Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Cardiovasc Diabetol ; 9: 5, 2010 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-20105289

RESUMO

BACKGROUND: Diabetes is associated with changes in myocardial stress-response pathways and is recognized as an independent risk factor for cardiac remodeling. Using spontaneously diabetic Goto Kakizaki rats as a model of type 2 DM we investigated whether post-translational modifications in the Akt - FOXO3a pathway, Sirt1 - p53 pathway and the mitogen activated protein kinase p38 regulator are involved in post-infarct cardiac remodeling METHODS: Experimental myocardial infarction (MI) was induced by left anterior descending coronary artery ligation in spontaneously diabetic Goto-Kakizaki rats and non-diabetic Wistar controls. Cardiac function was studied by echocardiography. Myocardial hypertrophy, cardiomyocyte apoptosis and cardiac fibrosis were determined histologically 12 weeks post MI or Sham operation. Western blotting was used to study Caspase-3, Bax, Sirt1, acetylation of p53 and phosphorylation of p38, Akt and FOXO3a. Electrophoretic mobility shift assay was used to assess FOXO3a activity and its nuclear localization. RESULTS: Post-infarct heart failure in diabetic GK rats was associated with pronounced cardiomyocyte hypertrophy, increased interstitial fibrosis and sustained cardiomyocyte apoptosis as compared with their non-diabetic Wistar controls. In the GK rat myocardium, Akt- and FOXO3a-phosphorylation was decreased and nuclear localization of FOXO3a was increased concomitantly with increased PTEN protein expression. Furthermore, increased Sirt1 protein expression was associated with decreased p53 acetylation, and phosphorylation of p38 was increased in diabetic rats with MI. CONCLUSIONS: Post-infarct heart failure in diabetic GK rats was associated with more pronounced cardiac hypertrophy, interstitial fibrosis and sustained cardiomyocyte apoptosis as compared to their non-diabetic controls. The present study suggests important roles for Akt-FOXO3a, Sirt1 - p53 and p38 MAPK in the regulation of post-infarct cardiac remodeling in type 2 diabetes.


Assuntos
Complicações do Diabetes/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Infarto do Miocárdio/patologia , Sirtuína 1/metabolismo , Animais , Colágeno/metabolismo , Complicações do Diabetes/diagnóstico por imagem , Complicações do Diabetes/patologia , Complicações do Diabetes/fisiopatologia , Diástole/fisiologia , Ecocardiografia , Proteína Forkhead Box O3 , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/patologia , Processamento de Proteína Pós-Traducional , Ratos , Ratos Endogâmicos , Ratos Wistar , Sirtuína 1/genética , Volume Sistólico , Sístole/fisiologia
2.
Basic Clin Pharmacol Toxicol ; 98(1): 74-8, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16433895

RESUMO

In order to clarify the mechanisms of the positive inotropic actions of levosimendan and its optical isomer, dextrosimendan, we compared their concentration-dependent effects in intact papillary muscles, permeabilized cardiomyocytes and in purified phosphodiesterase enzyme preparations of guinea-pig hearts. In papillary muscles twitch tension increased with EC50 values of 60 nM and 2.8 microM for levosimendan and dextrosimendan, respectively. Hence, the two enantiomers exhibited a 47 times potency difference in their positive inotropic effects in a preparation where theoretically Ca2+-sensitization and phosphodiesterase inhibition could both contribute to the positive inotropic effects. In guinea-pig cardiomyocytes, levosimendan and dextrosimendan increased isometric force production (at pCa 6.2) due to Ca2+-sensitization with EC50 values of 8.4 nM and 0.64 microM, respectively, with a similar relative potency difference of 76. A major difference appeared in their relative pharmacological potencies, however, when the inhibitory effects of the two enantiomers were assayed on phosphodiesterase III, purified from guinea pig left ventricle (i.e. the phosphodiesterase isoenzyme which is dominant in that tissue). Levosimendan was a 427 times more potent phosphodiesterase inhibitor than dextrosimendan, with IC50 values of 7.5 nM, and 3.2 microM, respectively. Taken together, our data support the hypothesis that levosimendan and dextrosimendan exert their positive inotropic effects via a stereoselective Ca2+-sensitizing mechanism and not via stereoselective inhibition of phosphodiesterase III in the myocardium.


Assuntos
Cálcio/metabolismo , Cardiotônicos/farmacologia , Hidrazonas/farmacologia , Piridazinas/farmacologia , 3',5'-AMP Cíclico Fosfodiesterases/antagonistas & inibidores , 3',5'-AMP Cíclico Fosfodiesterases/metabolismo , Animais , Cardiotônicos/química , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3 , Relação Dose-Resposta a Droga , Cobaias , Ventrículos do Coração/citologia , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/enzimologia , Hidrazonas/química , Técnicas In Vitro , Isomerismo , Contração Miocárdica , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/enzimologia , Músculos Papilares/citologia , Músculos Papilares/efeitos dos fármacos , Músculos Papilares/enzimologia , Inibidores de Fosfodiesterase/química , Inibidores de Fosfodiesterase/farmacologia , Piridazinas/química , Simendana
3.
Eur J Pharmacol ; 486(1): 67-74, 2004 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-14751410

RESUMO

The concentration dependences of the Ca(2+)-sensitizing and the phosphodiesterase-inhibitory effects of levosimendan (the (-) enantiomer of [[4-(1,4,5,6-tetrahydro-4-methyl-6-oxo-3-pyridazinyl)phenyl]hydrazono]propanedinitrile) and its active metabolite, OR-1896 (the (-) enantiomer of N-[4-(1,4,5,6-tetrahydro-4-methyl-6-oxo-3-pyridazinyl)phenyl] acetamide), were compared with their positive inotropic effects to reveal their mechanisms of action in guinea pig hearts. In Langendorff-perfused hearts, left ventricular +dP/dt(max) increased by 26+/-4% and 25+/-3% (mean+/-S.E.M.), with EC(50) values of 15+/-2 and 25+/-1 nM for levosimendan and OR-1896, respectively. In permeabilized myocyte-sized preparations, levosimendan and OR-1896 both increased isometric force production via Ca(2+) sensitization (at pCa 6.2), by 51+/-7% and 52+/-6%, with EC(50) values of 8+/-1 and 36+/-7 nM (P<0.05), respectively. Thus, the two molecules could be defined as Ca(2+) sensitizers and positive inotropes with very similar concentration dependences. However, major differences appeared when the phosphodiesterase-inhibitory effects of levosimendan and OR-1896 were probed on the two phosphodiesterase isoforms (phosphodiesterases III and IV) dominant in the left ventricular cardiac tissue. Levosimendan was a 40-fold more potent and a 3-fold more selective phosphodiesterase III inhibitor (IC(50) for phosphodiesterase III=2.5 nM, and IC(50) for phosphodiesterase IV=25 microM, selectivity factor approximately 10000) than OR-1896 (IC(50) for phosphodiesterase III=94 nM, and IC(50) for phosphodiesterase IV=286 microM, selectivity factor approximately 3000). Hence, our data support the hypothesis that levosimendan and OR-1896 both exert positive inotropy via a Ca(2+)-sensitizing mechanism and not via simultaneous inhibition of the phosphodiesterases III and IV isozymes in the myocardium at their maximal free plasma concentrations.


Assuntos
Acetamidas/farmacologia , Ventrículos do Coração/efeitos dos fármacos , Hidrazonas/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Diester Fosfórico Hidrolases/metabolismo , Piridazinas/farmacologia , Acetamidas/química , Animais , Separação Celular , Relação Dose-Resposta a Droga , Feminino , Cobaias , Ventrículos do Coração/enzimologia , Hidrazonas/química , Técnicas In Vitro , Isoenzimas/metabolismo , Masculino , Miócitos Cardíacos/enzimologia , Inibidores de Fosfodiesterase/farmacologia , Piridazinas/química , Simendana , Função Ventricular
4.
Basic Clin Pharmacol Toxicol ; 109(5): 387-97, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21699655

RESUMO

The calcium sensitizer levosimendan has shown beneficial effects on cardiac remodelling in spontaneously diabetic Goto-Kakizaki (GK) rats 12 weeks after experimental myocardial infarction (MI). However, the short-term effects and the cellular mechanisms remain partially unresolved. The aim was to study the effects of oral levosimendan treatment on the myocardial gene expression profile in diabetic GK rats 4 weeks after MI/sham operation. MI was induced to diabetic GK rats. Twenty-four hours after surgery, rats were randomized into four groups: MI, MI +levosimendan (1 mg/kg/day), sham-operated and sham-operated +levosimendan. Cardiac function and histology were examined 1, 4 and 12 weeks after MI. The effects of levosimendan on cardiac gene expression profile were investigated by microarray analysis. Levosimendan ameliorated post-infarct heart failure and cardiac remodelling. Levosimendan altered the expression of 264 of MI and sham rats, respectively; these changes were associated with alterations in two Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Levosimendan up-regulated 3 genes in the renin-angiotensin system pathway [angiotensin receptor 1 (Agtr1), chymase 1 (Cma1) and thimet oligopeptidase 1 (Thop1)] and down-regulated 3 genes in the glycerolipid metabolism pathway [diacylglycerol kinase gamma (Dgkg), carboxyl ester lipase (Cel) and Diacylglycerol kinase iota]. Levosimendan induced opposite effects on the gene expression of pleckstrin homology (PH) domain containing family f (Plekhf1), carboxymethylenebutenolidase homologue (Cmbl) (up-regulation) and hydroxyprostaglandin dehydrogenase 15 (Hpgd) (down-regulation) as compared with MI. MI versus sham affected 420 genes and was associated with alterations in 12 KEGG pathways. The beneficial effects of levosimendan on cardiac hypertrophy in sham-operated GK rats was associated with altered expression in 522 genes and associated with three KEGG pathways including purine metabolism, cell cycle pathway and pathways in cancer. Levosimendan protects against post-infarct heart failure and cardiac remodelling. Analysis of the cardiac transcriptome revealed several genes that are regulated by levosimendan. These genes may represent novel drug targets for heart failure and diabetic cardiomyopathy.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Perfilação da Expressão Gênica/métodos , Coração/fisiopatologia , Hidrazonas/farmacologia , Piridazinas/farmacologia , Remodelação Ventricular/efeitos dos fármacos , Animais , Regulação para Baixo , Coração/efeitos dos fármacos , Insuficiência Cardíaca/tratamento farmacológico , Imuno-Histoquímica , Análise em Microsséries , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , Ratos , Ratos Wistar , Sistema Renina-Angiotensina , Simendana , Transcriptoma , Regulação para Cima
5.
Br J Pharmacol ; 160(1): 142-52, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20412071

RESUMO

BACKGROUND AND PURPOSE: Levosimendan is a novel, short half-life calcium sensitizer used as pharmacological inotropic support in acute decompensated heart failure. After oral administration, levosimendan is metabolized to OR-1855, which, in rats, is further metabolized into OR-1896. OR-1896 is a long-lasting metabolite of levosimendan sharing the pharmacological properties of the parent compound. EXPERIMENTAL APPROACH: Effects of oral OR-1896 treatment on post-infarct heart failure and cardiac remodelling were assessed in diabetic Goto-Kakizaki (GK) rats, an animal model of type II diabetes. Myocardial infarction (MI) was produced to GK rats by coronary ligation. Twenty-four hours after MI or sham operation, the rats were randomized into four groups: (i) MI; (ii) MI + OR-1896 treatment; (iii) sham; and (iv) sham + OR-1896. Cardiac function and markers of cardiac remodelling were assessed 1, 4 and 12 weeks after MI. KEY RESULTS: OR-1896 increased ejection fraction and fractional shortening in GK rats with MI. OR-1896 ameliorated post-infarct cardiac hypertrophy, and prevented the MI-induced increase in cardiac mRNA for atrial natriuretic peptide, monocyte chemoattractant protein-1 and connective tissue growth factor, markers of pressure/volume overload, inflammation and fibrosis respectively. OR-1896 also suppressed mRNA for senescence-associated p16(INK4A) and p19(ARF). The beneficial effects of OR-1896 were more prominent at week 12 than at week 4. OR-1896 did not influence systolic blood pressure, blood glucose level, myocardial infarct size or cardiovascular mortality. CONCLUSIONS AND IMPLICATIONS: Oral treatment with calcium sensitizer OR-1896 protects against post-infarct heart failure and cardiac remodelling in experimental model of type II diabetes.


Assuntos
Acetamidas/farmacologia , Diabetes Mellitus Tipo 2/tratamento farmacológico , Insuficiência Cardíaca/tratamento farmacológico , Infarto do Miocárdio/etiologia , Piridazinas/farmacologia , Acetamidas/uso terapêutico , Animais , Biomarcadores/metabolismo , Volume Cardíaco/efeitos dos fármacos , Senescência Celular , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/fisiopatologia , Fibrose , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Inflamação/metabolismo , Mitocôndrias Cardíacas/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Piridazinas/uso terapêutico , Distribuição Aleatória , Ratos , Tetrazóis/uso terapêutico , Fatores de Tempo , Valina/análogos & derivados , Valina/uso terapêutico , Valsartana , Vasodilatadores/uso terapêutico , Pressão Ventricular/efeitos dos fármacos
6.
Hypertens Res ; 33(10): 1004-11, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20811386

RESUMO

Calcium-sensitizing agents improve cardiac function in acute heart failure; however, their long-term effects on cardiovascular mortality are unknown. We tested the hypothesis that levosimendan, an inodilator that acts through calcium sensitization, opening of ATP-dependent potassium channels and phosphodiesterase III inhibition, improves cardiac function and survival in double transgenic rats harboring human renin and angiotensinogen genes (dTGRs), a model of angiotensin II (Ang II)-induced hypertensive heart failure. Levosimendan (1 mg kg(-1)) was administered orally to 4-week-old dTGRs and normotensive Sprague-Dawley rats for 4 weeks. Untreated dTGRs developed severe hypertension, cardiac hypertrophy, heart failure with impaired diastolic relaxation, and exhibited a high mortality rate at the age of 8 weeks. Levosimendan did not decrease blood pressure and did not prevent cardiac hypertrophy. However, levosimendan improved systolic function, decreased cardiac atrial natriuretic peptide mRNA expression, ameliorated Ang II-induced cardiac damage and decreased mortality. Levosimendan did not correct Ang II-induced diastolic dysfunction and did not influence heart rate. In a separate survival study, levosimendan increased dTGR survival by 58% and median survival time by 27% (P=0.004). Our findings suggest that levosimendan ameliorates Ang II-induced hypertensive heart failure and reduces mortality. The results also support the notion that the effects of levosimendan in dTGRs are mediated by blood pressure-independent mechanisms and include improved systolic function and amelioration of Ang II-induced coronary and cardiomyocyte damage.


Assuntos
Angiotensina II/efeitos adversos , Insuficiência Cardíaca/tratamento farmacológico , Coração/fisiologia , Hidrazonas/uso terapêutico , Hipertensão/tratamento farmacológico , Piridazinas/uso terapêutico , Angiotensina II/metabolismo , Angiotensinogênio/genética , Angiotensinogênio/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Cardiotônicos/farmacologia , Cardiotônicos/uso terapêutico , Modelos Animais de Doenças , Coração/efeitos dos fármacos , Insuficiência Cardíaca/fisiopatologia , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Humanos , Hidrazonas/farmacologia , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Complexo Principal de Histocompatibilidade/fisiologia , Masculino , Piridazinas/farmacologia , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Renina/genética , Renina/metabolismo , Sistema Renina-Angiotensina/efeitos dos fármacos , Sistema Renina-Angiotensina/fisiologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Simendana , Proteína X Associada a bcl-2/metabolismo
7.
Hypertension ; 55(2): 508-15, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20065148

RESUMO

Angiotensin II (Ang II) induces mitochondrial dysfunction. We tested whether Ang II alters the "metabolomic" profile. We harvested hearts from 8-week-old double transgenic rats harboring human renin and angiotensinogen genes (dTGRs) and controls (Sprague-Dawley), all with or without Ang II type 1 receptor (valsartan) blockade. We used gas chromatography coupled with time-of-flight mass spectrometry to detect 247 intermediary metabolites. We used a partial least-squares discriminate analysis and identified 112 metabolites that differed significantly after corrections (false discovery rate q <0.05). We found great differences in the use of fatty acids as an energy source, namely, decreased levels of octanoic, oleic, and linoleic acids in dTGR (all P<0.01). The increase in cardiac hypoxanthine levels in dTGRs suggested an increase in purine degradation, whereas other changes supported an increased ketogenic amino acid tyrosine level, causing energy production failure. The metabolomic profile of valsartan-treated dTGRs more closely resembled Sprague-Dawley rats than untreated dTGRs. Mitochondrial respiratory chain activity of cytochrome C oxidase was decreased in dTGRs, whereas complex I and complex II were unaltered. Mitochondria from dTGR hearts showed morphological alterations suggesting increased mitochondrial fusion. Cardiac expression of the redox-sensitive and the cardioprotective metabolic sensor sirtuin 1 was increased in dTGRs. Interestingly, valsartan changed the level of 33 metabolites and induced mitochondrial biogenesis in Sprague-Dawley rats. Thus, distinct patterns of cardiac substrate use in Ang II-induced cardiac hypertrophy are associated with mitochondrial dysfunction. The finding underscores the importance of Ang II in the regulation of mitochondrial biogenesis and cardiac metabolomics, even in healthy hearts.


Assuntos
Angiotensina II/farmacologia , Cardiomegalia/metabolismo , Mitocôndrias Cardíacas/patologia , Sirtuína 1/metabolismo , Tetrazóis/farmacologia , Valina/análogos & derivados , Animais , Animais Geneticamente Modificados , Biomarcadores/análise , Cardiomegalia/induzido quimicamente , Cardiomegalia/mortalidade , Cardiomegalia/patologia , Modelos Animais de Doenças , Ácidos Graxos/metabolismo , Feminino , Humanos , Hipoxantina/metabolismo , Ácidos Linoleicos/metabolismo , Masculino , Metabolômica/métodos , Estresse Oxidativo/fisiologia , Probabilidade , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Valores de Referência , Taxa de Sobrevida , Valina/farmacologia , Valsartana
8.
J Hypertens ; 27(10): 2094-107, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19730126

RESUMO

BACKGROUND: Diabetes increases the risk for fatal myocardial infarction and development of heart failure. Levosimendan, an inodilator acting both via calcium sensitization and opening of ATP-dependent potassium channels, is used intravenously for acute decompensated heart failure. The long-term effects of oral levosimendan on postinfarct heart failure are largely unknown. OBJECTIVE: To examine whether oral treatment with levosimendan could improve cardiac functions and prevent cardiac remodeling after myocardial infarction in a rodent model of type 2 diabetes, the Goto-Kakizaki rat. METHODS: Myocardial infarction (MI) was induced to diabetic Goto-Kakizaki and nondiabetic Wistar rats by coronary ligation. Twenty-four hours after surgery, Goto-Kakizaki and Wistar rats were randomized into four groups: MI group without treatment, MI group with levosimendan for 12 weeks (1 mg/kg per day), sham-operated group, sham-operated group with levosimendan. Blood pressure, cardiac functions as wells as markers of cardiac remodeling were determined. RESULTS: In Goto-Kakizaki rats, MI induced systolic heart failure, pronounced cardiac hypertrophy in the remote area, and sustained cardiomyocyte apoptosis. Postinfarct cardiac remodeling was associated with increased atrial natriuretic peptide, interleukin-6 and connective tissue growth factor mRNA expressions, as well as three-fold increased cardiomyocyte senescence, measured as cardiac p16 mRNA expression. Levosimendan improved cardiac function and prevented postinfarct cardiomyocyte hypertrophy, cardiomyocyte apoptosis, and cellular senescence. Levosimendan also ameliorated MI-induced atrial natriuretic peptide, IL-6, and connective tissue growth factor overexpression as well as MI-induced disturbances in calcium-handling proteins (SERCA2, Na-Ca exchanger) without changes in diabetic status or systemic blood pressure. In nondiabetic Wistar rats, MI induced systolic heart failure; however, the postinfarct cardiac remodeling was associated with less pronounced cardiac hypertrophy, cardiomyocyte apoptosis, inflammatory reaction, and induction of cellular senescence. Levosimendan only partially prevented postinfarct heart failure and cardiac remodeling in Wistar rats. CONCLUSION: Our findings suggest a therapeutic role for oral levosimendan in prevention of postinfarct heart failure and cardiac remodeling in type 2 diabetes and underscore the importance of sustained cardiomyocyte apoptosis and induction of cellular senescence in the pathogenesis.


Assuntos
Cardiotônicos/farmacologia , Complicações do Diabetes/prevenção & controle , Insuficiência Cardíaca/prevenção & controle , Hidrazonas/farmacologia , Infarto do Miocárdio/tratamento farmacológico , Piridazinas/farmacologia , Remodelação Ventricular/efeitos dos fármacos , Administração Oral , Animais , Apoptose/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Cardiomegalia/etiologia , Cardiomegalia/patologia , Cardiomegalia/prevenção & controle , Inibidor p16 de Quinase Dependente de Ciclina/genética , Complicações do Diabetes/patologia , Diabetes Mellitus Tipo 2/complicações , Modelos Animais de Doenças , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/patologia , Proteínas de Homeodomínio/metabolismo , Masculino , Infarto do Miocárdio/complicações , Infarto do Miocárdio/patologia , RNA Mensageiro/metabolismo , Ratos , Ratos Mutantes , Ratos Wistar , Fator de Transcrição STAT1/efeitos dos fármacos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Simendana
9.
J Cardiovasc Pharmacol ; 43(4): 555-61, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15085067

RESUMO

Levosimendan is a novel calcium sensitizer that increases contraction force without change in intracellular calcium ([Ca2+]i); milrinone is a phosphodiesterase inhibitor that exerts a positive inotropic effect by increasing [Ca2+]i. The effects of levosimendan and milrinone on oxygen consumption in the isolated guinea-pig heart were studied. Isolated guinea-pig hearts were paced (280 beats/min) and perfused according to the Langendorff technique. Levosimendan (0.01-1 microM) or milrinone (0.1-10 microM) were added cumulatively and changes from baseline for diastolic and systolic pressure (LVEDP and LVSP), contractility and relaxation (+dP/dt and -dP/dt), and coronary flow and oxygen consumption (CF and VO2) were calculated. Levosimendan was found to be 10 to 30 times more potent than milrinone as an inotropic agent. The effect on VO2 was markedly lower in levosimendan-perfused hearts than in milrinone-perfused hearts (P = 0.031 between the concentration-dependent effects of the two drugs). The maximum increase in VO2 was 10 +/- 4% in the levosimendan group and 38 +/- 15% in the milrinone group. The economy of the contraction was more advantageous in levosimendan-perfused hearts (P

Assuntos
Coração/efeitos dos fármacos , Hidrazonas/farmacologia , Milrinona/farmacologia , Consumo de Oxigênio/efeitos dos fármacos , Piridazinas/farmacologia , Animais , Relação Dose-Resposta a Droga , Cobaias , Coração/fisiologia , Técnicas In Vitro , Contração Miocárdica/efeitos dos fármacos , Contração Miocárdica/fisiologia , Consumo de Oxigênio/fisiologia , Simendana
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA