differential effects on Ang II-induced Ald synthesis, and ANP may help to block Ald synthesis when the dose of ARB is not sufficient to block its secretion.


Assuntos
Glândulas Suprarrenais/metabolismo , Aldosterona/biossíntese , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Fator Natriurético Atrial/metabolismo , Hipertensão , Glândulas Suprarrenais/patologia , Fator Natriurético Atrial/efeitos dos fármacos , Células Cultivadas , Humanos , Hipertensão/tratamento farmacológico , Hipertensão/metabolismo , Hipertensão/patologia
5.
J Cardiovasc Pharmacol ; 61(6): 545-52, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23474841

RESUMO

: Caveolin-1-deficient (cav1) mice display a severely diseased cardiac phenotype with systolic and diastolic heart failure. Accumulating evidence supports a causative role of uncoupled endothelial nitric oxide synthase in the development of these abnormalities. Interestingly, a similar molecular mechanism was proposed for anthracycline-induced cardiomyopathy. Currently, dexrazoxane is approved for the prevention of anthracycline-induced cardiomyopathy. Given the molecular similarities between the anthracycline-induced cardiomyopathy and the cardiomyopathy in cav1 mice, we questioned whether dexrazoxane may also prevent the evolution of the cardiac pathologies in cav1 mice. We evaluated dexrazoxane treatment for 6 weeks in cav1 mice and wild-type controls. This study provides the first evidence for a reduced reactive oxygen species formation in the vessels of dexrazoxane-treated cav1 mice. This reduced oxidative stress resulted in a markedly reduced rate of apoptosis, which finally was translated into a significantly improved heart function in dexrazoxane-treated cav1 mice. These hemodynamic improvements were accompanied by significantly lowered proatrial natriuretic peptide levels. Notably, these protective properties of dexrazoxane were not evident in wild-type animals. Taken together, these novel findings indicate that dexrazoxane significantly reduces vascular reactive oxygen species formation cav1. Because this is paralleled by an improved cardiac performance in cav1 mice, our data suggest dexrazoxane as a novel therapeutic strategy in this specific cardiomyopathy.


Assuntos
Cardiomiopatias/prevenção & controle , Fármacos Cardiovasculares/uso terapêutico , Caveolina 1/deficiência , Razoxano/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Função Ventricular Esquerda/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Fator Natriurético Atrial/sangue , Fator Natriurético Atrial/efeitos dos fármacos , Cardiomiopatias/genética , Cardiomiopatias/metabolismo , Cardiomiopatias/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase/efeitos dos fármacos , Óxido Nítrico Sintase/metabolismo , Fenótipo , Resultado do Tratamento
6.
J Anesth ; 27(6): 901-8, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23736823

RESUMO

PURPOSE: Atrial natriuretic peptide (ANP) has been known to be protective against hepatic ischemia/reperfusion injury. The purpose of this study was to verify the hypothesis that ANP conserves microvascular circulation and reduces ischemia-reperfusion injury in the in vivo rabbit model. METHODS: With IRB approval, 30 male Japanese white rabbits under pentobarbital anesthesia were studied. These animals were randomly assigned to the following three groups (n = 10 each): control, ANP, and sham group. Animals in the ANP group received continuous infusion of ANP at 0.1 µg/kg/min throughout the study period. Animals in control and ANP groups underwent 90 min of partial hepatic ischemia by clamping the right hepatic artery and portal vein. Descending aortic blood flow (AoF) was monitored with a transit-time ultrasound flowmeter. Hepatic tissue microvascular blood flow (HTBF) at both right (ischemic) and left (nonischemic) lobe was intermittently evaluated with the hydrogen clearance method. After 180 min of reperfusion, hepatic injury was determined with serum AST and ALT. Galactose clearance of reperfused right lobe was also measured as an indicator of hepatic metabolic function. Histopathological change and the number of apoptotic hepatocytes were also evaluated. RESULTS: Systemic hemodynamic data including mean arterial pressure, heart rate, and AoF did not differ among the three groups during the study period. ANP attenuated ischemia-induced right HTBF decrease. ANP also suppressed histopathological degeneration, apoptosis, and decline in galactose clearance after reperfusion. CONCLUSIONS: ANP attenuated hepatic microvascular dysfunction and hepatocyte injury after reperfusion without significant hemodynamic change.


Assuntos
Fator Natriurético Atrial/metabolismo , Fator Natriurético Atrial/farmacologia , Fígado/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo , Animais , Apoptose/efeitos dos fármacos , Pressão Arterial/efeitos dos fármacos , Fator Natriurético Atrial/efeitos dos fármacos , Galactose/metabolismo , Frequência Cardíaca/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Circulação Hepática/efeitos dos fármacos , Testes de Função Hepática/métodos , Masculino , Microcirculação/efeitos dos fármacos , Coelhos , Distribuição Aleatória
7.
Am J Physiol Renal Physiol ; 302(1): F141-9, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21957179

RESUMO

Angiotensin-converting enzyme (ACE) inhibitors ameliorate the progression of renal disease. In combination with vitamin D receptor activators, they provide additional benefits. In the present study, uremic (U) rats were treated as follows: U+vehicle (UC), U+enalapril (UE; 25 mg/l in drinking water), U+paricalcitol (UP; 0.8 µg/kg ip, 3 × wk), or U+enalapril+paricalcitol (UEP). Despite hypertension in UP rats, proteinuria decreased by 32% vs. UC rats. Enalapril alone, or in combination with paricalcitol, further decreased proteinuria (≈70%). Glomerulosclerosis and interstitial infiltration increased in UC rats. Paricalcitol and enalapril inhibited this. The increase in cardiac atrial natriuretic peptide (ANP) seen in UC rats was significantly decreased by paricalcitol. Enalapril produced a more dramatic reduction in ANP. Renal oxidative stress plays a critical role in inflammation and progression of sclerosis. The marked increase in p22(phox), a subunit of NADPH oxidase, and decrease in endothelial nitric oxide synthase were inhibited in all treated groups. Cotreatment with both compounds inhibited the uremia-induced increase in proinflammatory inducible nitric oxide synthase (iNOS) and glutathione peroxidase activity better than either compound alone. Glutathione reductase was also increased in UE and UP rats vs. UC. Kidney 4-hydroxynonenal was significantly increased in the UC group compared with the normal group. Combined treatment with both compounds significantly blunted this increase, P < 0.05, while either compound alone had no effect. Additionally, the expression of Mn-SOD was increased and CuZn-SOD decreased by uremia. This was ameliorated in all treatment groups. Cotreatment with enalapril and paricalcitol had an additive effect in increasing CuZn-SOD expression. In conclusion, like enalapril, paricalcitol alone can improve proteinuria, glomerulosclerosis, and interstitial infiltration and reduce renal oxidative stress. The effects of paricalcitol may be amplified when an ACE inhibitor is added since cotreatment with both compounds seems to have an additive effect on ameliorating uremia-induced changes in iNOS and CuZn-SOD expression, peroxidase activity, and renal histomorphometry.


Assuntos
Enalapril/uso terapêutico , Ergocalciferóis/uso terapêutico , Glomerulonefrite/tratamento farmacológico , Proteinúria/tratamento farmacológico , Receptores de Calcitriol/efeitos dos fármacos , Aldeídos/metabolismo , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Fator Natriurético Atrial/efeitos dos fármacos , Feminino , Rim/efeitos dos fármacos , NADPH Oxidases/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Proteinúria/metabolismo , Ratos , Superóxido Dismutase/metabolismo , Uremia/metabolismo
8.
Biomed Pharmacother ; 146: 112433, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35062052

RESUMO

Oryeongsan (ORS), a herbal medicine formula, has long been used for the treatment of impaired body water balance in Asian countries. Recently, it was shown that ORS administration modulates the renin-angiotensin system (RAS). Purpose of the present study was to determine characteristics of atrial ANP secretion and effects of ORS on the secretion in the atria from spontaneously hypertensive rats (SHR). Normotensive WKY groups (WKY-V, WKY-ORS, WKY-LOS) and hypertensive SHR groups (SHR-V, SHR-ORS, SHR-LOS) treated with vehicle, ORS, and losartan as a positive control group, respectively, were used. Experiments were performed in perfused beating atria (1.3 Hz) allowing atrial distension, acetylcholine (ACh) stimulation, and serial collection of atrial perfusates. The secreted ANP concentration was measured using radioimmunoassay. Interstitial fluid (ISF) translocation was measured using [3H]inulin clearance. Stepwise increase in atrial distension by 1.1, 2.0, and 2.7 cmH2O above basal distension further increased ANP secretion proportionally in the atria from WKY-V, but the response was significantly suppressed in the atria from SHR-V. Cardiomyocyte ANP release, the first step of atrial ANP secretion, was suppressed in the atria from SHR-V compared to those from WKY-V (-8.02 ±â€¯2.86, -15.86 ±â€¯2.27, and -20.09 ±â€¯3.62%; n = 8, for SHR-V vs. 8.59 ±â€¯2.81, 15.65 ±â€¯7.14, and 38.12 ±â€¯8.28%; n = 8, for WKY-V; p < 0.001 for all stepwise distension, respectively). Chronic treatment with ORS reversed the suppressed ANP release in atria from SHR-ORS group (6.76 ±â€¯3.92, 9.12 ±â€¯2.85, and 28.79 ±â€¯1.79% for SHR-ORS; n = 5 vs. SHR-V; n = 8; p = 0.01, p < 0.001, p < 0.001, respectively). The effects of ORS were comparable to those of losartan. Trans-endocardial translocation of ISF, the second step of atrial ANP secretion was similar in the atria from the hypertensive SHR-V and normotensive WKY-V. ACh-induced ANP secretion and cardiomyocyte ANP release were also suppressed in the atria from SHR-V compared to WKY-V and ORS reversed the suppression. These findings were accompanied with accentuation of the AT1 receptor expression and suppression of the AT2/Mas receptor, M2 mACh receptor and GIRK4, a molecular component of KACh channel, expression in the atria from SHR-V. Further, treatment with ORS or losartan reversed the expressions in the groups of SHR-ORS and SHR-LOS. These results show that ANP secretion is suppressed in the atria from SHR in association with accentuation of AT1 receptor and suppression of AT2/Mas receptor and KACh channel expression. Treatment with ORS ameliorates impaired ANP secretion through improving cardiomyocyte ANP release with modulation of the cardiac RAS and muscarinic signaling. These findings provide experimental evidence which supports the effect of ORS on the regulation of atrial ANP secretion in the atria from SHR.


Assuntos
Fator Natriurético Atrial/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Líquido Extracelular/efeitos dos fármacos , Extratos Vegetais/farmacologia , Aldosterona/metabolismo , Animais , Medicamentos de Ervas Chinesas/administração & dosagem , Humanos , Masculino , Ratos , Ratos Endogâmicos WKY , Renina/efeitos dos fármacos
9.
Acta Cardiol ; 66(1): 21-7, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21446378

RESUMO

OBJECTIVE: The Anglo-Scandinavian Cardiac Outcomes Trial (ASCOT-BPLA) showed that an amlodipine-based regimen prevented more cardiovascular events than an atenolol-based regimen in patients at high risk of hypertension.The basis of this difference is partly unknown and may be due to their divergent effects on the remodelling process of hypertensive heart disease. METHODS AND RESULTS: We measured plasma levels of aminoterminal propeptide of atrial natriuretic peptide (NT-proANP) and aminoterminal propeptide of B-type natriuretic peptide and serum levels of the aminoterminal propeptide of type I procollagen (PINP), aminoterminal propeptide of type III procollagen and type I collagen telopeptide in 93 patients randomized in the ASCOT study at baseline and after two and four years and compared them with echocardiographic parameters and blood pressure. NT-proANP decreased at two years by 22 (-484 - 153) pmol/l in the amlodipine-based regimen and increased by 109 (-297 - 1545) pmol/l in the atenolol-based regimen (P < 0.001), whereas no significant difference in NT-proBNP between the arms was found. PINP levels increased by 1.8 (-29 -31) microg/l in the amlodipine-based regimen and decreased by 4.7 (-27- 31) microg/I in the atenolol-based regimen, whereas no differences were found in other collagen markers between the arms. Major echocardiographic changes were not found. CONCLUSIONS: Our results show that the two treatment regimens of ASCOT-BPLA had different effects on plasma natriuretic peptides and serological markers of collagen turnover, probably reflecting divergent effects in cardiac remodelling.


Assuntos
Anlodipino/uso terapêutico , Anti-Hipertensivos/uso terapêutico , Atenolol/uso terapêutico , Fator Natriurético Atrial/sangue , Biomarcadores/sangue , Colágeno/metabolismo , Hipertensão/tratamento farmacológico , Remodelação Ventricular/efeitos dos fármacos , Fator Natriurético Atrial/efeitos dos fármacos , Colágeno Tipo I , Humanos , Hipertensão/sangue , Hipertensão/fisiopatologia , Peptídeo Natriurético Encefálico/sangue , Fragmentos de Peptídeos/sangue , Fragmentos de Peptídeos/efeitos dos fármacos , Peptídeos , Pró-Colágeno/sangue , Pró-Colágeno/efeitos dos fármacos , Remodelação Ventricular/fisiologia
10.
Am J Physiol Regul Integr Comp Physiol ; 298(4): R1007-16, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20071611

RESUMO

Leptin is a circulating adipocyte-derived hormone that influences blood pressure (BP) and metabolism. This study was designed to define the possible role of leptin in regulation of the atrial natriuretic peptide (ANP) system using acute and chronic experiments. Intravenous infusion of rat leptin (250 microg/kg injection plus 2 microg.kg(-1).min(-1) for 20 min) into Sprague-Dawley rats increased BP by 25 mmHg and decreased plasma level of ANP from 80.3 +/- 3.45 to 51.8 +/- 3.3 pg/ml. Reserpinization attenuated the rise in BP, but not the reduction of plasma ANP during leptin infusion. N(omega)-nitro-l-arginine methyl ester prevented the effects of leptin on the reduction of ANP level. In hyperleptinemic rats that received adenovirus containing rat leptin cDNA (AdCMV-leptin), BP increased during first 2 days and then recovered to control value. Plasma concentration of ANP and expression of ANP mRNA, but not of atrial ANP, in hyperleptinemic rats were lower than in the control groups on the first and second week after administration of AdCMV-leptin. These effects were not observed by the pretreatment with N(omega)-nitro-l-arginine methyl ester. No differences in renal function and ANP receptor density in the kidney were found between hyperleptinemic and control rats. Basal ANP secretion and isoproterenol-induced suppression of ANP secretion from isolated, perfused atria of hyperleptinemic rats were not different from those of other control groups. These data suggest that leptin inhibits ANP secretion indirectly through nitric oxide without changing basal or isoproterenol-induced ANP secretion.


Assuntos
Fator Natriurético Atrial/sangue , Leptina/farmacologia , Óxido Nítrico/fisiologia , Animais , Fator Natriurético Atrial/efeitos dos fármacos , Fator Natriurético Atrial/genética , Fator Natriurético Atrial/metabolismo , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/fisiologia , Hipertensão/sangue , Hipertensão/induzido quimicamente , Isoproterenol/farmacologia , Veias Jugulares/efeitos dos fármacos , Veias Jugulares/fisiologia , Rim/efeitos dos fármacos , Rim/fisiologia , Leptina/sangue , Masculino , NG-Nitroarginina Metil Éster/farmacologia , RNA Mensageiro/sangue , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Reserpina/farmacologia
11.
J Sex Med ; 7(11): 3610-9, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20102442

RESUMO

INTRODUCTION: Receptors for natriuretic peptides have been demonstrated as potential targets for the treatment of male erectile dysfunction. AIM: This study investigates the relaxant effects of the atrial natriuretic peptide (ANP) and uroguanylin (UGN), and expression of natriuretic peptide receptors on strips of human corpora cavernosa (HCC). MAIN OUTCOME MEASURES: Quantitative analysis of natriuretic receptor expression and relaxation of precontracted strips were used to assess the membrane-bound guanylate cyclase-cyclic guanosine monophosphate (cGMP) pathway in HCC strips. METHODS: HCC was obtained from a cadaver donor at the time of collection of organs for transplantation (14-47 years) and strips were mounted in organ baths for isometric studies. RESULTS: ANP and UGN both induced concentration-dependent relaxation on HCC strips with a maximal response attained at 300 nM, corresponding to 45.4±4.0% and 49±4.8%, respectively. The relaxation is not affected by 30 µM 1H-[1,2,4]oxaolodiazolo[4,3-a]quinoxalin-1-one (ODQ) (a soluble guanylate cyclase inhibitor), but it is significantly blocked by 10 µM isatin, a nonspecific particulate guanylate cyclase (pGC) inhibitor. UGN was unable to potentiate electrical field stimulation (EFS) or acetylcholine-induced relaxations. The potential role of pGC activation and cGMP generation in this effect is reinforced by the potentiation of this effect by phosphodiesterase-5 inhibitor vardenafil (55.0±7.5-UGN vs. 98.6±1.4%-UGN+vardenafil; P<0.05). The relaxant effect was also partially (37.6%) blocked by the combination iberitoxin-apamin but was insensitive to glybenclamide. The expression of guanylate cyclase receptors (GC-A, GC-B, GC-C) and the expression of the natriuretic peptide "clearance" receptor (NPR-C) were confirmed by real-time polymerase chain reaction. The exposure of HCC strips to ANP (1 µM) and UGN (10 µM) significantly increased cGMP, but not cyclic adenosine monophosphate (cAMP) levels. CONCLUSIONS: UGN relaxes HCC strips by a guanylate cyclase and K(ca)-channel-dependent mechanism. These findings obtained in HCC reveal that the natriuretic peptide receptors are potential targets for the development of new drugs for the treatment of erectile dysfunction.


Assuntos
Fator Natriurético Atrial/metabolismo , Disfunção Erétil/tratamento farmacológico , Peptídeos Natriuréticos/farmacologia , Pênis/cirurgia , Adolescente , Adulto , Fator Natriurético Atrial/efeitos dos fármacos , Cadáver , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/metabolismo , Disfunção Erétil/enzimologia , Disfunção Erétil/metabolismo , Guanilato Ciclase/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Relaxamento Muscular/efeitos dos fármacos , Relaxamento Muscular/fisiologia , Músculo Liso/efeitos dos fármacos , Músculo Liso/fisiologia , Peptídeos Natriuréticos/metabolismo , Pênis/efeitos dos fármacos , Receptores do Fator Natriurético Atrial , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Peptídeos/efeitos dos fármacos , Receptores de Peptídeos/metabolismo , Guanilil Ciclase Solúvel , Adulto Jovem
12.
Heart Fail Clin ; 6(1): 11-23, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19945056

RESUMO

This article reviews information on the role of genetic variability in the development, progression, and treatment of heart failure. It focuses primarily on genetic variation in neurohormonal systems, where adrenergic receptors and the renin-angiotensin-aldosterone system are the two most explored areas. The article also reviews the endothelin system and natriuretic peptides. Clinical trial design issues are examined, and suggestions for pharmacogenomic clinical trials are presented. Finally, lessons from the oncology field and the changing regulatory landscape are discussed.


Assuntos
Insuficiência Cardíaca/tratamento farmacológico , Anti-Hipertensivos/uso terapêutico , Fator Natriurético Atrial/efeitos dos fármacos , Fator Natriurético Atrial/genética , Cardiotônicos/uso terapêutico , Endotelinas/efeitos dos fármacos , Endotelinas/genética , Insuficiência Cardíaca/genética , Humanos , Polimorfismo Genético , Receptores Adrenérgicos alfa/efeitos dos fármacos , Receptores Adrenérgicos alfa/genética , Receptores Adrenérgicos beta/efeitos dos fármacos , Receptores Adrenérgicos beta/genética , Sistema Renina-Angiotensina/efeitos dos fármacos , Sistema Renina-Angiotensina/genética
13.
J Cell Biol ; 156(5): 905-19, 2002 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-11864993

RESUMO

Members of the protein kinase C (PKC) isozyme family are important signal transducers in virtually every mammalian cell type. Within the heart, PKC isozymes are thought to participate in a signaling network that programs developmental and pathological cardiomyocyte hypertrophic growth. To investigate the function of PKC signaling in regulating cardiomyocyte growth, adenoviral-mediated gene transfer of wild-type and dominant negative mutants of PKC alpha, beta II, delta, and epsilon (only wild-type zeta) was performed in cultured neonatal rat cardiomyocytes. Overexpression of wild-type PKC alpha, beta II, delta, and epsilon revealed distinct subcellular localizations upon activation suggesting unique functions of each isozyme in cardiomyocytes. Indeed, overexpression of wild-type PKC alpha, but not betaI I, delta, epsilon, or zeta induced hypertrophic growth of cardiomyocytes characterized by increased cell surface area, increased [(3)H]-leucine incorporation, and increased expression of the hypertrophic marker gene atrial natriuretic factor. In contrast, expression of dominant negative PKC alpha, beta II, delta, and epsilon revealed a necessary role for PKC alpha as a mediator of agonist-induced cardiomyocyte hypertrophy, whereas dominant negative PKC epsilon reduced cellular viability. A mechanism whereby PKC alpha might regulate hypertrophy was suggested by the observations that wild-type PKC alpha induced extracellular signal-regulated kinase1/2 (ERK1/2), that dominant negative PKC alpha inhibited PMA-induced ERK1/2 activation, and that dominant negative MEK1 (up-stream of ERK1/2) inhibited wild-type PKC alpha-induced hypertrophic growth. These results implicate PKC alpha as a necessary mediator of cardiomyocyte hypertrophic growth, in part, through a ERK1/2-dependent signaling pathway.


Assuntos
Cardiomiopatia Hipertrófica/enzimologia , Diferenciação Celular/fisiologia , Coração/crescimento & desenvolvimento , Isoenzimas/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Miocárdio/enzimologia , Proteína Quinase C/metabolismo , Transdução de Sinais/fisiologia , Animais , Animais Recém-Nascidos , Fator Natriurético Atrial/efeitos dos fármacos , Fator Natriurético Atrial/metabolismo , Cardiomiopatia Hipertrófica/patologia , Cardiomiopatia Hipertrófica/fisiopatologia , Compartimento Celular/efeitos dos fármacos , Compartimento Celular/fisiologia , Diferenciação Celular/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Citosol/efeitos dos fármacos , Citosol/metabolismo , Coração/efeitos dos fármacos , Coração/fisiologia , Imuno-Histoquímica , Isoenzimas/efeitos dos fármacos , MAP Quinase Quinase 1 , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Miocárdio/citologia , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C-alfa , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Tubulina (Proteína)/metabolismo
14.
Folia Med Cracov ; 50(3-4): 35-42, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-21853869

RESUMO

Vasopeptidase inhibitors (VPI) are a new promising class of drugs, that simultaneously inhibit Angiotensin - Converting Enzyme (ACE) and an enzyme Neutral Endopeptidase (NEP), that cleaves the natriuretic peptides. These drugs, such as omapatrilat, sampatrilat, fasidotrilat, by combined inhibition of ACE and degradation of natriuretic peptides and in turn by inhibiting the Renin - Angiotensin - Aldosterone system and potentiating the Natriuretic Peptide system and Kinin system should decrease the mortality rate in the group of patients with hypertension being not adequately controlled with ACE inhibitors. Thus, finding the new therapeutic strategy using drugs that act on the hormonal systems other than Renin - Angiotensin - Aldosterone system seems to be crucial. The aim of the study was to compare the molecular aspects of the conventional schemes that are being used in the antihypertension therapy to the new drugs from the vasopeptidase inhibitors group--with focusing on the natriuretic peptide system (NPS)--and, taking these considerations, making clues about therapeutical implications to reveal promising results in antihypertension treatment.


Assuntos
Anti-Hipertensivos/uso terapêutico , Hipertensão/tratamento farmacológico , Inibidores de Proteases/uso terapêutico , Sistema Renina-Angiotensina/efeitos dos fármacos , Resistência Vascular/efeitos dos fármacos , Alanina/análogos & derivados , Alanina/uso terapêutico , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Anti-Hipertensivos/farmacologia , Fator Natriurético Atrial/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Mesilatos/uso terapêutico , Inibidores de Proteases/farmacologia , Piridinas/uso terapêutico , Sistema Nervoso Simpático/efeitos dos fármacos , Tiazepinas/uso terapêutico , Tirosina/análogos & derivados , Tirosina/uso terapêutico
15.
Circulation ; 116(22): 2587-96, 2007 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-18025526

RESUMO

BACKGROUND: We have previously shown that deficiency for the z-disc protein calsarcin-1 (CS1) sensitizes the heart to calcineurin signaling and to stimuli of pathological hypertrophy. In the present study we asked whether overexpression of CS1 might exhibit antihypertrophic effects, and therefore we tested this hypothesis both in vitro and in vivo. METHODS AND RESULTS: Adenoviral gene transfer of CS1 into neonatal cardiomyocytes inhibited hypertrophy as a result of Gq-agonist stimulation, including angiotensin-II (Ang-II), endothelin-1, and phenylephrine. Consistently, Adenoviral gene transfer of CS1 also led to the reduction of increased levels of atrial natriuretic factor (mRNA) and the calcineurin-sensitive gene MCIP1.4, suggesting that CS1 inhibits calcineurin-dependent signaling. Furthermore, we generated CS1-overexpressing transgenic mice (CS1Tg). Unchallenged CS1Tg mice did not exhibit a pathological phenotype as assessed by echocardiography and analysis of cardiac gene expression. Likewise, when subjected to long-term infusion of Ang-II, both CS1Tg and wild-type mice developed a similar degree of arterial hypertension. Yet, in contrast to wild-type mice, Ang-II-treated CS1Tg animals did not display cardiac hypertrophy. Despite the absence of hypertrophy, both fractional shortening and dP/dt(max) were preserved in CS1Tg Ang-II-treated mice as assessed by echocardiography and cardiac catheterization, respectively. Moreover, induction of the hypertrophic gene program (atrial natriuretic factor, brain natriuretic peptide) was markedly blunted, and expression of the calcineurin-dependent gene MCIP1.4 was significantly reduced in CS1Tg mice, again consistent with an inhibitory role of CS1 on calcineurin. CONCLUSIONS: The sarcomeric protein CS1 prevents Ang-II-induced cardiomyocyte hypertrophy at least in part via inhibition of calcineurin signaling. Thus, overexpression of CS1 might represent a novel approach to attenuate pathological cardiac hypertrophy.


Assuntos
Angiotensina II/farmacologia , Calcineurina/metabolismo , Cardiomegalia/terapia , Proteínas de Transporte/farmacologia , Terapia Genética/métodos , Proteínas Musculares/farmacologia , Animais , Animais Recém-Nascidos , Fator Natriurético Atrial/efeitos dos fármacos , Fator Natriurético Atrial/genética , Proteínas de Ligação ao Cálcio , Cardiomegalia/etiologia , Proteínas de Transporte/administração & dosagem , Peptídeos e Proteínas de Sinalização Intracelular/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Transgênicos , Proteínas Musculares/administração & dosagem , Proteínas Musculares/efeitos dos fármacos , Proteínas Musculares/genética , RNA Mensageiro/análise , RNA Mensageiro/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
17.
Circulation ; 113(7): 977-85, 2006 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-16476851

RESUMO

BACKGROUND: The effect of beta-blockade on the cardiac natriuretic peptides is poorly understood but could contribute to their beneficial treatment effect and may be relevant to clinical use of plasma brain natriuretic peptide (BNP)/N-terminal pro brain natriuretic peptide (NTproBNP) measurements in risk stratification and in titration of anti-heart failure therapy. METHODS AND RESULTS: Sixteen men with mild, stable heart failure (NYHA class II to III; left ventricular ejection fraction <40%) underwent serial blood sampling for plasma natriuretic peptide levels and received infusions of atrial natriuretic peptide (ANP) and BNP before and 6 weeks after the introduction and uptitration of metoprolol or 6 weeks unchanged therapy in a randomized, parallel-group design. Plasma natriuretic peptides (BNP, NTproBNP, ANP, and NTproANP) were increased by metoprolol (P<0.01 for all). The natriuretic responses to ANP and BNP infusions were sustained with the introduction of metoprolol despite reduced renal perfusion pressure. The levels of the noninfused natriuretic peptide were increased by both ANP and BNP infusions, and this effect was enhanced by metoprolol. The early plasma half-life (t(1/2)alpha) of BNP was prolonged by metoprolol (5.6+/-0.45 to 11+/-1.3 versus 5.7+/-0.8 to 6.6+/-1.3 minutes in control subjects; P=0.019). CONCLUSIONS: Plasma cardiac natriuretic peptide levels increase significantly with the introduction of metoprolol in heart failure as a result of effects on secretion and clearance. Natriuretic responses to NP infusions are sustained with beta-blockade despite reduced renal perfusion pressure. Clinicians should be aware that the introduction of metoprolol causes a rise in plasma BNP/NTproBNP that is unrelated to deterioration in clinical status and must be considered when measurements are undertaken for risk stratification or titration of treatment.


Assuntos
Insuficiência Cardíaca/tratamento farmacológico , Metoprolol/farmacologia , Peptídeo Natriurético Encefálico/efeitos dos fármacos , Peptídeo Natriurético Encefálico/farmacocinética , Antagonistas Adrenérgicos beta/farmacologia , Idoso , Fator Natriurético Atrial/sangue , Fator Natriurético Atrial/efeitos dos fármacos , GMP Cíclico/urina , Insuficiência Cardíaca/sangue , Humanos , Masculino , Metoprolol/administração & dosagem , Pessoa de Meia-Idade , Peptídeo Natriurético Encefálico/administração & dosagem , Peptídeo Natriurético Encefálico/sangue , Fragmentos de Peptídeos/sangue , Fragmentos de Peptídeos/efeitos dos fármacos
18.
Neuroscience ; 147(1): 247-57, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17524563

RESUMO

The regulation of fluid and electrolyte homeostasis involves the participation of several neuropeptides and hormones that utilize hypothalamic cholinergic, alpha-adrenergic and angiotensinergic neurotransmitters and pathways. Additionally, it has been suggested that hypothalamus-pituitary-adrenal axis activity modulates hormonal responses to blood volume expansion. In the present study, we evaluated the effect of dexamethasone on atrial natriuretic peptide (ANP), oxytocin (OT) and vasopressin (AVP) responses to i.c.v. microinjections of 0.15 M and 0.30 M NaCl, angiotensin-II (ANG-II) and carbachol. We also evaluated the Fos protein immunoreactivity in the median preoptic (MnPO), paraventricular (PVN) and supraoptic (SON) nuclei. Male Wistar rats received an i.p. injection of dexamethasone (1 mg/kg) or vehicle (0.15 M NaCl) 2 h before the i.c.v. microinjections. Blood samples for plasma ANP, OT, AVP and corticosterone determinations were collected at 5 and 20 min after stimulus. Another set of rats was perfused 120 min after stimulation. A significant increase in plasma ANP, OT, AVP and corticosterone levels was observed at 5 and 20 min after each central stimulation compared with isotonic saline-injected group. Pre-treatment with dexamethasone decreased plasma corticosterone and OT levels, with no changes in the AVP secretion. On the other hand, dexamethasone induced a significant increase in plasma ANP levels. A significant increase in the number of Fos immunoreactive neurons was observed in the MnPO, PVN and SON after i.c.v. stimulations. Pre-treatment with dexamethasone induced a significant decrease in Fos immunoreactivity in these nuclei compared with the vehicle. These results indicate that central osmotic, cholinergic, and angiotensinergic stimuli activate MnPO, PVN and SON, with a subsequent OT, AVP, and ANP release. The present data also suggest that these responses are modulated by glucocorticoids.


Assuntos
Fator Natriurético Atrial/sangue , Hipotálamo/fisiologia , Ocitocina/sangue , Vasopressinas/sangue , Equilíbrio Hidroeletrolítico/fisiologia , Adaptação Fisiológica , Angiotensina II/fisiologia , Animais , Fator Natriurético Atrial/efeitos dos fármacos , Carbacol/farmacologia , Agonistas Colinérgicos/farmacologia , Corticosterona/sangue , Dexametasona/farmacologia , Glucocorticoides/farmacologia , Hipotálamo/citologia , Hipotálamo/efeitos dos fármacos , Injeções Intraventriculares , Masculino , Ocitocina/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Wistar , Estimulação Química , Vasopressinas/efeitos dos fármacos
19.
Regul Pept ; 140(3): 136-41, 2007 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-17223208

RESUMO

Atrial natriuretic peptide (ANP) is a hormone secreted in response to atrial or ventricular volume expansion and pressure overload, respectively. However, it has been found in studies with animals and patients an increase in ANP plasma concentration, during advanced septic shock, despite the fall in mean arterial pressure (MAP). Several studies support the hypothesis that NO may be involved in the regulation of ANP release. Since NO may have an effect on ANP release, we hypothesized that NO pathway may participate in the control of the ANP release induced by the endotoxemic shock. Thus, the purpose of the present study was to assess the effect of the intravenous (i.v.) and intracereboventricular (i.c.v.) administration of aminoguanidine, an iNOS blocker, on plasma ANP levels and MAP during experimental endotoxemic shock. Experiments were performed on adult male Wistar rats weighing 180-240 g. Rats were injected i.v. by bolus injection with 1.5 mg/kg of Lipopolysaccharide (LPS) or saline (0.5 mL) and were decapitated 2, 4 and 6 h after LPS injection for ANP determination by radioimmunoassay. In a separate set of experiments, rats received intravenous (i.v.) (100 mg/kg) or intracerebroventricular (i.c.v.) (250 microg in a final volume of 2 microL) injection of aminoguanidine (AG). Thirty minutes after the i.c.v. or i.v. injections, animals received LPS and were decapitated 2, 4 and 6 h later to determine plasma ANP concentration. In the two set of experiments MAP and heart rate (HR) were measured each 15 min for a period of 6 h using a polygraph. When animals were injected with LPS, a reduction (p<0.01) in MPA and an increase in HR occurred. A significant increase in plasma ANP concentration occurred, coinciding with the period of drop in blood pressure. We found a significant increase in plasma ANP concentration after AG plus LPS injection, when compared to the rats treated with LPS plus saline. Further, the administration of AG plus LPS attenuated the decrease in the MAP after LPS and attenuated the increase in the HR when compared to the rats treated with LPS plus saline. Our study suggests that inducible NOS pathway may activate an inhibitory control mechanism that attenuates ANP secretion, which is not regulated by the changes in blood pressure.


Assuntos
Fator Natriurético Atrial/sangue , Fator Natriurético Atrial/efeitos dos fármacos , Guanidinas/farmacologia , Óxido Nítrico Sintase Tipo II/efeitos dos fármacos , Choque Séptico/sangue , Animais , Inibidores Enzimáticos/farmacologia , Lipopolissacarídeos/farmacologia , Masculino , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Ratos , Ratos Wistar
20.
Life Sci ; 181: 45-52, 2017 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-28535942

RESUMO

AIMS: Gallic acid (GA) protects against myocardial ischemia-reperfusion (I/R) injury, prevents cardiac hypertrophy and fibrosis, and has anti-inflammatory activity in the heart. However, its effects in regulating atrial natriuretic peptide (ANP) secretion are unknown. The aim of this study was to determine the function of GA in regulating ANP secretion and atrial dynamics in rat atria. KEY FINDINGS: GA (0.01, 0.05, and 0.1µmol/L) significantly increased atrial ANP secretion and induced positive inotropy dose-dependently. GA (0.1µmol/L) also increased plasma level of ANP and hemodynamics in rats. These effects were accompanied by upregulation of atrial protein kinase C subtypes ß and ε (PKCß and PKCε), which was completely blocked by LY333531 and EAVSLKPT, antagonists of protein PKCß and PKCε, respectively. GA-induced ANP secretion was also attenuated by Gö6983 but not rottlerin, antagonists of PKCα and PKCδ, and the positive inotropy was reversed by Gö6983. U-73122, a phospholipase C (PLC) antagonist, mitigated the effects of GA on ANP secretion and mechanical dynamics and downregulated Phospho-PLCß at Ser537 (pPLCß S537), Phospho-PLCß at Ser1105 (pPLCß S1105), PKCß and PKCε levels, whereas KN62, an inhibitor of Ca2+/calmodulin-dependent kinase II, was not modified the GA-induced ANP secretion and suppressed GA-induced mechanical dynamics. SIGNIFICANCE: GA promotes ANP secretion and effects positive inotropy with regard to mechanical dynamics through the activation of PLC-PKC signaling in rat atria.


Assuntos
Fator Natriurético Atrial/efeitos dos fármacos , Ácido Gálico/farmacologia , Átrios do Coração/efeitos dos fármacos , Fosfolipases Tipo C/metabolismo , Animais , Fator Natriurético Atrial/sangue , Fator Natriurético Atrial/metabolismo , Relação Dose-Resposta a Droga , Feminino , Ácido Gálico/administração & dosagem , Átrios do Coração/metabolismo , Masculino , Proteína Quinase C beta/metabolismo , Proteína Quinase C-épsilon/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA