Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Cell Mol Med ; 21(12): 3277-3287, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28608983

RESUMO

The use of doxorubicin (DOXO) as a chemotherapeutic drug has been hampered by cardiotoxicity leading to cardiomyopathy and heart failure. Folic acid (FA) is a modulator of endothelial nitric oxide (NO) synthase (eNOS), which in turn is an important player in diseases associated with NO insufficiency or NOS dysregulation, such as pressure overload and myocardial infarction. However, the role of FA in DOXO-induced cardiomyopathy is poorly understood. The aim of this study was to test the hypothesis that FA prevents DOXO-induced cardiomyopathy by modulating eNOS and mitochondrial structure and function. Male C57BL/6 mice were randomized to a single dose of DOXO (20 mg/kg intraperitoneal) or sham. FA supplementation (10 mg/day per oral) was started 7 days before DOXO injection and continued thereafter. DOXO resulted in 70% mortality after 10 days, with the surviving mice demonstrating a 30% reduction in stroke volume compared with sham groups. Pre-treatment with FA reduced mortality to 45% and improved stroke volume (both P < 0.05 versus DOXO). These effects of FA were underlain by blunting of DOXO-induced cardiomyocyte atrophy, apoptosis, interstitial fibrosis and impairment of mitochondrial function. Mechanistically, pre-treatment with FA prevented DOXO-induced increases in superoxide anion production by reducing the eNOS monomer:dimer ratio and eNOS S-glutathionylation, and attenuated DOXO-induced decreases in superoxide dismutase, eNOS phosphorylation and NO production. Enhancing eNOS function by restoring its coupling and subsequently reducing oxidative stress with FA may be a novel therapeutic approach to attenuate DOXO-induced cardiomyopathy.


Assuntos
Antioxidantes/farmacologia , Cardiomiopatias/prevenção & controle , Cardiotônicos/farmacologia , Cardiotoxicidade/prevenção & controle , Doxorrubicina/antagonistas & inibidores , Doxorrubicina/toxicidade , Ácido Fólico/farmacologia , Animais , Antibióticos Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/enzimologia , Cardiomiopatias/mortalidade , Cardiotoxicidade/enzimologia , Cardiotoxicidade/mortalidade , Cardiotoxicidade/patologia , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Mitocôndrias/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosforilação , Volume Sistólico/efeitos dos fármacos , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxidos/antagonistas & inibidores , Superóxidos/metabolismo , Análise de Sobrevida
2.
J Mol Cell Cardiol ; 88: 145-54, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26436984

RESUMO

Nitric oxide (NO) produced by endothelial NO synthase (eNOS) exerts beneficial effects in a variety of cardiovascular disease states. Studies on the benefit of eNOS activity in pressure-overload cardiac hypertrophy and dysfunction produced by aortic stenosis are equivocal, which may be due to different expression levels of eNOS or different severities of pressure-overload. Consequently, we investigated the effects of eNOS-expression level on cardiac hypertrophy and dysfunction produced by mild or severe pressure-overload. To unravel the impact of eNOS on pressure-overload cardiac dysfunction we subjected eNOS deficient, wildtype and eNOS overexpressing transgenic (eNOS-Tg) mice to 8weeks of mild or severe transverse aortic constriction (TAC) and studied cardiac geometry and function at the whole organ and tissue level. In both mild and severe TAC, lack of eNOS ameliorated, whereas eNOS overexpression aggravated, TAC-induced cardiac remodeling and dysfunction. Moreover, the detrimental effects of eNOS in severe TAC were associated with aggravation of TAC-induced NOS-dependent oxidative stress and by further elevation of eNOS monomer levels, consistent with enhanced eNOS uncoupling. In the presence of TAC, scavenging of reactive oxygen species with N-acetylcysteine reduced eNOS S-glutathionylation, eNOS monomer and NOS-dependent superoxide levels in eNOS-Tg mice to wildtype levels. Accordingly, N-acetylcysteine improved cardiac function in eNOS-Tg but not in wildtype mice with TAC. In conclusion, independent of the severity of TAC, eNOS aggravates cardiac remodeling and dysfunction, which appears due to TAC-induced eNOS uncoupling and superoxide production.


Assuntos
Cardiomegalia/enzimologia , Cardiomegalia/genética , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico/metabolismo , Remodelação Ventricular , Acetilcisteína/farmacologia , Animais , Aorta/cirurgia , Cardiomegalia/etiologia , Cardiomegalia/patologia , Constrição Patológica/complicações , Constrição Patológica/cirurgia , Ativação Enzimática , Feminino , Sequestradores de Radicais Livres/farmacologia , Deleção de Genes , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo III/metabolismo , Estresse Oxidativo , Índice de Gravidade de Doença , Superóxidos/antagonistas & inibidores , Superóxidos/metabolismo
3.
Circ Res ; 109(12): 1410-4, 2011 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-22034490

RESUMO

RATIONALE: One of the physiological mechanisms by which the heart adapts to a rise in blood pressure is by augmenting myocyte stretch-mediated intracellular calcium, with a subsequent increase in contractility. This slow force response was first described over a century ago and has long been considered compensatory, but its underlying mechanisms and link to chronic adaptations remain uncertain. Because levels of the matricellular protein thrombospondin-4 (TSP4) rapidly rise in hypertension and are elevated in cardiac stress overload and heart failure, we hypothesized that TSP4 is involved in this adaptive mechanism. OBJECTIVE: To determine the mechano-transductive role that TSP4 plays in cardiac regulation to stress. METHODS AND RESULTS: In mice lacking TSP4 (Tsp4⁻/⁻), hearts failed to acutely augment contractility or activate stretch-response pathways (ERK1/2 and Akt) on exposure to acute pressure overload. Sustained pressure overload rapidly led to greater chamber dilation, reduced function, and increased heart mass. Unlike controls, Tsp4⁻/⁻ cardiac trabeculae failed to enhance contractility and cellular calcium after a stretch. However, the contractility response was restored in Tsp4⁻/⁻ muscle incubated with recombinant TSP4. Isolated Tsp4⁻/⁻ myocytes responded normally to stretch, identifying a key role of matrix-myocyte interaction for TSP4 contractile modulation. CONCLUSION: These results identify TSP4 as myocyte-interstitial mechano-signaling molecule central to adaptive cardiac contractile responses to acute stress, which appears to play a crucial role in the transition to chronic cardiac dilatation and failure.


Assuntos
Contração Miocárdica/fisiologia , Miócitos Cardíacos/fisiologia , Estresse Fisiológico/fisiologia , Trombospondinas/fisiologia , Animais , Insuficiência Cardíaca/fisiopatologia , Hipertensão/fisiopatologia , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Miócitos Cardíacos/citologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Ratos , Trombospondinas/deficiência , Trombospondinas/genética
4.
J Mol Cell Cardiol ; 52(6): 1213-25, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22465037

RESUMO

The utility of anthracycline antineoplastic agents in the clinic is compromised by the risk of cardiotoxicity. It has been calculated that approximately 10% of patients treated with doxorubicin or its derivatives will develop cardiac complications up to 10 years after the cessation of chemotherapy. Oxidative stress has been established as the primary cause of cardiotoxicity. However, interventions reducing oxidative stress have not been successful at reducing the incidence of cardiotoxicity in patients treated with doxorubicin. New insights into the cardiomyocyte response to oxidative stress demonstrate that underlying differences between in vitro and in vivo toxicities may modulate the response to superoxide radicals and related compounds. This has led to potentially new uses for pre-existing drugs and new avenues of exploration to find better pharmacotherapies and interventions for the prevention of cardiotoxicity. However, much work still must be done to validate the clinical utility of these new approaches and proposed mechanisms. In this review, the authors have reviewed the molecular mechanisms of the pathogenesis of acute and chronic doxorubicin-induced cardiotoxicity and propose potential pharmacological interventions and treatment options to prevent or reverse this specific type of heart failure.


Assuntos
Antibióticos Antineoplásicos/efeitos adversos , Cardiomiopatias/induzido quimicamente , Doxorrubicina/efeitos adversos , Animais , Cardiomiopatias/diagnóstico , Cardiomiopatias/metabolismo , Cardiomiopatias/terapia , Humanos
5.
Am J Physiol Endocrinol Metab ; 302(5): E481-95, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22167522

RESUMO

Endothelial nitric oxide synthase (eNOS) serves as a critical enzyme in maintaining vascular pressure by producing nitric oxide (NO); hence, it has a crucial role in the regulation of endothelial function. The bioavailability of eNOS-derived NO is crucial for this function and might be affected at multiple levels. Uncoupling of eNOS, with subsequently less NO and more superoxide generation, is one of the major underlying causes of endothelial dysfunction found in atherosclerosis, diabetes, hypertension, cigarette smoking, hyperhomocysteinemia, and ischemia/reperfusion injury. Therefore, modulating eNOS uncoupling by stabilizing eNOS activity, enhancing its substrate, cofactors, and transcription, and reversing uncoupled eNOS are attractive therapeutic approaches to improve endothelial function. This review provides an extensive overview of the important role of eNOS uncoupling in the pathogenesis of endothelial dysfunction and the potential therapeutic interventions to modulate eNOS for tackling endothelial dysfunction.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico/metabolismo , Doenças Vasculares/tratamento farmacológico , Doenças Vasculares/metabolismo , Animais , Endotélio Vascular/fisiopatologia , Indução Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Estabilidade Enzimática/efeitos dos fármacos , Humanos , Óxido Nítrico/agonistas , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/química , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Células-Tronco/efeitos dos fármacos , Células-Tronco/enzimologia , Células-Tronco/metabolismo , Doenças Vasculares/diagnóstico , Doenças Vasculares/fisiopatologia
6.
J Mol Cell Cardiol ; 51(4): 559-63, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21458460

RESUMO

Tetrahydrobiopterin (BH(4)) is an essential cofactor for aromatic amino acid hydroxylases and for all three nitric oxide synthase (NOS) isoforms. It also has a protective role in the cell as an antioxidant and scavenger of reactive nitrogen and oxygen species. Experimental studies in humans and animals demonstrate that decreased BH(4)-bioavailability, with subsequent uncoupling of endothelial NOS (eNOS) plays an important role in the pathogenesis of endothelial dysfunction, hypertension, ischemia-reperfusion injury, and pathologic cardiac remodeling. Synthetic BH(4) is clinically approved for the treatment of phenylketonuria, and experimental studies support its capacity for ameliorating cardiovascular pathophysiologies. To date, however, the translation of these studies to human patients remains limited, and early results have been mixed. In this review, we discuss the pathophysiologic role of decreased BH(4) bioavailability, molecular mechanisms regulating its metabolism, and its potential therapeutic use as well as pitfalls as an NOS-modulating drug. This article is part of a special issue entitled ''Key Signaling Molecules in Hypertrophy and Heart Failure.''


Assuntos
Antioxidantes/uso terapêutico , Biopterinas/análogos & derivados , Endotélio Vascular/fisiopatologia , Coração/fisiopatologia , Animais , Biopterinas/biossíntese , Biopterinas/uso terapêutico , Doenças Cardiovasculares/tratamento farmacológico , Endotélio Vascular/efeitos dos fármacos , Coração/efeitos dos fármacos , Humanos , Isoenzimas/metabolismo , Terapia de Alvo Molecular , Miocárdio/enzimologia , Óxido Nítrico Sintase/metabolismo
7.
J Mol Cell Cardiol ; 51(4): 564-9, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21645517

RESUMO

The exogenous administration of tetrahydrobiopterin (BH4), an essential cofactor of nitric oxide synthase (NOS), has been shown to reduce left ventricular hypertrophy, fibrosis, and cardiac dysfunction in mice with pre-established heart disease induced by pressure-overload. In this setting, BH4 re-coupled endothelial NOS (eNOS), with subsequent reduction of NOS-dependent oxidative stress and reversal of maladaptive remodeling. However, recent studies suggest the effective BH4 dosing may be narrower than previously thought, potentially due to its oxidation upon oral consumption. Accordingly, we assessed the dose response of daily oral synthetic sapropterin dihydrochloride (6-R-l-erythro-5,6,7,8-tetrahydrobiopterin, 6R-BH4) on pre-established pressure-overload cardiac disease. Mice (n=64) were administered 0-400mg/kg/d BH4 by ingesting small pre-made pellets (consumed over 15-30 min). In a dose range of 36-200mg/kg/d, 6R-BH4 suppressed cardiac chamber remodeling, hypertrophy, fibrosis, and oxidative stress with pressure-overload. However, at both lower and higher doses, BH4 had less or no ameliorative effects. The effective doses correlated with a higher myocardial BH4/BH2 ratio. However, BH2 rose linearly with dose, and at the 400mg/kg/d, this lowered the BH4/BH2 ratio back toward control. These results expose a potential limitation for the clinical use of BH4, as variability of cellular redox and perhaps heart disease could produce a variable therapeutic window among individuals. This article is part of a special issue entitled ''Key Signaling Molecules in Hypertrophy and Heart Failure.''


Assuntos
Biopterinas/análogos & derivados , Cardiotônicos/uso terapêutico , Insuficiência Cardíaca/tratamento farmacológico , Hipertrofia Ventricular Esquerda/tratamento farmacológico , Remodelação Ventricular/efeitos dos fármacos , Análise de Variância , Animais , Biopterinas/metabolismo , Biopterinas/farmacocinética , Biopterinas/uso terapêutico , Cardiotônicos/farmacocinética , Relação Dose-Resposta a Droga , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Hipertrofia Ventricular Esquerda/etiologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Ligadura , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/patologia , Distribuição Aleatória , Superóxidos/metabolismo , Função Ventricular Esquerda
8.
Am J Physiol Heart Circ Physiol ; 301(3): H634-46, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21622818

RESUMO

The pathogenesis of many cardiovascular diseases is associated with reduced nitric oxide (NO) bioavailability and/or increased endothelial NO synthase (eNOS)-dependent superoxide formation. These findings support that restoring and conserving adequate NO signaling in the heart and blood vessels is a promising therapeutic intervention. In particular, modulating eNOS, e.g., through increasing the bioavailability of its substrate and cofactors, enhancing its transcription, and interfering with other modulators of eNOS pathway, such as netrin-1, has a high potential for effective treatments of cardiovascular diseases. This review provides an overview of the possibilities for modulating eNOS and how this may be translated to the clinic in addition to describing the genetic models used to study eNOS modulation.


Assuntos
Fármacos Cardiovasculares/uso terapêutico , Doenças Cardiovasculares/tratamento farmacológico , Endotélio Vascular/efeitos dos fármacos , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico/metabolismo , Animais , Doenças Cardiovasculares/enzimologia , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/fisiopatologia , Endotélio Vascular/enzimologia , Endotélio Vascular/fisiopatologia , Inibidores Enzimáticos/uso terapêutico , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Doadores de Óxido Nítrico/uso terapêutico , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/genética , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
9.
J Mol Cell Cardiol ; 48(6): 1088-95, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20184889

RESUMO

The presence of a third beta-adrenergic receptor (beta 3-AR) in the cardiovascular system has challenged the classical paradigm of sympathetic regulation by beta1- and beta2-adrenergic receptors. While beta 3-AR's role in the cardiovascular system remains controversial, increasing evidence suggests that it serves as a "brake" in sympathetic overstimulation - it is activated at high catecholamine concentrations, producing a negative inotropic effect that antagonizes beta1- and beta2-AR activity. The anti-adrenergic effects induced by beta 3-AR were initially linked to nitric oxide (NO) release via endothelial NO synthase (eNOS), although more recently it has been shown under some conditions to increase NO production in the cardiovascular system via the other two NOS isoforms, namely inducible NOS (iNOS) and neuronal NOS (nNOS). We summarize recent findings regarding beta 3-AR effects on the cardiovascular system and explore its prospective as a therapeutic target, particularly focusing on its emerging role as an important mediator of NO signaling in the pathogenesis of cardiovascular disorders.


Assuntos
Doenças Cardiovasculares/metabolismo , Regulação da Expressão Gênica , Receptores Adrenérgicos beta 3/metabolismo , Animais , Coração/fisiologia , Insuficiência Cardíaca/metabolismo , Humanos , Camundongos , Modelos Biológicos , Miocárdio/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Estresse Oxidativo , Transdução de Sinais
10.
Am J Physiol Heart Circ Physiol ; 299(5): H1283-99, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20833966

RESUMO

Lung ischemia-reperfusion injury remains one of the major complications after cardiac bypass surgery and lung transplantation. Due to its dual blood supply system and the availability of oxygen from alveolar ventilation, the pathogenetic mechanisms of ischemia-reperfusion injury in the lungs are more complicated than in other organs, where loss of blood flow automatically leads to hypoxia. In this review, an extensive overview is given of the molecular and cellular mechanisms that are involved in the pathogenesis of lung ischemia-reperfusion injury and the possible therapeutic strategies to reduce or prevent it. In addition, the roles of neutrophils, alveolar macrophages, cytokines, and chemokines, as well as the alterations in the cell-death related pathways, are described in detail.


Assuntos
Pneumopatias/fisiopatologia , Traumatismo por Reperfusão/fisiopatologia , Animais , Ponte Cardiopulmonar/efeitos adversos , Humanos , Pneumopatias/prevenção & controle , Pneumopatias/terapia , Transplante de Pulmão/efeitos adversos , Modelos Animais , Fluxo Sanguíneo Regional , Traumatismo por Reperfusão/prevenção & controle , Traumatismo por Reperfusão/terapia
11.
J Mol Cell Cardiol ; 47(5): 576-85, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19766235

RESUMO

Stimulation of the beta-adrenergic system is important in the pathological response to sustained cardiac stress, forming the rationale for the use of beta-blockers in heart failure. The beta3-adrenoreceptor (AR) is thought to couple to the inhibitory G-protein, G(i), with downstream signaling through nitric oxide, although its role in the heart remains controversial. In this study, we tested whether lack of beta3-AR influences the myocardial response to pressure-overload. Baseline echocardiography in mice lacking beta3-AR (beta3(-/-)) compared to wild type (WT) showed mild LV hypertrophy at 8 weeks that worsened as they aged. beta3(-/-) mice had much greater mortality after transverse aortic constriction (TAC) than WT controls. By 3 weeks of TAC, systolic function was worse. After 9 weeks of TAC, beta3(-/-) mice also had greater LV dilation, myocyte hypertrophy and enhanced fibrosis. NOS activity declined in beta3(-/-)TAC hearts after 9 weeks, and total and NOS-dependent superoxide rose, indicating heightened oxidative stress and NOS uncoupling. The level of eNOS phosphorylation in beta3(-/-)TAC hearts was diminished, and nNOS and iNOS expression levels were increased. GTP cyclohydrolase-1 expression was reduced, although total BH4 levels were not depleted. 3 weeks of BH4 treatment rescued beta3(-/-) mice from worsened remodeling after TAC, and lowered NOS-dependent superoxide. Thus, lack of beta3-AR signaling exacerbates cardiac pressure-overload induced remodeling and enhances NOS uncoupling and consequent oxidant stress, all of which can be rescued with exogenous BH4. These data suggest a cardioprotective role for the beta3-AR in modulating oxidative stress and adverse remodeling in the failing heart.


Assuntos
Hipertrofia Ventricular Esquerda/genética , Hipertrofia Ventricular Esquerda/patologia , Receptores Adrenérgicos beta 3/fisiologia , Remodelação Ventricular/genética , Remodelação Ventricular/fisiologia , Fatores Etários , Animais , Biopterinas/análogos & derivados , Biopterinas/uso terapêutico , Western Blotting , Cardiomiopatias/genética , Cardiomiopatias/patologia , Ecocardiografia , Hipertrofia Ventricular Esquerda/tratamento farmacológico , Masculino , Camundongos , Camundongos Mutantes , Óxido Nítrico Sintase/metabolismo , Estresse Oxidativo/genética , Estresse Oxidativo/fisiologia , Receptores Adrenérgicos beta 3/genética , Superóxidos/metabolismo , Vasoconstrição/fisiologia , Remodelação Ventricular/efeitos dos fármacos
12.
Circulation ; 117(14): 1810-9, 2008 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-18362233

RESUMO

BACKGROUND: The B vitamin folic acid (FA) is important to mitochondrial protein and nucleic acid synthesis, is an antioxidant, and enhances nitric oxide synthase activity. Here, we tested whether FA reduces myocardial ischemic dysfunction and postreperfusion injury. METHODS AND RESULTS: Wistar rats were pretreated with either FA (10 mg/d) or placebo for 1 week and then underwent in vivo transient left coronary artery occlusion for 30 minutes with or without 90 minutes of reperfusion (total n=131; subgroups used for various analyses). FA (4.5x10(-6) mol/L i.c.) pretreatment and global ischemia/reperfusion (30 minutes/30 minutes) also were performed in vitro (n=28). After 30 minutes of ischemia, global function declined more in controls than in FA-pretreated rats (Delta dP/dtmax, -878+/-586 versus -1956+/-351 mm Hg/s placebo; P=0.03), and regional thickening was better preserved (37.3+/-5.3% versus 5.1+/-0.6% placebo; P=0.004). Anterior wall perfusion fell similarly (-78.4+/-9.3% versus -71.2+/-13.8% placebo at 30 minutes), yet myocardial high-energy phosphates ATP and ADP reduced by ischemia in controls were better preserved by FA pretreatment (ATP: control, 2740+/-58 nmol/g; ischemia, 947+/-55 nmol/g; ischemia plus FA, 1332+/-101 nmol/g; P=0.02). Basal oxypurines (xanthine, hypoxanthine, and urate) rose with FA pretreatment but increased less during ischemia than in controls. Ischemic superoxide generation declined (3124+/-280 cpm/mg FA versus 5898+/-474 cpm/mg placebo; P=0.001). After reperfusion, FA-treated hearts had smaller infarcts (3.8+/-1.2% versus 60.3+/-4.1% placebo area at risk; P<0.002) and less contraction band necrosis, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling positivity, superoxide, and nitric oxide synthase uncoupling. Infarct size declined similarly with 1 mg/d FA. CONCLUSIONS: FA pretreatment blunts myocardial dysfunction during ischemia and ameliorates postreperfusion injury. This is coupled to preservation of high-energy phosphates, reducing subsequent reactive oxygen species generation, eNOS-uncoupling, and postreperfusion cell death.


Assuntos
Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Cardiotônicos/uso terapêutico , Oclusão Coronária/tratamento farmacológico , Ácido Fólico/uso terapêutico , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Pró-Fármacos/uso terapêutico , Animais , Cardiotônicos/farmacologia , Oclusão Coronária/metabolismo , Avaliação Pré-Clínica de Medicamentos , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Ácido Fólico/administração & dosagem , Ácido Fólico/farmacologia , Hiper-Homocisteinemia/tratamento farmacológico , Infarto do Miocárdio/etiologia , Infarto do Miocárdio/patologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico Sintase Tipo III , Estresse Oxidativo/efeitos dos fármacos , Pré-Medicação , Pró-Fármacos/administração & dosagem , Pró-Fármacos/farmacologia , Purinas/biossíntese , Ratos , Ratos Wistar , Superóxidos/metabolismo
13.
Circulation ; 117(20): 2626-36, 2008 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-18474817

RESUMO

BACKGROUND: Sustained pressure overload induces pathological cardiac hypertrophy and dysfunction. Oxidative stress linked to nitric oxide synthase (NOS) uncoupling may play an important role. We tested whether tetrahydrobiopterin (BH4) can recouple NOS and reverse preestablished advanced hypertrophy, fibrosis, and dysfunction. METHODS AND RESULTS: C57/Bl6 mice underwent transverse aortic constriction for 4 weeks, increasing cardiac mass (190%) and diastolic dimension (144%), lowering ejection fraction (-46%), and triggering NOS uncoupling and oxidative stress. Oral BH4 was then administered for 5 more weeks of pressure overload. Without reducing loading, BH4 reversed hypertrophy and fibrosis, recoupled endothelial NOS, lowered oxidant stress, and improved chamber and myocyte function, whereas untreated hearts worsened. If BH4 was started at the onset of pressure overload, it did not suppress hypertrophy over the first week when NOS activity remained preserved even in untreated transverse aortic constriction hearts. However, BH4 stopped subsequent remodeling when NOS activity was otherwise declining. A broad antioxidant, Tempol, also reduced oxidant stress yet did not recouple NOS or reverse worsened hypertrophy/fibrosis from sustained transverse aortic constriction. Microarray analysis revealed very different gene expression profiles for both treatments. BH4 did not enhance net protein kinase G activity. Finally, transgenic mice with enhanced BH4 synthesis confined to endothelial cells were unprotected against pressure overload, indicating that exogenous BH4 targeted myocytes and fibroblasts. CONCLUSIONS: NOS recoupling by exogenous BH4 ameliorates preexisting advanced cardiac hypertrophy/fibrosis and is more effective than a less targeted antioxidant approach (Tempol). These data highlight the importance of myocyte NOS uncoupling in hypertrophic heart disease and support BH4 as a potential new approach to treat this disorder.


Assuntos
Biopterinas/análogos & derivados , Cardiomegalia/tratamento farmacológico , Fibrose/tratamento farmacológico , Hipertensão/complicações , Miocárdio/patologia , Óxido Nítrico Sintase Tipo III/metabolismo , Animais , Biopterinas/farmacologia , Biopterinas/uso terapêutico , Óxidos N-Cíclicos/farmacologia , Óxidos N-Cíclicos/uso terapêutico , Modelos Animais de Doenças , GTP Cicloidrolase/genética , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Marcadores de Spin
15.
Circulation ; 115(16): 2159-67, 2007 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-17420342

RESUMO

BACKGROUND: Recent cell-based studies have found that cGMP synthesis and hydrolysis by phosphodiesterase (PDE) appear compartmentalized, with nitric oxide synthase-derived and/or PDE type 5 (PDE-5)-hydrolyzable cGMP undetected at the sarcolemmal membrane in contrast to cGMP stimulated by natriuretic peptide. In the present study, we determine the functional significance of such compartments with a comparison of beta-adrenergic modulation by PDE-5 inhibition to that of natriuretic peptide stimulation in both cardiomyocytes and intact hearts. The potential role of differential cGMP and protein kinase G stimulation by these 2 modulators was also studied. METHODS AND RESULTS: Intact C57/BL6 mouse hearts were studied with pressure-volume analysis, and adult isolated myocytes were studied with fluorescence microscopy. PDE-5 inhibition with 0.1 to 1 micromol/L sildenafil (SIL) suppressed isoproterenol (ISO)-stimulated contractility, whereas 10 micromol/L atrial natriuretic peptide (ANP) had no effect. ISO suppression by SIL was prevented in cells pretreated with a protein kinase G inhibitor. Surprisingly, myocardial cGMP changed little with SIL+ISO yet rose nearly 5-fold with ANP, whereas protein kinase G activation (vasodilator-stimulated protein phosphorylation; ELISA assay) displayed the opposite: increased with SIL+ISO but unaltered by ANP+ISO. PDE-5 and ANP compartments were functionally separated, as inhibition of nitric oxide synthase by N(w)-nitro-L-arginine methyl ester eliminated antiadrenergic effects of SIL, yet this was not restorable by co-stimulation with ANP. CONCLUSIONS: Regulation of cardiac beta-adrenergic response by cGMP is specifically linked to a nitric oxide-synthesis/PDE-5-hydrolyzed pool signaling via protein kinase G. Natriuretic peptide stimulation achieves greater detectable increases in cGMP but not protein kinase G activity and does not modulate beta-adrenergic response. Such disparities likely contribute to differential cardiac regulation by drugs that modulate cGMP synthesis and hydrolysis.


Assuntos
3',5'-GMP Cíclico Fosfodiesterases/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Contração Miocárdica/fisiologia , Miócitos Cardíacos/efeitos dos fármacos , Piperazinas/farmacologia , Receptores Adrenérgicos beta/fisiologia , Sulfonas/farmacologia , 3',5'-GMP Cíclico Fosfodiesterases/antagonistas & inibidores , Animais , Fator Natriurético Atrial/farmacologia , Compartimento Celular/fisiologia , Membrana Celular/enzimologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/fisiologia , GMP Cíclico/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5 , Citosol/enzimologia , Ativação Enzimática/efeitos dos fármacos , Guanilato Ciclase/metabolismo , Isoproterenol/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/fisiologia , Inibidores de Fosfodiesterase/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Purinas/farmacologia , Receptores Adrenérgicos beta/efeitos dos fármacos , Sistemas do Segundo Mensageiro/fisiologia , Citrato de Sildenafila
16.
Am J Cardiol ; 99(4): 476-81, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17293188

RESUMO

The aim of this study was to test the influence of high-dose folic acid (10 mg/d) on endothelial function in patients referred for coronary intervention after an acute myocardial infarction (AMI) and determine its relation to homocysteine levels. Flow-mediated dilation (FMD) of the brachial artery was performed in 40 patients after AMI (16 with normal homocysteine levels and 24 patients with elevated levels [>11 micromol/L]). Subjects were randomized to receive first folic acid (10 mg/day; group A) or placebo (group B) for 6 weeks in a double-blind crossover trial with a 2-week washout. Plasma folate, total homocysteine and its subtypes (oxidized, reduced, and protein-bound), FMD, and nitroglycerin-mediated dilation were assessed at baseline and at 6 and 14 weeks. In group A, folic acid improved FMD from 3.98 +/- 0.35% to 6.44 +/- 0.56% (p <0.001). This effect persisted after the crossover with placebo (5.42 +/- 0.59, p = 0.13). In group B, placebo did not increase FMD (4.01 +/- 0.34% vs 4.46 +/- 0.38, p = 0.38); however, a significant increase was observed in the second active treatment period (6.49 +/- 0.56%, p = 0.005). In both groups, improved FMD neither correlated with basal levels of homocysteine and its subtypes nor with changes induced during the folate treatment. Nitroglycerin-mediated dilation did not change significantly in either group. Folic acid increased FMD in both normo- and hyperhomocysteinanemic groups (p = 0.006 and p <0.001). In conclusion, 6-week treatment with high-dose folic acid improves endothelial function in post-AMI patients, independent from homocysteine status. Folic acid can be recommended to improve postinfarction endothelial dysfunction in patients with normo- and hyperhomocysteinemia.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Ácido Fólico/farmacologia , Infarto do Miocárdio/fisiopatologia , Complexo Vitamínico B/farmacologia , Artéria Braquial/efeitos dos fármacos , Distribuição de Qui-Quadrado , Estudos Cross-Over , Método Duplo-Cego , Endotélio Vascular/fisiopatologia , Feminino , Ácido Fólico/administração & dosagem , Homocisteína/sangue , Humanos , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento , Complexo Vitamínico B/administração & dosagem
17.
Arterioscler Thromb Vasc Biol ; 26(11): 2439-44, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16946131

RESUMO

Tetrahydrobiopterin (BH4) is an essential cofactor for the aromatic amino acid hydroxylases, which are essential in the formation of neurotransmitters, and for nitric oxide synthase. It is presently used clinically to treat some forms of phenylketonuria (PKU) that can be ameliorated by BH4 supplementation. Recent evidence supports potential cardiovascular benefits from BH4 replacement for the treatment of hypertension, ischemia-reperfusion injury, and cardiac hypertrophy with chamber remodeling. Such disorders exhibit BH4 depletion because of its oxidation and/or reduced synthesis, which can result in functional uncoupling of nitric oxide synthase (NOS). Uncoupled NOS generates more oxygen free radicals and less nitric oxide, shifting the nitroso-redox balance and having adverse consequences on the cardiovascular system. While previously difficult to use as a treatment because of chemical instability and cost, newer methods to synthesize stable BH4 suggest its novel potential as a therapeutic agent. This review discusses the biochemistry, physiology, and evolving therapeutic potential of BH4 for cardiovascular disease.


Assuntos
Biopterinas/análogos & derivados , Doenças Cardiovasculares/tratamento farmacológico , Animais , Aterosclerose/etiologia , Disponibilidade Biológica , Biopterinas/biossíntese , Biopterinas/química , Biopterinas/fisiologia , Biopterinas/uso terapêutico , Vasos Sanguíneos/efeitos dos fármacos , Coração/efeitos dos fármacos , Humanos , Óxido Nítrico Sintase/metabolismo , Estresse Oxidativo/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA