Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Dev Biol ; 399(1): 129-138, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25557619

RESUMEN

INTRODUCTION: GTP cyclohydrolase I (GTPCH) catalyses the first and rate-limiting reaction in the synthesis of the enzymatic cofactor, tetrahydrobiopterin (BH4). Loss of function mutations in the GCH1 gene lead to congenital neurological diseases such as DOPA-responsive dystonia and hyperphenylalaninemia. However, little is known about how GTPCH and BH4 affects embryonic development in utero, and in particular whether metabolic replacement or supplementation in pregnancy is sufficient to rescue genetic GTPCH deficiency in the developing embryo. METHODS AND RESULTS: Gch1 deficient mice were generated by the insertion of loxP sites flanking exons 2-3 of the Gch1 gene. Gch1(fl/fl) mice were bred with Sox2cre mice to generate mice with global Gch1 deficiency. Genetic ablation of Gch1 caused embryonic lethality by E13.5. Despite loss of Gch1 mRNA and GTPCH enzymatic activity, whole embryo BH4 levels were maintained until E11.5, indicating sufficient maternal transfer of BH4 to reach this stage of development. After E11.5, Gch1(-/-) embryos were deficient in BH4, but an unbiased metabolomic screen indicated that the lethality was not due to a gross disturbance in metabolic profile. Embryonic lethality in Gch1(-/-) embryos was not caused by structural abnormalities, but was associated with significant bradycardia at E11.5. Embryonic lethality was not rescued by maternal supplementation of BH4, but was partially rescued, up to E15.5, by maternal supplementation of BH4 and l-DOPA. CONCLUSION: These findings demonstrate a requirement for Gch1 in embryonic development and have important implications for the understanding of pathogenesis and treatment of genetic BH4 deficiencies, as well as the identification of new potential roles for BH4.


Asunto(s)
Biopterinas/análogos & derivados , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , GTP Ciclohidrolasa/metabolismo , Animales , Biopterinas/metabolismo , Cromatografía Líquida de Alta Presión , Embrión de Mamíferos/embriología , Femenino , GTP Ciclohidrolasa/genética , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Levodopa/metabolismo , Masculino , Espectrometría de Masas , Metabolómica , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
2.
Eur Heart J ; 35(29): 1971-80, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24135835

RESUMEN

AIMS: Predicting the likely success of primary PCI to salvage potential infarcted myocardium is desirable. We compared early invasive parameters of coronary microcirculation function with the levels of circulating endothelin (ET-1) and 6-month ejection fraction after STEMI. METHODS AND RESULTS: Forty-four STEMI patients underwent assessment of coronary flow reserve (CFR) and index of myocardial resistance (IMR) on completion of PPCI and one day later. Cardiac magnetic resonance (CMR) at 24 h and 6 months assessed ejection fraction, oedema, late gadolinium enhancement, and salvage. In patients with depressed EF, there was no difference in IMR or CFR measured immediately after PPCI compared with those with preserved EF. However, by Day 1, CFR was significantly lower in those with depressed EF [2.0(1.5-2.3) vs. 2.6(2.1-3.3), P = 0.008]. In multivariable models, higher CFR post-PPCI [EST: +8.9 (SE 3.7) per 1 CFR unit, P = 0.03] and greater increase in CFR between post-PPCI and Day 1 [EST: +8.5 (SE 3.4) per 1 CFR unit, P = 0.01] were associated with higher salvage index. Circulating endothelin levels were significantly elevated in the low EF group at both 6 and 24 h, and 24 h levels correlated with CFR. CONCLUSION: Changes of the coronary microcirculation in the first day after PPCI are associated with 6-month ejection fraction and myocardial salvage. Depressed CFR at 24 h is associated with CMR imaging indices of MVO and haemorrhage and elevated endothelin levels.


Asunto(s)
Circulación Coronaria/fisiología , Infarto del Miocardio/fisiopatología , Intervención Coronaria Percutánea , Oclusión Coronaria/fisiopatología , Vasos Coronarios/fisiología , Endotelina-1/metabolismo , Femenino , Hemorragia/fisiopatología , Humanos , Angiografía por Resonancia Magnética , Masculino , Microcirculación/fisiología , Microvasos/fisiología , Persona de Mediana Edad , Infarto del Miocardio/terapia , Estudios Prospectivos , Recuperación de la Función , Volumen Sistólico/fisiología , Resistencia Vascular/fisiología
3.
Circ Res ; 111(6): 718-27, 2012 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-22798524

RESUMEN

RATIONALE: Tetrahydrobiopterin (BH4) is an essential cofactor of nitric oxide synthases (NOS). Oral BH4 supplementation preserves cardiac function in animal models of cardiac disease; however, the mechanisms underlying these findings are not completely understood. OBJECTIVE: To study the effect of myocardial transgenic overexpression of the rate-limiting enzyme in BH4 biosynthesis, GTP cyclohydrolase 1 (GCH1), on NOS activity, myocardial function, and Ca2+ handling. METHODS AND RESULTS: GCH1overexpression significantly increased the biopterins level in left ventricular (LV) myocytes but not in the nonmyocyte component of the LV myocardium or in plasma. The ratio between BH4 and its oxidized products was lower in mGCH1-Tg, indicating that a large proportion of the myocardial biopterin pool was oxidized; nevertheless, myocardial NOS1 activity was increased in mGCH1-Tg, and superoxide release was significantly reduced. Isolated hearts and field-stimulated LV myocytes (3 Hz, 35°C) overexpressing GCH1 showed a faster relaxation and a PKA-mediated increase in the PLB Ser16 phosphorylated fraction and in the rate of decay of the [Ca2+]i transient. RyR2 S-nitrosylation and diastolic Ca2+ leak were larger in mGCH1-Tg and ICa density was lower; nevertheless the amplitude of the [Ca2+]i transient and contraction did not differ between genotypes, because of an increase in the SR fractional release of Ca2+ in mGCH1-Tg myocytes. Xanthine oxidoreductase inhibition abolished the difference in superoxide production but did not affect myocardial function in either group. By contrast, NOS1 inhibition abolished the differences in ICa density, Ser16 PLB phosphorylation, [Ca2+]i decay, and myocardial relaxation between genotypes. CONCLUSIONS: Myocardial GCH1 activity and intracellular BH4 are a limiting factor for constitutive NOS1 and SERCA2A activity in the healthy myocardium. Our findings suggest that GCH1 may be a valuable target for the treatment of LV diastolic dysfunction.


Asunto(s)
Biopterinas/análogos & derivados , GTP Ciclohidrolasa/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Animales , Biopterinas/metabolismo , Biopterinas/farmacología , Calcio/metabolismo , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Femenino , GTP Ciclohidrolasa/genética , Corazón/efectos de los fármacos , Corazón/fisiología , Humanos , Immunoblotting , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Miocardio/citología , Miocardio/enzimología , Miocitos Cardíacos/enzimología , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo , Superóxidos/metabolismo
4.
Eur Heart J ; 34(43): 3378-88, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23008511

RESUMEN

AIMS: Understanding endothelial cell repopulation post-stenting and how this modulates in-stent restenosis is critical to improving arterial healing post-stenting. We used a novel murine stent model to investigate endothelial cell repopulation post-stenting, comparing the response of drug-eluting stents with a primary genetic modification to improve endothelial cell function. METHODS AND RESULTS: Endothelial cell repopulation was assessed en face in stented arteries in ApoE(-/-) mice with endothelial-specific LacZ expression. Stent deployment resulted in near-complete denudation of endothelium, but was followed by endothelial cell repopulation, by cells originating from both bone marrow-derived endothelial progenitor cells and from the adjacent vasculature. Paclitaxel-eluting stents reduced neointima formation (0.423 ± 0.065 vs. 0.240 ± 0.040 mm(2), P = 0.038), but decreased endothelial cell repopulation (238 ± 17 vs. 154 ± 22 nuclei/mm(2), P = 0.018), despite complete strut coverage. To test the effects of selectively improving endothelial cell function, we used transgenic mice with endothelial-specific overexpression of GTP-cyclohydrolase 1 (GCH-Tg) as a model of enhanced endothelial cell function and increased NO production. GCH-Tg ApoE(-/-) mice had less neointima formation compared with ApoE(-/-) littermates (0.52 ± 0.08 vs. 0.26 ± 0.09 mm(2), P = 0.039). In contrast to paclitaxel-eluting stents, reduced neointima formation in GCH-Tg mice was accompanied by increased endothelial cell coverage (156 ± 17 vs. 209 ± 23 nuclei/mm(2), P = 0.043). CONCLUSION: Drug-eluting stents reduce not only neointima formation but also endothelial cell repopulation, independent of strut coverage. In contrast, selective targeting of endothelial cell function is sufficient to improve endothelial cell repopulation and reduce neointima formation. Targeting endothelial cell function is a rational therapeutic strategy to improve vascular healing and decrease neointima formation after stenting.


Asunto(s)
Aterosclerosis/patología , Células Endoteliales/patología , Endotelio Vascular/patología , Stents , Animales , Aspirina/farmacología , Stents Liberadores de Fármacos , Fibrinolíticos/farmacología , Masculino , Ratones , Ratones Endogámicos , Neointima/patología , Paclitaxel/farmacología , Moduladores de Tubulina/farmacología
5.
Circulation ; 124(10): 1107-17, 2011 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-21844076

RESUMEN

BACKGROUND: An altered nitric oxide-redox balance has been implicated in the pathogenesis of atrial fibrillation (AF). Statins inhibit NOX2-NADPH oxidases and prevent postoperative AF but are less effective in AF secondary prevention; the mechanisms underlying these findings are poorly understood. METHODS AND RESULTS: By using goat models of pacing-induced AF or of atrial structural remodeling secondary to atrioventricular block and right atrial samples from 130 patients undergoing cardiac surgery, we found that the mechanisms responsible for the NO-redox imbalance differ between atria and with the duration and substrate of AF. Rac1 and NADPH oxidase activity and the protein level of NOX2 and p22phox were significantly increased in the left atrium of goats after 2 weeks of AF and in patients who developed postoperative AF in the absence of differences in leukocytes infiltration. Conversely, in the presence of longstanding AF or atrioventricular block, uncoupled nitric oxide synthase activity (secondary to reduced BH4 content and/or increased arginase activity) and mitochondrial oxidases accounted for the biatrial increase in reactive oxygen species. Atorvastatin caused a mevalonate-reversible inhibition of Rac1 and NOX2-NADPH oxidase activity in right atrial samples from patients who developed postoperative AF, but it did not affect reactive oxygen species, nitric oxide synthase uncoupling, or BH4 in patients with permanent AF. CONCLUSIONS: Upregulation of atrial NADPH oxidases is an early but transient event in the natural history of AF. Changes in the sources of reactive oxygen species with atrial remodeling may explain why statins are effective in the primary prevention of AF but not in its management.


Asunto(s)
Antiarrítmicos/uso terapéutico , Fibrilación Atrial/prevención & control , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Arginasa/metabolismo , Bloqueo Atrioventricular/metabolismo , Modelos Animales de Enfermedad , Femenino , Cabras , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/metabolismo , Humanos , Masculino , Glicoproteínas de Membrana/biosíntesis , Persona de Mediana Edad , Mitocondrias/enzimología , NADPH Oxidasa 2 , NADPH Oxidasas/biosíntesis , NADPH Oxidasas/metabolismo , Oxidorreductasas/metabolismo , Proteína de Unión al GTP rac1/metabolismo
6.
J Mol Cell Cardiol ; 51(4): 564-9, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21645517

RESUMEN

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.''


Asunto(s)
Biopterinas/análogos & derivados , Cardiotónicos/uso terapéutico , Insuficiencia Cardíaca/tratamiento farmacológico , Hipertrofia Ventricular Izquierda/tratamiento farmacológico , Remodelación Ventricular/efectos de los fármacos , Análisis de Varianza , Animales , Biopterinas/metabolismo , Biopterinas/farmacocinética , Biopterinas/uso terapéutico , Cardiotónicos/farmacocinética , Relación Dosis-Respuesta a Droga , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/fisiopatología , Humanos , Hipertrofia Ventricular Izquierda/etiología , Hipertrofia Ventricular Izquierda/fisiopatología , Ligadura , Ratones , Ratones Endogámicos C57BL , Miocardio/patología , Distribución Aleatoria , Superóxidos/metabolismo , Función Ventricular Izquierda
8.
J Biol Chem ; 284(41): 28128-28136, 2009 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-19666465

RESUMEN

Tetrahyrobiopterin (BH4) is a required cofactor for the synthesis of nitric oxide by endothelial nitric-oxide synthase (eNOS), and BH4 bioavailability within the endothelium is a critical factor in regulating the balance between NO and superoxide production by eNOS (eNOS coupling). BH4 levels are determined by the activity of GTP cyclohydrolase I (GTPCH), the rate-limiting enzyme in de novo BH4 biosynthesis. However, BH4 levels may also be influenced by oxidation, forming 7,8-dihydrobiopterin (BH2), which promotes eNOS uncoupling. Conversely, dihydrofolate reductase (DHFR) can regenerate BH4 from BH2, but the functional importance of DHFR in maintaining eNOS coupling remains unclear. We investigated the role of DHFR in regulating BH4 versus BH2 levels in endothelial cells and in cell lines expressing eNOS combined with tet-regulated GTPCH expression in order to compare the effects of low or high levels of de novo BH4 biosynthesis. Pharmacological inhibition of DHFR activity by methotrexate or genetic knockdown of DHFR protein by RNA interference reduced intracellular BH4 and increased BH2 levels resulting in enzymatic uncoupling of eNOS, as indicated by increased eNOS-dependent superoxide but reduced NO production. In contrast to the decreased BH4:BH2 ratio induced by DHFR knockdown, GTPCH knockdown greatly reduced total biopterin levels but with no change in BH4:BH2 ratio. In cells expressing eNOS with low biopterin levels, DHFR inhibition or knockdown further diminished the BH4:BH2 ratio and exacerbated eNOS uncoupling. Taken together, these data reveal a key role for DHFR in eNOS coupling by maintaining the BH4:BH2 ratio, particularly in conditions of low total biopterin availability.


Asunto(s)
Biopterinas/análogos & derivados , Óxido Nítrico Sintasa de Tipo III/metabolismo , Tetrahidrofolato Deshidrogenasa/metabolismo , Animales , Antioxidantes/metabolismo , Biopterinas/metabolismo , Línea Celular , Células Cultivadas , Doxiciclina/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/enzimología , GTP Ciclohidrolasa/genética , GTP Ciclohidrolasa/metabolismo , Metotrexato/metabolismo , Ratones , Células 3T3 NIH , Óxido Nítrico Sintasa de Tipo III/genética , Oxidación-Reducción , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Superóxidos/metabolismo , Tetrahidrofolato Deshidrogenasa/genética
9.
Am J Physiol Heart Circ Physiol ; 299(1): H88-96, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20418482

RESUMEN

GTP cyclohydrolase I (GTPCH) is the rate-limiting enzyme for tetrahydrobiopterin (BH(4)) synthesis. Decreases in GTPCH activity and expression have been shown in late stages of acute cardiac rejection, suggesting a deficit in BH(4). We hypothesized that increasing intracellular levels of BH(4) by cardiac myocyte-targeted overexpression of GTPCH would diminish acute cardiac allograft rejection. Transgenic mice overexpressing GTPCH in the heart were generated and crossed on C57BL6 background. Wild-type and transgenic mouse donor hearts were transplanted into BALB/c recipient mice. Left ventricular (LV) function, histological rejection, BH(4) levels, and inflammatory cytokine gene expression (mRNA) were examined. Expression of human GTPCH was documented by PCR, Western analysis, and function by a significant (P < 0.001) increase in cardiac BH(4) levels. GTPCH transgene decreased histological rejection (46%; P < 0.003) and cardiac myocyte injury (eosin autofluorescence; 56%; P < 0.0001) independent of changes in inflammatory cytokine expression or nitric oxide content. GTPCH transgene decreased IL-2 (88%; P < 0.002), IL-1R2 (42%; P < 0.0001), and programmed cell death-1 (67%; P < 0.0001) expression, whereas it increased fms-like tyrosine kinase 3 (156%; P < 0.0001) and stromal-derived factor-1 (2; 190%; P < 0.0001) expression. There was no difference in ejection fraction or fractional shortening; however, LV mass was significantly increased (P < 0.05) only in wild-type grafts. The decreases in LV mass, cardiac injury, and histological rejection support a protective role of cardiac GTPCH overexpression and increased BH(4) synthesis in cardiac allografts. The mechanism of the decreased rejection appears related to decreased T cell proliferation and modulation of immune function by higher expression of genes involved in hematopoietic/stromal cell development and recruitment.


Asunto(s)
Biopterinas/análogos & derivados , GTP Ciclohidrolasa/metabolismo , Rechazo de Injerto/prevención & control , Trasplante de Corazón , Miocitos Cardíacos/enzimología , Enfermedad Aguda , Animales , Biopterinas/metabolismo , Citocinas/genética , Citocinas/metabolismo , GTP Ciclohidrolasa/genética , Genotipo , Rechazo de Injerto/diagnóstico por imagen , Rechazo de Injerto/enzimología , Rechazo de Injerto/inmunología , Rechazo de Injerto/fisiopatología , Trasplante de Corazón/efectos adversos , Humanos , Mediadores de Inflamación/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/patología , Óxido Nítrico/metabolismo , Fenotipo , ARN Mensajero/metabolismo , Trasplante Homólogo , Ultrasonografía , Función Ventricular Izquierda
10.
Clin Sci (Lond) ; 119(3): 131-42, 2010 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-20337596

RESUMEN

BH4 (tetrahydrobiopterin) supplementation improves endothelial function in models of vascular disease by maintaining eNOS (endothelial nitric oxide synthase) coupling and NO (nitric oxide) bioavailability. However, the cellular mechanisms through which enhanced endothelial function leads to reduced atherosclerosis remain unclear. We have used a pharmaceutical BH4 formulation to investigate the effects of BH4 supplementation on atherosclerosis progression in ApoE-KO (apolipoprotein E-knockout) mice. Single oral dose pharmacokinetic studies revealed rapid BH4 uptake into plasma and organs. Plasma BH4 levels returned to baseline by 8 h after oral dosing, but remained markedly increased in aorta at 24 h. Daily oral BH4 supplementation in ApoE-KO mice from 8 weeks of age, for a period of 8 or 12 weeks, had no effect on plasma lipids or haemodynamic parameters, but significantly reduced aortic root atherosclerosis compared with placebo-treated animals. BH4 supplementation significantly reduced VCAM-1 (vascular cell adhesion molecule 1) mRNA levels in aortic endothelial cells, markedly reduced the infiltration of T-cells, macrophages and monocytes into plaques, and reduced T-cell infiltration in the adjacent adventitia, but importantly had no effect on circulating leucocytes. GCH (GTP cyclohydrolase I)-transgenic mice, with a specific increase in endothelial BH4 levels, exhibited a similar reduction in vascular immune cell infiltration compared with BH4-deficient controls, suggesting that BH4 reduces vascular inflammation via endothelial cell signalling. In conclusion, BH4 supplementation reduces vascular immune cell infiltration in atherosclerosis and may therefore be a rational therapeutic approach to reduce the progression of atherosclerosis.


Asunto(s)
Enfermedades de la Aorta/tratamiento farmacológico , Apolipoproteínas E/deficiencia , Aterosclerosis/tratamiento farmacológico , Biopterinas/análogos & derivados , Administración Oral , Animales , Enfermedades de la Aorta/inmunología , Enfermedades de la Aorta/metabolismo , Apolipoproteínas E/genética , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Biopterinas/farmacocinética , Biopterinas/uso terapéutico , Quimiotaxis de Leucocito/efectos de los fármacos , Progresión de la Enfermedad , Esquema de Medicación , Evaluación Preclínica de Medicamentos , Endotelio Vascular/metabolismo , Hemodinámica/efectos de los fármacos , Lípidos/sangre , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/genética , Distribución Tisular , Molécula 1 de Adhesión Celular Vascular/biosíntesis , Molécula 1 de Adhesión Celular Vascular/metabolismo
11.
Circ Res ; 102(8): 923-32, 2008 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-18309100

RESUMEN

Oxidized low-density lipoproteins increase arginase activity and reciprocally decrease endothelial NO in human aortic endothelial cells. Here, we demonstrate that vascular endothelial arginase activity is increased in atherogenic-prone apolipoprotein E-null (ApoE(-/-)) and wild-type mice fed a high cholesterol diet. In ApoE(-/-) mice, selective arginase II inhibition or deletion of the arginase II gene (Arg II(-/-) mice) prevents high-cholesterol diet-dependent decreases in vascular NO production, decreases endothelial reactive oxygen species production, restores endothelial function, and prevents oxidized low-density lipoprotein-dependent increases in vascular stiffness. Furthermore, arginase inhibition significantly decreases plaque burden. These data indicate that arginase II plays a critical role in the pathophysiology of cholesterol-mediated endothelial dysfunction and represents a novel target for therapy in atherosclerosis.


Asunto(s)
Arginasa/fisiología , Aterosclerosis/etiología , Animales , Apolipoproteínas E/deficiencia , Arginasa/antagonistas & inhibidores , Arginasa/genética , Aterosclerosis/patología , Colesterol/administración & dosificación , Células Endoteliales/patología , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Ratones , Ratones Noqueados , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo III , Regulación hacia Arriba , Resistencia Vascular
12.
Circulation ; 117(8): 1045-54, 2008 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-18268143

RESUMEN

BACKGROUND: Tetrahydrobiopterin (BH4) is an essential cofactor of endothelial nitric oxide synthase (eNOS). When BH4 levels are decreased, eNOS becomes uncoupled to produce superoxide anion (O2(-)) instead of NO, which contributes to endothelial dysfunction. Deoxycorticosterone acetate (DOCA)-salt hypertension is characterized by a suppressed plasma renin level due to sodium retention but manifests in eNOS uncoupling; however, how endogenous BH4 regulates blood pressure is unknown. GTP cyclohydrolase I (GTPCH I) is the rate-limiting enzyme for de novo BH4 synthesis. This study tested the hypothesis that endothelium-specific GTPCH I overexpression retards the progression of hypertension through preservation of the structure and function of resistance mesenteric arteries. METHODS AND RESULTS: During 3 weeks of DOCA-salt treatment, arterial blood pressure was increased significantly in wild-type mice, as determined by radiotelemetry, but this increase was attenuated in transgenic mice with endothelium-specific GTPCH I overexpression (Tg-GCH). Arterial GTPCH I activity and BH4 levels were decreased significantly in wild-type DOCA-salt mice, but both were preserved in Tg-GCH mice despite DOCA-salt treatment. Significant remodeling of resistance mesenteric arteries (approximately 100-microm outside diameter) in wild-type DOCA-salt mice exists, evidenced by increased medial cross-sectional area, media thickness, and media-lumen ratio and overexpression of tenascin C, an extracellular matrix glycoprotein that contributes to hypertrophic remodeling; all of these effects were prevented in DOCA-salt-treated Tg-GCH mice. Furthermore, NO-mediated relaxation in mesenteric arteries was significantly improved in DOCA-salt-treated Tg-GCH mice, in parallel with reduced O2(-) levels. Finally, phosphorylation of eNOS at serine residue 1177 (eNOS-S1177), but not its dimer-monomer ratio, was decreased significantly in wild-type DOCA-salt mice compared with sham controls but was preserved in DOCA-salt-treated Tg-GCH mice. CONCLUSIONS: These results demonstrate that endothelium-specific GTPCH I overexpression abrogates O2(-) production and preserves eNOS phosphorylation, which results in preserved structural and functional integrity of resistance mesenteric arteries and lowered blood pressure in low-renin hypertension.


Asunto(s)
Endotelio Vascular/enzimología , GTP Ciclohidrolasa/metabolismo , Hipertensión/metabolismo , Hipertensión/fisiopatología , Renina/sangre , Animales , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Presión Sanguínea/fisiología , Peso Corporal , Vasos Coronarios/fisiología , Desoxicorticosterona , GTP Ciclohidrolasa/genética , Regulación Enzimológica de la Expresión Génica , Hipertensión/inducido químicamente , Masculino , Arterias Mesentéricas/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mineralocorticoides , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III , Especies Reactivas de Oxígeno/metabolismo , Cloruro de Sodio , Tenascina/metabolismo , Resistencia Vascular/fisiología , Vasodilatación/fisiología
13.
Circulation ; 118(14 Suppl): S71-7, 2008 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-18824773

RESUMEN

BACKGROUND: Vascular injury results in loss of endothelial nitric oxide (NO), production of reactive oxygen species (ROS), and the initiation of an inflammatory response. Both NO and ROS modulate inflammation through redox-sensitive pathways. Tetrahydrobiopterin (BH4) is an essential cofactor for endothelial nitric oxide synthase (eNOS) that regulates enzymatic synthesis of either nitric oxide or ROS. We hypothesized that endothelial BH4 is an important regulator of inflammation and vascular remodeling. METHODS AND RESULTS: Endothelium-targeted overexpression of GTP cyclohydrolase 1 (GCH), the rate limiting enzyme in BH4 synthesis, increased levels of tetrahydrobiopterin (BH4), reduced endothelial superoxide, improved eNOS coupling, and reduced vein graft atherosclerosis in transgenic GCH/ApoE-KO mice compared to ApoE-KO controls. Immunohistochemistry using anti-MAC-3 and MAC-1 antibody staining revealed a marked reduction in vein graft macrophage content, as did RT-PCR expression of macrophage marker CD68 mRNA levels in GCH/ApoE-KO mice. When we investigated the potential mediators of this reduction, we discovered that mRNA and protein levels of MCP-1 (CCL2) but not RANTES (CCL5) were significantly reduced in GCH/ApoE-KO aortic tissue. Consistent with this finding we found a decrease in CCR2-mediated, but not CCR5-mediated, chemotaxis in vascular tissue and plasma samples from GCH/ApoE-KO animals. CONCLUSIONS: Increased endothelial BH4 reduces vein graft neointimal hyperplasia and atherosclerosis through a reduction in vascular inflammation. These findings highlight the importance of MCP-1/CCR2 signaling in the response to vascular injury and identify novel pathways linking endothelial BH4 to inflammation and vascular remodeling.


Asunto(s)
Aterosclerosis/prevención & control , Biopterinas/análogos & derivados , Vasos Sanguíneos/lesiones , Quimiocina CCL2/metabolismo , Endotelio Vascular/metabolismo , Receptores CCR2/metabolismo , Vasculitis/prevención & control , Animales , Aorta/metabolismo , Apolipoproteínas E/deficiencia , Aterosclerosis/etiología , Biopterinas/metabolismo , Arterias Carótidas/cirugía , Quimiotaxis , Femenino , GTP Ciclohidrolasa/metabolismo , Humanos , Hiperplasia , Macrófagos/patología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Superóxidos/metabolismo , Túnica Íntima/patología , Regulación hacia Arriba , Vasculitis/complicaciones , Venas Cavas/metabolismo , Venas Cavas/patología , Venas Cavas/trasplante , Heridas y Lesiones/complicaciones
14.
Circulation ; 117(20): 2626-36, 2008 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-18474817

RESUMEN

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.


Asunto(s)
Biopterinas/análogos & derivados , Cardiomegalia/tratamiento farmacológico , Fibrosis/tratamiento farmacológico , Hipertensión/complicaciones , Miocardio/patología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Animales , Biopterinas/farmacología , Biopterinas/uso terapéutico , Óxidos N-Cíclicos/farmacología , Óxidos N-Cíclicos/uso terapéutico , Modelos Animales de Enfermedad , GTP Ciclohidrolasa/genética , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Marcadores de Spin
15.
Clin Sci (Lond) ; 114(8): 509-31, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18336368

RESUMEN

Cardiovascular disease, resulting from atherosclerosis, is a leading cause of global morbidity and mortality. Genetic predisposition and classical environmental risk factors explain much of the attributable risk for cardiovascular events in populations, but other risk factors for the development and progression of atherosclerosis, which can be identified and modified, may be important therapeutic targets. Infectious agents, such as Chlamydia pneumoniae, have been proposed as contributory factors in the pathogenesis of atherosclerosis. In the present review, we consider the experimental evidence that has accumulated over the last 20 years evaluating the role of C. pneumoniae in atherosclerosis and suggest areas for future research in this field.


Asunto(s)
Aterosclerosis/microbiología , Infecciones por Chlamydophila/complicaciones , Chlamydophila pneumoniae/aislamiento & purificación , Animales , Antibacterianos/uso terapéutico , Aterosclerosis/epidemiología , Aterosclerosis/prevención & control , Infecciones por Chlamydophila/tratamiento farmacológico , Infecciones por Chlamydophila/epidemiología , ADN Bacteriano/análisis , Modelos Animales de Enfermedad , Humanos , Factores de Riesgo , Estudios Seroepidemiológicos
16.
Arterioscler Thromb Vasc Biol ; 27(4): 833-40, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17204666

RESUMEN

OBJECTIVE: We aimed to develop and validate a model of angioplasty and stenting in mice that would allow investigation of the response to stent injury using genetically modified mouse strains. METHODS AND RESULTS: Aortic segments from either C57BL/6 wild-type or atherosclerotic ApoE-KO mice underwent balloon angioplasty alone or balloon angioplasty and stenting with a 1.25x2.5 mm stainless steel stent. Vessels were carotid-interposition grafted into genetically identical littermate recipients and harvested at 1, 7, 14, or 28 days. In wild-type mice, stenting generated an inflammatory vascular injury response between days 1 to 7, leading to the development of neointimal hyperplasia by day 14, which further increased in area by day 28 leading to the development of in-stent stenosis. Uninjured vessels and vessels injured by balloon angioplasty alone developed minimal neointimal hyperplasia. In stented ApoE-KO mice, neointimal area at 28 days was 30% greater compared with wild-type mice. CONCLUSIONS: By reproducing important features of human stenting in atherosclerotic mice, we provide the potential to investigate molecular pathways and evaluate novel therapeutic targets for stent injury and restenosis.


Asunto(s)
Angioplastia de Balón/efectos adversos , Apolipoproteínas E/deficiencia , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Stents/efectos adversos , Animales , Aorta Torácica/lesiones , Aorta Torácica/patología , Aorta Torácica/trasplante , Arterias Carótidas/cirugía , Constricción Patológica/etiología , Femenino , Hiperplasia , Inmunoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Tiempo , Túnica Íntima/patología , Vasculitis/etiología , Vasculitis/patología
17.
Arterioscler Thromb Vasc Biol ; 27(7): 1632-7, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17463333

RESUMEN

OBJECTIVE: When the availability of tetrahydrobiopterin (BH4) is deficient, endothelial nitric oxide synthase (eNOS) produces superoxide rather than NO (uncoupled eNOS). We have shown that the atherosclerotic lesion size was augmented in apolipoprotein E-deficient (ApoE-KO) mice overexpressing eNOS because of the enhanced superoxide production. In this study, we addressed the specific importance of uncoupled eNOS in atherosclerosis, and the potential mechanistic role for specific versus nonspecific antioxidant strategies in restoring eNOS coupling. METHODS AND RESULTS: We crossed mice overexpressing eNOS in the endothelium (eNOS-Tg) with mice overexpressing GTP-cyclohydrolase I (GCH), the rate-limiting enzyme in BH4 synthesis, to generate ApoE-KO/eNOS-Tg/GCH-Tg mice. As a comparison, ApoE-KO/eNOS-Tg mice were treated with vitamin C. Atherosclerotic lesion formation was increased in ApoE-KO/eNOS-Tg mice compared with ApoE-KO mice. GCH overexpression in ApoE-KO/eNOS-Tg/GCH-Tg mice increased vascular BH4 levels and reduced plaque area. This reduction was associated with decreased superoxide production from uncoupled eNOS. Vitamin C treatment failed to reduce atherosclerotic lesion size in ApoE-KO/eNOS-Tg mice, despite reducing overall vascular superoxide production. CONCLUSION: In contrast to vitamin C treatment, augmenting BH4 levels in the endothelium by GCH overexpression reduced the accelerated atherosclerotic lesion formation in ApoE-KO/eNOS-Tg mice, associated with a reduction of superoxide production from uncoupled eNOS.


Asunto(s)
Antioxidantes/farmacología , Aterosclerosis/fisiopatología , Biopterinas/análogos & derivados , Óxido Nítrico Sintasa de Tipo III/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Análisis de Varianza , Animales , Apolipoproteínas E/deficiencia , Ácido Ascórbico/farmacología , Aterosclerosis/metabolismo , Biopterinas/metabolismo , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Femenino , GTP Ciclohidrolasa/análisis , GTP Ciclohidrolasa/metabolismo , Ratones , Ratones Endogámicos C57BL , Probabilidad , Especies Reactivas de Oxígeno/metabolismo , Sensibilidad y Especificidad , Superóxidos/metabolismo
19.
J Clin Invest ; 112(5): 725-35, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12952921

RESUMEN

Increased production of reactive oxygen species and loss of endothelial NO bioactivity are key features of vascular disease states such as diabetes mellitus. Tetrahydrobiopterin (BH4) is a required cofactor for eNOS activity; pharmacologic studies suggest that BH4 may mediate some of the adverse effects of diabetes on eNOS function. We have now investigated the importance and mechanisms of BH4 availability in vivo using a novel transgenic mouse model with endothelial-targeted overexpression of the rate-limiting enzyme in BH4 synthesis, guanosine triphosphate-cyclohydrolase I (GTPCH). Transgenic (GCH-Tg) mice demonstrated selective augmentation of endothelial BH4 levels. In WT mice, induction of diabetes with streptozotocin (STZ) increased vascular oxidative stress, resulting in oxidative loss of BH4, forming BH2 and biopterin. Endothelial cell superoxide production in diabetes was increased, and NO-mediated endothelium-dependent vasodilatation was impaired. In diabetic GCH-Tg mice, superoxide production from the endothelium was markedly reduced compared with that of WT mice, endothelial BH4 levels were maintained despite some oxidative loss of BH4, and NO-mediated vasodilatation was preserved. These findings indicate that BH4 is an important mediator of eNOS regulation in diabetes and is a rational therapeutic target to restore NO-mediated endothelial function in diabetes and other vascular disease states.


Asunto(s)
Biopterinas/análogos & derivados , Biopterinas/fisiología , Diabetes Mellitus Experimental/metabolismo , Endotelio Vascular/fisiología , GTP Ciclohidrolasa/fisiología , Óxido Nítrico/fisiología , Animales , Diabetes Mellitus Experimental/terapia , GTP Ciclohidrolasa/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico Sintasa de Tipo III , Estrés Oxidativo , ARN Mensajero/análisis , Estreptozocina , Superóxidos/metabolismo
20.
Clin Sci (Lond) ; 113(2): 47-63, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17555404

RESUMEN

NO produced by eNOS (endothelial nitric oxide synthase) is a key mediator of vascular homoeostasis. NO bioavailability is reduced early in vascular disease states, such as hypercholesterolaemia, diabetes and hypertension, and throughout the progression of atherosclerosis. This is a result of both reduced NO synthesis and increased NO consumption by reactive oxygen species. eNOS enzymatic activity appears to be determined by the availability of its cofactor BH4 (tetrahydrobiopterin). When BH4 levels are adequate, eNOS produces NO; when BH4 levels are limiting, eNOS becomes enzymatically uncoupled and generates superoxide, contributing to vascular oxidative stress and endothelial dysfunction. BH4 bioavailability is determined by a balance of enzymatic de novo synthesis and recycling, versus oxidative degradation in dysfunctional endothelium. Augmenting vascular BH4 levels by pharmacological supplementation, by enhancing the rate of de novo biosynthesis or by measures to reduce BH4 oxidation have been shown in experimental studies to enhance NO bioavailability. Thus BH4 represents a potential therapeutic target for preserving eNOS function in vascular disease.


Asunto(s)
Aterosclerosis/metabolismo , Biopterinas/análogos & derivados , Endotelio Vascular/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo , Animales , Ácido Ascórbico/uso terapéutico , Aterosclerosis/tratamiento farmacológico , Biopterinas/biosíntesis , Biopterinas/metabolismo , Progresión de la Enfermedad , Homeostasis , Humanos , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Vitaminas/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA