Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cardiovasc Diabetol ; 18(1): 35, 2019 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-30885203

RESUMEN

BACKGROUND: This pathophysiological study addressed the hypothesis that soluble epoxide hydrolase (sEH), which metabolizes the vasodilator and anti-inflammatory epoxyeicosatrienoic acids (EETs) to dihydroxyeicosatrienoic acids (DHETs), contributes to conduit artery endothelial dysfunction in type 2 diabetes. METHODS AND RESULTS: Radial artery endothelium-dependent flow-mediated dilatation in response to hand skin heating was reduced in essential hypertensive patients (n = 9) and type 2 diabetic subjects with (n = 19) or without hypertension (n = 10) compared to healthy subjects (n = 36), taking into consideration cardiovascular risk factors, flow stimulus and endothelium-independent dilatation to glyceryl trinitrate. Diabetic patients but not non-diabetic hypertensive subjects displayed elevated whole blood reactive oxygen species levels and loss of NO release during heating, assessed by measuring local plasma nitrite variation. Moreover, plasma levels of EET regioisomers increased during heating in healthy subjects, did not change in hypertensive patients and decreased in diabetic patients. Correlation analysis showed in the overall population that the less NO and EETs bioavailability increases during heating, the more flow-mediated dilatation is reduced. The expression and activity of sEH, measured in isolated peripheral blood mononuclear cells, was elevated in diabetic but not hypertensive patients, leading to increased EETs conversion to DHETs. Finally, hyperglycemic and hyperinsulinemic euglycemic clamps induced a decrease in flow-mediated dilatation in healthy subjects and this was associated with an altered EETs release during heating. CONCLUSIONS: These results demonstrate that an increased EETs degradation by sEH and altered NO bioavailability are associated with conduit artery endothelial dysfunction in type 2 diabetic patients independently from their hypertensive status. The hyperinsulinemic and hyperglycemic state in these patients may contribute to these alterations. Trial registration NCT02311075. Registered December 8, 2014.


Asunto(s)
Diabetes Mellitus Tipo 2/sangre , Angiopatías Diabéticas/sangre , Eicosanoides/sangre , Hipertensión Esencial/sangre , Arteria Radial/metabolismo , Vasodilatación , Anciano , Biomarcadores/sangre , Estudios de Casos y Controles , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/fisiopatología , Angiopatías Diabéticas/diagnóstico , Angiopatías Diabéticas/fisiopatología , Epóxido Hidrolasas/metabolismo , Hipertensión Esencial/diagnóstico , Hipertensión Esencial/fisiopatología , Femenino , Humanos , Hipertermia Inducida , Masculino , Persona de Mediana Edad , Óxido Nítrico/metabolismo , Nitritos/sangre , Nitroglicerina/administración & dosificación , Arteria Radial/efectos de los fármacos , Arteria Radial/fisiopatología , Vasodilatación/efectos de los fármacos , Vasodilatadores/administración & dosificación
2.
Am J Physiol Heart Circ Physiol ; 314(6): H1279-H1288, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29569957

RESUMEN

We have previously shown that protein tyrosine phosphatase 1B (PTP1B) inactivation in mice [PTP1B-deficient (PTP1B-/-) mice] improves left ventricular (LV) angiogenesis, perfusion, remodeling, and function and limits endothelial dysfunction after myocardial infarction. However, whether PTP1B inactivation slows aging-associated cardiovascular dysfunction remains unknown. Wild-type (WT) and PTP1B-/- mice were allowed to age until 18 mo. Compared with old WT mice, in which aging increased the LV mRNA expression of PTP1B, old PTP1B-/- mice had 1) reduced cardiac hypertrophy with decreased LV mRNA levels of hypertrophic markers and atrial and brain natriuretic peptides, 2) lower LV fibrosis (collagen: 16 ± 3% in WT mice and 5 ± 3% in PTP1B-/- mice, P < 0.001) with decreased mRNA levels of transforming growth-factor-ß1 and matrix metalloproteinase-2, and 3) higher LV capillary density and lower LV mRNA level of hypoxic inducible factor-1α, which was associated over time with a higher rate of proangiogenic M2 type macrophages and a stable LV mRNA level of VEGF receptor-2. Echocardiography revealed an age-dependent LV increase in end-diastolic volume in WT mice together with alterations of fractional shortening and diastole (transmitral Doppler E-to-A wave ratio). Invasive hemodynamics showed better LV systolic contractility and better diastolic compliance in old PTP1B-/- mice (LV end-systolic pressure-volume relation: 13.9 ± 0.9 in WT mice and 18.4 ± 1.6 in PTP1B-/- mice; LV end-diastolic pressure-volume relation: 5.1 ± 0.8 mmHg/relative volume unit in WT mice and 1.2 ± 0.3 mmHg/relative volume unit in PTP1B-/- mice, P < 0.05). In addition, old PTP1B-/- mice displayed a reduced amount of LV reactive oxygen species. Finally, in isolated resistance mesenteric arteries, PTP1B inactivation reduced aging-associated endothelial dysfunction (flow-mediated dilatation: -0.4 ± 2.1% in WT mice and 8.2 ± 2.8% in PTP1B-/- mice, P < 0.05). We conclude that PTP1B inactivation slows aging-associated LV remodeling and dysfunction and reduces endothelial dysfunction in mesenteric arteries. NEW & NOTEWORTHY The present study shows that protein tyrosine phosphatase 1B inactivation in aged mice improves left ventricular systolic and diastolic function associated with reduced adverse cardiac remodeling (hypertrophy, fibrosis, and capillary rarefaction) and limits vascular endothelial dysfunction. This suggests that protein tyrosine phosphatase 1B inhibition could be an interesting treatment approach in age-related cardiovascular dysfunction.


Asunto(s)
Insuficiencia Cardíaca/prevención & control , Ventrículos Cardíacos/enzimología , Hipertrofia Ventricular Izquierda/prevención & control , Proteína Tirosina Fosfatasa no Receptora Tipo 1/deficiencia , Disfunción Ventricular Izquierda/prevención & control , Función Ventricular Izquierda , Remodelación Ventricular , Factores de Edad , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Modelos Animales de Enfermedad , Fibrosis , Regulación Enzimológica de la Expresión Génica , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Hemodinámica , Hipertrofia Ventricular Izquierda/enzimología , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/fisiopatología , Masculino , Arterias Mesentéricas/enzimología , Arterias Mesentéricas/fisiopatología , Ratones Endogámicos BALB C , Ratones Noqueados , Neovascularización Fisiológica , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Disfunción Ventricular Izquierda/enzimología , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/fisiopatología
3.
Diabetes Obes Metab ; 20(10): 2399-2407, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29862614

RESUMEN

AIM: To determine whether non-steroidal mineralocorticoid receptor (MR) antagonists oppose metabolic syndrome-related end-organ, i.e. cardiac, damage. MATERIALS AND METHODS: In Zucker fa/fa rats, a rat model of metabolic syndrome, we assessed the effects of the non-steroidal MR antagonist finerenone (oral 2 mg/kg/day) on left ventricular (LV) function, haemodynamics and remodelling (using echocardiography, magnetic resonance imaging and biochemical methods). RESULTS: Long-term (90 days) finerenone modified neither systolic blood pressure nor heart rate, but reduced LV end-diastolic pressure and LV end-diastolic pressure-volume relationship, without modifying LV end-systolic pressure and LV end-systolic pressure-volume relationship. Simultaneously, long-term finerenone reduced both LV systolic and diastolic diameters, associated with reductions in LV weight and LV collagen density, while proteinuria and renal nGAL expression were reduced. Short-term (7 days) finerenone improved LV haemodynamics and reduced LV systolic diameter, without modifying LV diastolic diameter. Moreover, short-term finerenone increased myocardial tissue perfusion and reduced myocardial reactive oxygen species, while plasma nitrite levels, an indicator of nitric oxide (NO) bio-availability, were increased. CONCLUSIONS: In rats with metabolic syndrome, the non-steroidal MR antagonist finerenone opposed metabolic syndrome-related diastolic cardiac dysfunction and nephropathy. This involved acute effects, such as improved myocardial perfusion, reduced oxidative stress/increased NO bioavailability, as well as long-term effects, such as modifications in the myocardial structure.


Asunto(s)
Enfermedades Cardiovasculares/prevención & control , Enfermedades Renales/prevención & control , Síndrome Metabólico/tratamiento farmacológico , Antagonistas de Receptores de Mineralocorticoides/administración & dosificación , Naftiridinas/administración & dosificación , Animales , Enfermedades Cardiovasculares/complicaciones , Esquema de Medicación , Frecuencia Cardíaca/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Enfermedades Renales/complicaciones , Masculino , Síndrome Metabólico/complicaciones , Síndrome Metabólico/patología , Síndrome Metabólico/fisiopatología , Antagonistas de Receptores de Mineralocorticoides/efectos adversos , Naftiridinas/efectos adversos , Óxido Nítrico/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Zucker , Factores de Tiempo , Función Ventricular Izquierda/efectos de los fármacos
4.
Planta ; 246(6): 1109-1124, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28815300

RESUMEN

MAIN CONCLUSION: A chemical screen of plant-derived compounds identified holaphyllamine, a steroid, able to trigger defense responses in Arabidopsis thaliana and improve resistance against the pathogenic bacterium Pseudomonas syringae pv tomato DC3000. A chemical screen of 1600 plant-derived compounds was conducted and allowed the identification of a steroid able to activate defense responses in A. thaliana at a concentration of 1 µM without altering growth. The identified compound is holaphyllamine (HPA) whose chemical structure is similar to steroid pregnanes of mammals. Our data show that HPA, which is not constitutively present in A. thaliana, is able to trigger the formation of reactive oxygen species, deposition of callose and expression of several pathogenesis-related genes of the salicylic and jasmonic acid pathways. In addition, the results show that pre-treatment of A. thaliana seedlings with HPA before infection with the pathogenic bacterium Pseudomonas syringae pv tomato DC3000 results in a significant reduction of symptoms (i.e., reduction of bacterial colonies). Using A. thaliana mutants, we have found that the activation of defense responses by HPA does not depend on BRI1/BAK1 receptor kinases. Finally, a structure/function study reveals that the minimal structure required for activity is a 5-pregnen-20-one steroid with an equatorial nucleophilic group in C-3. Together, these findings demonstrate that HPA can activate defense responses that lead to improved resistance against bacterial infection in A. thaliana.


Asunto(s)
Arabidopsis/efectos de los fármacos , Resistencia a la Enfermedad , Regulación de la Expresión Génica de las Plantas/efectos de los fármacos , Fitosteroles/farmacología , Enfermedades de las Plantas/inmunología , Pseudomonas syringae/fisiología , Arabidopsis/genética , Arabidopsis/inmunología , Arabidopsis/microbiología , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Células Cultivadas , Ciclopentanos/metabolismo , Glucanos/metabolismo , Mutación , Oxilipinas/metabolismo , Fitosteroles/química , Enfermedades de las Plantas/microbiología , Reguladores del Crecimiento de las Plantas/metabolismo , Hojas de la Planta/efectos de los fármacos , Hojas de la Planta/genética , Hojas de la Planta/inmunología , Hojas de la Planta/microbiología , Especies Reactivas de Oxígeno/metabolismo , Estallido Respiratorio/efectos de los fármacos , Ácido Salicílico/metabolismo , Plantones/efectos de los fármacos , Plantones/genética , Plantones/inmunología , Plantones/microbiología , Bibliotecas de Moléculas Pequeñas , Nicotiana/efectos de los fármacos
5.
Circulation ; 130(7): 554-64, 2014 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-24928681

RESUMEN

BACKGROUND: Obesity and diabetes mellitus are independently associated with the development of heart failure. In this study, we determined the respective effects of obesity, insulin resistance, and diabetes mellitus on the intrinsic contraction and mitochondrial function of the human myocardium before the onset of cardiomyopathy. METHODS AND RESULTS: Right atrial myocardium was obtained from 141 consecutive patients presenting no sign of cardiomyopathy. We investigated ex vivo isometric contraction, mitochondrial respiration and calcium retention capacity, and respiratory chain complex activities and oxidative stress status. Diabetes mellitus was associated with a pronounced impairment of intrinsic contraction, mitochondrial dysfunction, and increased myocardial oxidative stress, regardless of weight status. In contrast, obesity was associated with less pronounced contractile dysfunction without any significant perturbation of mitochondrial function or oxidative stress status. Tested as continuous variables, glycated hemoglobin A1C, but neither body mass index nor the insulin resistance index (homeostasis model assessment-insulin resistance), was independently associated with cardiac mitochondrial function. Furthermore, diabetes mellitus was associated with cardiac mitochondrial network fragmentation and significantly decreased expression of the mitochondrial fusion related protein MFN1. Myocardial MFN1 content was inversely proportional to hemoglobin A1C. CONCLUSION: Worsening of intrinsic myocardial contraction in the transition from obesity to diabetes mellitus is likely related to worsening of cardiac mitochondrial function because impaired mitochondrial function and dynamics and contractile dysfunction are observed in diabetic patients but not in "metabolically healthy" obese patients at early stage in insulin resistance.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Mitocondrias Cardíacas/fisiología , Contracción Miocárdica/fisiología , Obesidad/fisiopatología , Anciano , Función del Atrio Derecho/fisiología , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/sangre , Femenino , Humanos , Resistencia a la Insulina/fisiología , Masculino , Persona de Mediana Edad , Obesidad/sangre , Técnicas de Cultivo de Órganos , Estudios Prospectivos
6.
Kidney Int ; 87(2): 465-72, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25029430

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is a renal hereditary disorder associated with increased cardiovascular mortality, due to mutations in polycystin-1 and polycystin-2 genes. Endothelial polycystin-deficient cells have an altered mechanosensitivity to fluid shear stress and subsequent deficit in calcium-induced nitric oxide release, prevented by dopamine receptor stimulation. However, the impact of polycystin deficiency on endothelial function in ADPKD patients is still largely unknown. Here we assessed endothelium-dependent flow-mediated dilatation in 21 normotensive ADPKD patients and 21 healthy control subjects, during sustained (hand skin heating) and transient (postischemic hyperemia) flow stimulation. Flow-mediated dilatation was less marked in ADPKD patients than in controls during heating, but it was similar during postischemic hyperemia. There was no difference in endothelium-independent dilatation in response to glyceryl trinitrate. Local plasma nitrite, an indicator of nitric oxide availability, increased during heating in controls but not in patients. Brachial infusion of dopamine in a subset of ADPKD patients stimulated plasma nitrite increase during heating and improved flow-mediated dilatation. Thus, ADPKD patients display a loss of nitric oxide release and an associated reduction in endothelium-dependent dilatation of conduit arteries during sustained blood flow increase. The correction of these anomalies by dopamine suggests future therapeutic strategies that could reduce the occurrence of cardiovascular events in ADPKD.


Asunto(s)
Hemodinámica/fisiología , Riñón Poliquístico Autosómico Dominante/fisiopatología , Canales Catiónicos TRPP/deficiencia , Adulto , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/fisiopatología , Estudios de Casos y Controles , Dopamina/fisiología , Endotelio Vascular/fisiopatología , Femenino , Humanos , Masculino , Mutación , Óxido Nítrico/fisiología , Riñón Poliquístico Autosómico Dominante/complicaciones , Riñón Poliquístico Autosómico Dominante/genética , Arteria Radial/fisiopatología , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/fisiología , Vasodilatación/fisiología , Adulto Joven
7.
Am J Physiol Heart Circ Physiol ; 308(9): H1020-9, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25724490

RESUMEN

This study addressed the hypothesis that inhibiting the soluble epoxide hydrolase (sEH)-mediated degradation of epoxy-fatty acids, notably epoxyeicosatrienoic acids, has an additional impact against cardiovascular damage in insulin resistance, beyond its previously demonstrated beneficial effect on glucose homeostasis. The cardiovascular and metabolic effects of the sEH inhibitor trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid (t-AUCB; 10 mg/l in drinking water) were compared with those of the sulfonylurea glibenclamide (80 mg/l), both administered for 8 wk in FVB mice subjected to a high-fat diet (HFD; 60% fat) for 16 wk. Mice on control chow diet (10% fat) and nontreated HFD mice served as controls. Glibenclamide and t-AUCB similarly prevented the increased fasting glycemia in HFD mice, but only t-AUCB improved glucose tolerance and decreased gluconeogenesis, without modifying weight gain. Moreover, t-AUCB reduced adipose tissue inflammation, plasma free fatty acids, and LDL cholesterol and prevented hepatic steatosis. Furthermore, only the sEH inhibitor improved endothelium-dependent relaxations to acetylcholine, assessed by myography in isolated coronary arteries. This improvement was related to a restoration of epoxyeicosatrienoic acid and nitric oxide pathways, as shown by the increased inhibitory effects of the nitric oxide synthase and cytochrome P-450 epoxygenase inhibitors l-NA and MSPPOH on these relaxations. Moreover, t-AUCB decreased cardiac hypertrophy, fibrosis, and inflammation and improved diastolic function, as demonstrated by the increased E/A ratio (echocardiography) and decreased slope of the end-diastolic pressure-volume relation (invasive hemodynamics). These results demonstrate that sEH inhibition improves coronary endothelial function and prevents cardiac remodeling and diastolic dysfunction in obese insulin-resistant mice.


Asunto(s)
Benzoatos/farmacología , Vasos Coronarios/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Epóxido Hidrolasas/antagonistas & inhibidores , Cardiopatías/prevención & control , Resistencia a la Insulina , Obesidad/tratamiento farmacológico , Urea/análogos & derivados , Vasodilatación/efectos de los fármacos , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Vasos Coronarios/enzimología , Vasos Coronarios/fisiopatología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Eicosanoides/metabolismo , Endotelio Vascular/enzimología , Endotelio Vascular/fisiopatología , Epóxido Hidrolasas/metabolismo , Gliburida/farmacología , Cardiopatías/enzimología , Cardiopatías/etiología , Cardiopatías/fisiopatología , Hipoglucemiantes/farmacología , Mediadores de Inflamación/metabolismo , Lípidos/sangre , Masculino , Ratones , Óxido Nítrico/metabolismo , Obesidad/sangre , Obesidad/complicaciones , Obesidad/enzimología , Obesidad/fisiopatología , Factores de Tiempo , Urea/farmacología , Vasodilatadores/farmacología , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos
8.
Crit Care Med ; 43(10): e420-30, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26110489

RESUMEN

OBJECTIVES: Total liquid ventilation provides ultrafast and potently neuro- and cardioprotective cooling after shockable cardiac arrest and myocardial infarction in animals. Our goal was to decipher the effect of hypothermic total liquid ventilation on the systemic and cerebral response to asphyxial cardiac arrest using an original pressure- and volume-controlled ventilation strategy in rabbits. DESIGN: Randomized animal study. SETTING: Academic research laboratory. SUBJECTS: New Zealand Rabbits. INTERVENTIONS: Thirty-six rabbits were submitted to 13 minutes of asphyxia, leading to cardiac arrest. After resumption of spontaneous circulation, they underwent either normothermic life support (control group, n = 12) or hypothermia induced by either 30 minutes of total liquid ventilation (total liquid ventilation group, n = 12) or IV cold saline (conventional cooling group, n = 12). MEASUREMENTS AND MAIN RESULTS: Ultrafast cooling with total liquid ventilation (32 °C within 5 min in the esophagus) dramatically attenuated the post-cardiac arrest syndrome regarding survival, neurologic dysfunction, and histologic lesions (brain, heart, kidneys, liver, and lungs). Final survival rate achieved 58% versus 0% and 8% in total liquid ventilation, control, and conventional cooling groups (p < 0.05), respectively. This was accompanied by an early preservation of the blood-brain barrier integrity and cerebral hemodynamics as well as reduction in the immediate reactive oxygen species production in the brain, heart, and kidneys after cardiac arrest. Later on, total liquid ventilation also mitigated the systemic inflammatory response through alteration of monocyte chemoattractant protein-1, interleukin-1ß, and interleukin-8 transcripts levels compared with control. In the conventional cooling group, cooling was achieved more slowly (32 °C within 90-120 min in the esophagus), providing none of the above-mentioned systemic or organ protection. CONCLUSIONS: Ultrafast cooling by total liquid ventilation limits the post-cardiac arrest syndrome after asphyxial cardiac arrest in rabbits. This protection involves an early limitation in reactive oxidative species production, blood-brain barrier disruption, and delayed preservation against the systemic inflammatory response.


Asunto(s)
Encefalopatías/etiología , Encefalopatías/prevención & control , Paro Cardíaco/complicaciones , Hipotermia Inducida , Ventilación Liquida , Animales , Asfixia/complicaciones , Barrera Hematoencefálica , Paro Cardíaco/etiología , Paro Cardíaco/fisiopatología , Hemodinámica , Hipotermia Inducida/métodos , Ventilación Liquida/métodos , Masculino , Conejos , Distribución Aleatoria , Sepsis/fisiopatología
9.
Arterioscler Thromb Vasc Biol ; 34(5): 1032-44, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24578383

RESUMEN

OBJECTIVE: Cardiovascular dysfunction is a major cause of mortality in patients with sepsis. Recently, we showed that gene deletion or pharmacological inhibition of protein tyrosine phosphatase 1B (PTP1B) improves endothelial dysfunction and reduces the severity of experimental heart failure. However, the cardiovascular effect of PTP1B invalidation in sepsis is unknown. Thus, we explored the beneficial therapeutic effect of PTP1B gene deletion on lipopolysaccharide (LPS)-induced cardiovascular dysfunction, inflammation, and mortality. APPROACH AND RESULTS: PTP1B(-/-) or wild-type mice received LPS (15 mg/kg) or vehicle followed by subcutaneous fluid resuscitation (saline, 30 mL/kg). α-1-dependent constriction and endothelium-dependent dilatation, assessed on isolated perfused mesenteric arteries, were impaired 8 hours after LPS and significantly improved in PTP1B(-/-) mice. This was associated with reduced vascular expression of interleukin1-ß, intercellular adhesion molecule-1, vascular cell adhesion molecule-1, cyclooxygenase-2, and inducible nitric oxide synthase mRNA. PTP1B gene deletion also limited LPS-induced cardiac dysfunction assessed by echocardiography, left ventricular pressure-volume curves, and in isolated perfused hearts. PTP1B(-/-) mice also displayed reduced LPS-induced cardiac expression of tumor necrosis factor-α, interleukin1-ß, intercellular adhesion molecule-1, vascular cell adhesion molecule-1, and Gp91phox, as well as of several markers of cellular infiltration. PTP1B deficiency also reduced cardiac P38 and extracellular signal-regulated protein kinase 1 and 2 phosphorylation and increased phospholamban phosphorylation. Finally, PTP1B(-/-) mice displayed a markedly reduced LPS-induced mortality, an effect also observed using a pharmacological PTP1B inhibitor. PTP1B deletion also improved survival in a cecal ligation puncture model of sepsis. CONCLUSIONS: PTP1B gene deletion protects against septic shock-induced cardiovascular dysfunction and mortality, and this may be the result of the profound reduction of cardiovascular inflammation. PTP1B is an attractive target for the treatment of sepsis.


Asunto(s)
Enfermedades Cardiovasculares/prevención & control , Músculo Liso Vascular/enzimología , Miocardio/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/deficiencia , Sepsis/enzimología , Animales , Presión Sanguínea , Enfermedades Cardiovasculares/enzimología , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/fisiopatología , Ciego/microbiología , Ciego/cirugía , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica , Frecuencia Cardíaca , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Ligadura , Lipopolisacáridos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Arterias Mesentéricas/enzimología , Arterias Mesentéricas/fisiopatología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Músculo Liso Vascular/fisiopatología , NADPH Oxidasa 2 , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Punciones , ARN Mensajero/metabolismo , Sepsis/inducido químicamente , Sepsis/complicaciones , Sepsis/genética , Sepsis/microbiología , Transducción de Señal , Factores de Tiempo , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo , Vasodilatación
10.
J Cardiovasc Pharmacol ; 66(4): 399-408, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26222991

RESUMEN

BACKGROUND: Enhanced heart rate observed in metabolic syndrome (MS) contributes to the deterioration of left ventricular (LV) function via impaired LV filling and relaxation, increased myocardial O2 consumption, and reduced coronary perfusion. However, whether heart rate reduction (HRR) opposes LV dysfunction observed in MS is unknown. METHODS: We assessed in Zucker fa/fa rats, a rat model of MS, the cardiovascular effects of HRR induced by the If current inhibitor S38844 (3 mg · kg(-1) · d(-1)). RESULTS: Delayed short-term (4 days) and long-term (90 days) HRR induced by S38844 reduced LV end-diastolic pressure and LV end-diastolic pressure-volume relation, increased myocardial tissue perfusion, decreased myocardial oxidized glutathione levels, and preserved cardiac output, without modifying LV end-systolic pressure and LV end-systolic pressure-volume relation, although only long-term S38844 opposed LV collagen accumulation. Long-term S38844 improved flow-induced endothelium-dependent dilatation of mesenteric arteries, while metabolic parameters, such as plasma glucose levels, and Hb1c, were never modified. CONCLUSIONS: In rats with MS, HRR induced by the If inhibitor S38844 improved LV diastolic function and endothelium-dependent vascular dilatation, independent from modifications in metabolic status. Moreover, this improvement in cardiac function involves not only immediate effects such as improved myocardial perfusion and reduced oxidative stress but also long-term effects such as modifications in the myocardial structure.


Asunto(s)
Fármacos Cardiovasculares/uso terapéutico , Frecuencia Cardíaca/efectos de los fármacos , Síndrome Metabólico/tratamiento farmacológico , Disfunción Ventricular Izquierda/prevención & control , Función Ventricular Izquierda/efectos de los fármacos , Animales , Fármacos Cardiovasculares/administración & dosificación , Diástole/efectos de los fármacos , Electrocardiografía , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Hemodinámica/efectos de los fármacos , Canales Iónicos/antagonistas & inhibidores , Masculino , Síndrome Metabólico/complicaciones , Síndrome Metabólico/fisiopatología , Estrés Oxidativo/efectos de los fármacos , Ratas Zucker , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/fisiopatología
11.
Prostaglandins Other Lipid Mediat ; 120: 148-54, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26022136

RESUMEN

This study addressed the hypothesis that inhibition of the EETs degrading enzyme soluble epoxide hydrolase affords renal protection in the early stage of diabetic nephropathy. The renal effects of the sEH inhibitor t-AUCB (10mg/l in drinking water) were compared to those of the sulfonylurea glibenclamide (80mg/l), both administered for 8 weeks in FVB mice subjected to a high-fat diet (HFD, 60% fat) for 16 weeks. Mice on control chow diet (10% fat) and non-treated HFD mice served as controls. Compared with non-treated HFD mice, HFD mice treated with t-AUCB had a decreased EET degradation, as shown by their higher plasma EETs-to-DHETs ratio, and an increased EET production, as shown by the increase in EETs+DHETs levels, which was associated with induction of CYP450 epoxygenase expression. Both agents similarly reduced fasting glycemia but only t-AUCB prevented the increase in the urinary albumine-to-creatinine ratio in HFD mice. Histopathological analysis showed that t-AUCB reduced renal inflammation, which was associated with an increased mRNA expression of the NFκB inhibitor Iκ≡ and related decrease in MCP-1, COX2 and VCAM-1 expressions. Finally, there was a marginally significant increase in reactive oxygen species production in HFD mice, together with an enhanced NOX2 expression. Both agents did not modify these parameters but t-AUCB increased the expression of the antioxidant enzyme superoxide dismutase 1. These results demonstrate that, independently from its glucose-lowering effect, sEH inhibition prevents microalbuminuria and renal inflammation in overweight hyperglycemic mice, suggesting that this pharmacological strategy could be useful in the management of diabetic nephropathy.


Asunto(s)
Glucemia/metabolismo , Inhibidores Enzimáticos/farmacología , Epóxido Hidrolasas/antagonistas & inhibidores , Epóxido Hidrolasas/química , Riñón/efectos de los fármacos , Sobrepeso/sangre , Animales , Benzoatos/farmacología , Peso Corporal/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Eicosanoides/metabolismo , Ayuno/sangre , Gliburida/farmacología , Riñón/metabolismo , Riñón/patología , Riñón/fisiopatología , Ratones , Ratones Obesos , Tamaño de los Órganos/efectos de los fármacos , Sobrepeso/metabolismo , Sobrepeso/patología , Sobrepeso/fisiopatología , Estrés Oxidativo/efectos de los fármacos , Solubilidad , Urea/análogos & derivados , Urea/farmacología
12.
Nephrol Dial Transplant ; 29(2): 414-22, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24235073

RESUMEN

BACKGROUND: Middle molecular weight uraemic toxins are considered to play an important role in vascular dysfunction and cardiovascular outcomes in end-stage renal disease (ESRD) patients. Recent dialysis techniques based on convection, specifically high-efficiency on-line haemodiafiltration (HDF), enhance the removal of middle molecular weight toxins and reduce all-cause mortality in haemodialysis (HD) patients. However, the mechanisms of these improved outcomes remain to be established. METHODS: This prospective study randomly assigned 42 ESRD patients to switch from high-flux HD to high-efficiency on-line HDF (n=22) or to continue HD (n=20). Brachial artery endothelium-dependent flow-mediated dilatation, central pulse pressure, carotid artery intima-media thickness (IMT), internal diastolic diameter and distensibility and circulating markers of uraemia, inflammation and oxidative stress were blindly assessed before and after a 4-month follow-up. RESULTS: Brachial flow-mediated dilatation and carotid artery distensibility increased significantly in the HDF group compared with HD, while carotid IMT and diameter remained similar. HDF decreased predialysis levels of the uraemic toxins ß2-microglobulin, phosphate and blood TNFα mRNA expression. Oxidative stress markers were not different between the HD and HDF groups. Blood mRNA expression of protein kinase C ß2, an endothelial NO-synthase (eNOS) inhibitor, decreased significantly with HDF. CONCLUSIONS: High-efficiency on-line HDF prevents the endothelial dysfunction and stiffening of the conduit arteries in ESRD patients compared with high-flux HD. HDF decreases uraemic toxins, vascular inflammation, and is associated with subsequent improvement in eNOS functionality. These results suggest that reduced endothelial dysfunction may be an intermediate mechanism explaining the beneficial outcomes associated with HDF.


Asunto(s)
Endotelio Vascular/fisiopatología , Hemodiafiltración/métodos , Fallo Renal Crónico/terapia , Vasodilatación , Adulto , Anciano , Anciano de 80 o más Años , Derivación Arteriovenosa Quirúrgica , Arteria Braquial/diagnóstico por imagen , Arteria Braquial/fisiopatología , Arterias Carótidas/diagnóstico por imagen , Arterias Carótidas/fisiopatología , Grosor Intima-Media Carotídeo , Endotelio Vascular/diagnóstico por imagen , Femenino , Estudios de Seguimiento , Humanos , Fallo Renal Crónico/fisiopatología , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Diálisis Renal/métodos , Factores de Tiempo
13.
Nat Commun ; 15(1): 5403, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38926363

RESUMEN

Idiopathic inflammatory myopathies (IIMs) are severe autoimmune diseases with poorly understood pathogenesis and unmet medical needs. Here, we examine the role of interferon γ (IFNγ) using NOD female mice deficient in the inducible T cell co-stimulator (Icos), which have previously been shown to develop spontaneous IFNγ-driven myositis mimicking human disease. Using muscle proteomic and spatial transcriptomic analyses we reveal profound myofiber metabolic dysregulation in these mice. In addition, we report muscle mitochondrial abnormalities and oxidative stress in diseased mice. Supporting a pathogenic role for oxidative stress, treatment with a reactive oxygen species (ROS) buffer compound alleviated myositis, preserved muscle mitochondrial ultrastructure and respiration, and reduced inflammation. Mitochondrial anomalies and oxidative stress were diminished following anti-IFNγ treatment. Further transcriptomic analysis in IIMs patients and human myoblast in vitro studies supported the link between IFNγ and mitochondrial dysfunction observed in mice. These results suggest that mitochondrial dysfunction, ROS and inflammation are interconnected in a self-maintenance loop, opening perspectives for mitochondria therapy and/or ROS targeting drugs in myositis.


Asunto(s)
Interferón gamma , Miositis , Estrés Oxidativo , Especies Reactivas de Oxígeno , Animales , Interferón gamma/metabolismo , Miositis/metabolismo , Miositis/patología , Miositis/genética , Humanos , Femenino , Especies Reactivas de Oxígeno/metabolismo , Ratones , Ratones Endogámicos NOD , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Modelos Animales de Enfermedad , Mitocondrias Musculares/metabolismo , Mitocondrias Musculares/patología , Ratones Noqueados , Mioblastos/metabolismo
14.
Therapie ; 79(1): 61-74, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38102030

RESUMEN

Healthcare product procurement accounts for around 50% of the French healthcare system's greenhouse gas emissions. This lesson learned from the publication of the Shift Project's work in November 2021 has been a catalyst within the healthcare system, accelerating the consideration and implementation of actions aimed at reducing the environmental impact of the healthcare system, before, during and after care. In addition to their carbon footprint, healthcare products have a wide range of environmental impacts, including on water, air and soil, throughout their entire life cycle. We have chosen to divide this life cycle into four main stages: from research and development to production, distribution and market access, use and finally end-of-life management. Analysis of the regulatory framework at each stage and of existing initiatives described in the literature or by those in the field have structured and fuelled our thinking. We found that existing regulations focus exclusively on the health risk, with little or no consideration of the environmental risk, which is in itself a health risk. Furthermore, the implementation of certain structuring actions during the first 3 stages of the life cycle would make it possible to simplify or even eliminate the major problem of waste management associated with the end-of-life of healthcare products. With this in mind, we have produced 9 recommendations to ensure that the environmental impact of healthcare products is better taken into account throughout their life cycle.


Asunto(s)
Huella de Carbono , Efecto Invernadero , Humanos , Animales , Atención a la Salud , Estadios del Ciclo de Vida , Muerte
15.
Circulation ; 125(10): 1266-75, 2012 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-22412088

RESUMEN

BACKGROUND: We sought to clarify, using functional and biological approaches, the role of epoxyeicosatrienoic acids, nitric oxide (NO)/reactive oxygen species balance, and endothelin-1 in conduit artery endothelial dysfunction during essential hypertension. METHODS AND RESULTS: Radial artery diameter and mean wall shear stress were determined in 28 untreated patients with essential hypertension and 30 normotensive control subjects during endothelium-dependent flow-mediated dilatation induced by hand skin heating. The role of epoxyeicosatrienoic acids and NO was assessed with the brachial infusion of inhibitors of cytochrome P450 epoxygenases (fluconazole) and NO synthase (N(G)-monomethyl-l-arginine [L-NMMA]). Compared with controls, hypertensive patients exhibited a decreased flow-mediated dilatation in response to postischemic hyperemia as well as to heating, as shown by the lesser slope of their diameter-shear stress relationship. In controls, heating-induced flow-mediated dilatation was reduced by fluconazole, L-NMMA, and, to a larger extent, by L-NMMA+fluconazole. In patients, flow-mediated dilatation was not affected by fluconazole and was reduced by L-NMMA and L-NMMA+fluconazole to a lesser extent than in controls. Furthermore, local plasma epoxyeicosatrienoic acids increased during heating in controls (an effect diminished by fluconazole) but not in patients. Plasma nitrite, an indicator of NO availability, increased during heating in controls (an effect abolished by L-NMMA) and, to a lesser extent, in patients, whereas, inversely, reactive oxygen species increased more in patients (an effect diminished by L-NMMA). Plasma endothelin-1 decreased during heating in controls but not in patients. CONCLUSIONS: These results show that an impaired role of epoxyeicosatrienoic acids contributes, together with an alteration in NO/reactive oxygen species balance and endothelin-1 pathway, to conduit artery endothelial dysfunction in essential hypertension. CLINICAL TRIAL REGISTRATION: https://www.eudract.ema.europa.eu. Unique identifier: RCB2007-A001-10-53.


Asunto(s)
Eicosanoides/metabolismo , Endotelina-1/metabolismo , Endotelio Vascular/metabolismo , Hipertensión/metabolismo , Óxido Nítrico/metabolismo , Inhibidores de 14 alfa Desmetilasa/administración & dosificación , Adulto , Endotelio Vascular/efectos de los fármacos , Inhibidores Enzimáticos/administración & dosificación , Femenino , Fluconazol/administración & dosificación , Calor , Humanos , Hiperemia/metabolismo , Hipertensión/fisiopatología , Masculino , Persona de Mediana Edad , Flujo Pulsátil/efectos de los fármacos , Flujo Pulsátil/fisiología , Arteria Radial/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Piel/irrigación sanguínea , Estrés Mecánico , omega-N-Metilarginina/administración & dosificación
16.
Med Sci (Paris) ; 39(10): 769-775, 2023 Oct.
Artículo en Francés | MEDLINE | ID: mdl-37943138

RESUMEN

Healthy volunteers participating in biomedical research benefit from varying levels of protection in different parts of the world since they are too rarely identified as a specific subset of study participants with specific vulnerabilities and risks. These differences in protection can lead to unfair and ethically unacceptable situations. Healthy volunteers are subject to a number of risks, not only regarding the respect of their rights and of their health but they are also at risk of being exploited because of their financial situation, educational level and motivations. In the end, the scientific validity of the studies may also be called into question. Through its work, the VolREthics (Volunteers in Research and Ethics) initiative, set up by the Inserm ethics committee, outlines the ethical issues raised by the involvement of healthy volunteers in biomedical research, and highlights the need to improve their protection worldwide. Healthy volunteers are essential to scientific progress and society, and their potential vulnerabilities must be recognized and taken into account.


Title: VolREthics - Une initiative internationale de l'Inserm pour définir la protection des volontaires sains. Abstract: Les volontaires sains qui participent aux recherches biomédicales sont très rarement identifiés comme un groupe spécifique. Pourtant, de par leur vulnérabilité et les risques potentiels auxquels ils sont exposés, ils ne bénéficient que d'un niveau de protection qui reste variable selon les régions du monde. Il en résulte différents risques, non seulement pour le respect de leurs droits, de leur santé, mais également pour la validité scientifique des recherches. L'initiative internationale VolREthics (pour volontaires sains en recherche et éthique, ou Volunteers in Research and Ethics), lancée par le comité d'éthique de l'Inserm, a mis en évidence les questions éthiques soulevées par la participation des volontaires sains dans la recherche biomédicale. Elle insiste également sur la nécessité d'améliorer la protection de ces volontaires lors des recherches menées à travers le monde.


Asunto(s)
Investigación Biomédica , Humanos , Voluntarios Sanos , Escolaridad , Motivación
17.
J Mol Cell Cardiol ; 52(3): 660-6, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22155238

RESUMEN

The study addressed the hypothesis that soluble epoxide hydrolase (sEH) inhibition, which increases cardiovascular protective epoxyeicosatrienoic acids (EETs), exerts beneficial effects in an established chronic heart failure (CHF) model. In CHF rats, left ventricular (LV) function, perfusion and remodeling were assessed using MRI and invasive hemodynamics after 42-day (starting 8 days after coronary ligation) and delayed 3-day (starting 47 days after coronary ligation) treatments with the sEH inhibitor AUDA (twice 0.25 mg/day). Delayed 3-day and 42-day AUDA increased plasma EETs demonstrating the effective inhibition of sEH. Delayed 3-day and 42-day AUDA enhanced cardiac output without change in arterial pressure, thus reducing total peripheral resistance. Both treatment periods increased the slope of the LV end-systolic pressure-volume relation, but only 42-day AUDA decreased LV end-diastolic pressure, relaxation constant Tau and the slope of the LV end-diastolic pressure-volume relation, associated with a reduced LV diastolic volume and collagen density. Delayed 3-day and, to a larger extent, 42-day AUDA increased LV perfusion associated with a decreased LV hypoxia-inducible factor-1alpha. Both treatment periods decreased reactive oxygen species level and increased reduced-oxidized glutathione ratio. Finally, MSPPOH, an inhibitor of the EET-synthesizing enzyme cytochrome epoxygenases, abolished the beneficial effects of 3-day AUDA on LV function and perfusion. Augmentation of EET availability by pharmacological inhibition of sEH increases LV diastolic and systolic functions in established CHF. This notably results from short-term processes, i.e. increased LV perfusion, reduced LV oxidative stress and peripheral vasodilatation, but also from long-term effects, i.e. reduced LV remodeling.


Asunto(s)
Circulación Coronaria , Epóxido Hidrolasas/antagonistas & inhibidores , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/fisiopatología , Miocardio/enzimología , Adamantano/administración & dosificación , Adamantano/análogos & derivados , Adamantano/farmacología , Animales , Circulación Coronaria/efectos de los fármacos , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Epóxido Hidrolasas/metabolismo , Insuficiencia Cardíaca/tratamiento farmacológico , Hemodinámica/efectos de los fármacos , Ácidos Láuricos/administración & dosificación , Ácidos Láuricos/farmacología , Masculino , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/enzimología , Infarto del Miocardio/fisiopatología , Óxido Nítrico/metabolismo , Estrés Oxidativo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Transducción de Señal , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos
18.
Neurobiol Dis ; 45(3): 871-86, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22209711

RESUMEN

In industrialized countries, cerebral palsy affects 2.5‰ of preterm and term infants. At a neurochemical level, the massive release of glutamate constitutes a major process leading to excitotoxicity and neonatal brain lesions. Previous studies, conducted in the laboratory, revealed that, in (δ/δ)VEGF(A) transgenic mice, glutamate-induced brain lesions are exacerbated suggesting that VEGF(A) could play a protective action against excitotoxicity. Using a model of cultured cortical brain slices, the aim of the study was to characterize the central effects of VEGF against glutamate-induced excitotoxicity in neonates. Exposure of brain slices to glutamate induced a strong increase of necrotic cell death in the deep cortical layer VI and a decrease of apoptotic death in superficial layers II-IV. When administered alone, a 6-h treatment with VEGF(A) had no effect on both apoptotic and necrotic deaths. In contrast, VEGF(A) abolished the glutamate-induced necrosis observed in layer VI. While MEK and PI3-K inhibitors had no effect on the protective action of VEGF(A), L-NAME, a pan inhibitor of NOS, abrogated the effect of VEGF(A) and exacerbated the excitotoxic action of glutamate. Calcimetry experiments performed on brain slices revealed that VEGF(A) reduced the massive calcium influx induced by glutamate in layer VI and this effect was blocked by L-NAME. Neuroprotective effect of VEGF(A) was also blocked by LNIO and NPLA, two inhibitors of constitutive NOS, while AGH, an iNOS inhibitor, had no effect. Nitrite measurements, electron paramagnetic resonance spectroscopy and immunohistochemistry indicated that glutamate was a potent inducer of NO production via activation of nNOS in the cortical layer VI. In vivo administration of nNOS siRNA promoted excitotoxicity and mimicked the effects of L-NAME, LNIO and NPLA. A short-term glutamate treatment increased nNOS Ser1412 phosphorylation, while a long-term exposure inhibited nNOS/NR2B protein-protein interactions. Altogether, these findings indicate that, in deep cortical layers of mice neonates, glutamate stimulates nNOS activity. Contrasting with mature brain, NO production induced by high concentrations of glutamate is neuroprotective and is required for the anti-necrotic effect of VEGF(A).


Asunto(s)
Apoptosis/efectos de los fármacos , Corteza Cerebral/crecimiento & desarrollo , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Óxido Nítrico/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Análisis de Varianza , Animales , Animales Recién Nacidos , Calcio/metabolismo , Caspasa 3/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Citrulina/metabolismo , Relación Dosis-Respuesta a Droga , Espectroscopía de Resonancia por Spin del Electrón/métodos , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática/métodos , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Glutamato Descarboxilasa/genética , Ácido Glutámico/toxicidad , Proteínas Fluorescentes Verdes/genética , Técnicas In Vitro , L-Lactato Deshidrogenasa/metabolismo , Ratones , Ratones Transgénicos , NADPH Deshidrogenasa/metabolismo , Neuronas/efectos de los fármacos , Óxido Nítrico Sintasa/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , S-Nitroso-N-Acetilpenicilamina/farmacología , Factores de Tiempo
19.
Am J Physiol Heart Circ Physiol ; 300(6): H2035-43, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21441311

RESUMEN

The deleterious effects of aldosterone excess demonstrated in cardiovascular diseases might be linked in part to coronary vascular dysfunction. However, whether such vascular dysfunction is a cause or a consequence of the changes occurring in the cardiomyocytes is unclear. Moreover, the possible link between mineralocorticoid receptor (MR)-mediated effects on the cardiomyocyte and the coronary arteries is unknown. Thus we used a mouse model with conditional, cardiomyocyte-specific overexpression of human MR (hMR) and observed the effects on endothelial function in isolated coronary segments. hMR overexpression decreased the nitric oxide (NO)-mediated relaxing responses to acetylcholine in coronary arteries (but not in peripheral arteries), and this was prevented by a 1-mo treatment either with an MR antagonist, vitamin E/vitamin C, or a NADPH oxidase inhibitor. hMR overexpression did not affect coronary endothelial NO synthase content nor its level of phosphorylation on serine 1177, but increased cardiac levels of reactive oxygen species, cardiac NADPH oxidase (NOX) activity, and expression of the NOX subunit gp91phox, which was limited to endothelial cells. Thus an increase in hMR activation, restricted to cardiomyocytes, is sufficient to induce a severe coronary endothelial dysfunction. We suggest a new paracrine mechanism by which cardiomyocytes trigger a NOX-dependent, reactive oxygen species-mediated coronary endothelial dysfunction.


Asunto(s)
Comunicación Celular/fisiología , Vasos Coronarios/fisiopatología , Endotelio Vascular/fisiopatología , Miocitos Cardíacos/metabolismo , Receptores de Mineralocorticoides/metabolismo , Acetilcolina/farmacología , Animales , Ácido Ascórbico/farmacología , Vasos Coronarios/citología , Endotelio Vascular/citología , Humanos , Ratones , Ratones Transgénicos , Antagonistas de Receptores de Mineralocorticoides , Modelos Animales , Miocitos Cardíacos/citología , Óxido Nítrico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Mineralocorticoides/efectos de los fármacos , Vitamina E/farmacología
20.
Arterioscler Thromb Vasc Biol ; 30(12): 2562-7, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20847304

RESUMEN

OBJECTIVE: To assess the coronary endothelial protective effects of 17ß-estradiol (E2) and the role of estrogen receptor (ER) α in ischemia/reperfusion (I/R). METHODS AND RESULTS: E2 exerts protective effects in cardiac I/R. However, the implication in vivo of the endothelium and the cellular targets of the anti-ischemic effects of E2 are unknown. Mice were subjected to I/R (30 minutes of I and 1 hour of R) in vivo, after which acetylcholine-induced relaxation of isolated coronary segments was assessed ex vivo. I/R induced a coronary endothelial dysfunction in untreated ovariectomized mice that was prevented by long-term treatment with E2 in wild-type, but not in ERα(-/-), mice. Chimeric mice inactivated for ERα in the hematopoietic compartment remained protected by E2. Further inactivation of endothelial ERα abolished the protective action of E2 on coronary endothelial function in Tie2-Cre(+) ERα(f/f) mice. More importantly, E2 significantly limited infarct size in wild-type mice but not in mice deficient in endothelial ERα, even in the presence of hematopoietic ERα. CONCLUSIONS: Endothelial ERα plays a crucial role in the E2-induced prevention of endothelial dysfunction after I/R. To our knowledge, we demonstrate for the first time, by using unique genetically modified mice, that targeting endothelial protection per se can confer cardiomyocyte protection in I/R.


Asunto(s)
Vasos Coronarios/metabolismo , Células Endoteliales/metabolismo , Estradiol/administración & dosificación , Receptor alfa de Estrógeno/metabolismo , Terapia de Reemplazo de Estrógeno , Daño por Reperfusión Miocárdica/prevención & control , Animales , Presión Sanguínea , Trasplante de Médula Ósea , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/fisiopatología , Vasos Coronarios/ultraestructura , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Implantes de Medicamentos , Células Endoteliales/ultraestructura , Receptor alfa de Estrógeno/deficiencia , Receptor alfa de Estrógeno/genética , Femenino , Frecuencia Cardíaca , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Ovariectomía , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo , Quimera por Trasplante , Útero/efectos de los fármacos , Vasodilatación , Vasodilatadores/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA