Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Science ; 356(6335): 233-234, 2017 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-28428376
2.
Am J Physiol Heart Circ Physiol ; 310(1): H71-9, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26519030

RESUMEN

Hydrogen sulfide (H2S) has emerged as an important gasotransmitter in the vasculature. In this study, we tested the hypothesis that H2S contributes to coronary vasoregulation and evaluated the physiological relevance of two sources of H2S, namely, cystathionine-γ-lyase (CSE) and 3-mercaptypyruvate sulfertransferase (MPST). MPST was detected in human coronary artery endothelial cells as well as rat and mouse coronary artery; CSE was not detected in the coronary vasculature. Rat coronary artery homogenates produced H2S through the MPST pathway but not the CSE pathway in vitro. In vivo coronary vasorelaxation response was similar in CSE knockout mice, wild-type mice (WT), and WT mice treated with the CSE inhibitor propargylglycine, suggesting that CSE-produced H2S does not have a significant role in coronary vasoregulation in vivo. Ex vivo, the MPST substrate 3-mercaptopyruvate (3-MP) and H2S donor sodium hydrosulfide (NaHS) elicited similar coronary vasoreactivity responses. Pyruvate did not have any effects on vasoreactivity. The vasoactive effect of H2S appeared to be nitric oxide (NO) dependent: H2S induced coronary vasoconstriction in the presence of NO and vasorelaxation in its absence. Maximal endothelial-dependent relaxation was intact after 3-MP and NaHS induced an increase in preconstriction tone, suggesting that endothelial NO synthase activity was not significantly inhibited. In vitro, H2S reacted with NO, which may, in part explain the vasoconstrictive effects of 3-MP and NaHS. Taken together, these data show that MPST rather than CSE generates H2S in coronary artery, mediating its effects through direct modulation of NO. This has important implications for H2S-based therapy in healthy and diseased coronary arteries.


Asunto(s)
Vasos Coronarios/enzimología , Cistationina gamma-Liasa/metabolismo , Sulfuro de Hidrógeno/metabolismo , Sulfurtransferasas/metabolismo , Animales , Células Cultivadas , Vasos Coronarios/efectos de los fármacos , Cistationina gamma-Liasa/antagonistas & inhibidores , Cistationina gamma-Liasa/deficiencia , Cistationina gamma-Liasa/genética , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Humanos , Cinética , Masculino , Ratones Noqueados , Óxido Nítrico/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
3.
J Vis Exp ; (94)2014 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-25489936

RESUMEN

We present a protocol for measuring in vivo aortic stiffness in mice using high-resolution ultrasound imaging. Aortic diameter is measured by ultrasound and aortic blood pressure is measured invasively with a solid-state pressure catheter. Blood pressure is raised then lowered incrementally by intravenous infusion of vasoactive drugs phenylephrine and sodium nitroprusside. Aortic diameter is measured for each pressure step to characterize the pressure-diameter relationship of the ascending aorta. Stiffness indices derived from the pressure-diameter relationship can be calculated from the data collected. Calculation of arterial compliance is described in this protocol. This technique can be used to investigate mechanisms underlying increased aortic stiffness associated with cardiovascular disease and aging. The technique produces a physiologically relevant measure of stiffness compared to ex vivo approaches because physiological influences on aortic stiffness are incorporated in the measurement. The primary limitation of this technique is the measurement error introduced from the movement of the aorta during the cardiac cycle. This motion can be compensated by adjusting the location of the probe with the aortic movement as well as making multiple measurements of the aortic pressure-diameter relationship and expanding the experimental group size.


Asunto(s)
Aorta/diagnóstico por imagen , Aorta/patología , Rigidez Vascular/fisiología , Animales , Ratones , Ultrasonografía Doppler/métodos
4.
Radiat Res ; 176(4): 474-85, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21787183

RESUMEN

Ionizing radiation has been implicated in the development of significant cardiovascular complications. Since radiation exposure is associated with space exploration, astronauts are potentially at increased risk of accelerated cardiovascular disease. This study investigated the effect of high atomic number, high-energy (HZE) iron-ion radiation on vascular and endothelial function as a model of space radiation. Rats were exposed to a single whole-body dose of iron-ion radiation at doses of 0, 0.5 or 1 Gy. In vivo aortic stiffness and ex vivo aortic tension responses were measured 6 and 8 months after exposure as indicators of chronic vascular injury. Rats exposed to 1 Gy iron ions demonstrated significantly increased aortic stiffness, as measured by pulse wave velocity. Aortic rings from irradiated rats exhibited impaired endothelial-dependent relaxation consistent with endothelial dysfunction. Acute xanthine oxidase (XO) inhibition or reactive oxygen species (ROS) scavenging restored endothelial-dependent responses to normal. In addition, XO activity was significantly elevated in rat aorta 4 months after whole-body irradiation. Furthermore, XO inhibition, initiated immediately after radiation exposure and continued until euthanasia, completely inhibited radiation-dependent XO activation. ROS production was elevated after 1 Gy irradiation while production of nitric oxide (NO) was significantly impaired. XO inhibition restored NO and ROS production. Finally, dietary XO inhibition preserved normal endothelial function and vascular stiffness after radiation exposure. These results demonstrate that radiation induced XO-dependent ROS production and nitroso-redox imbalance, leading to chronic vascular dysfunction. As a result, XO is a potential target for radioprotection. Enhancing the understanding of vascular radiation injury could lead to the development of effective methods to ameliorate radiation-induced vascular damage.


Asunto(s)
Aorta/patología , Endotelio Vascular/enzimología , Endotelio Vascular/efectos de la radiación , Hierro/efectos adversos , Xantina Oxidasa/metabolismo , Animales , Aorta/efectos de los fármacos , Aorta/efectos de la radiación , Fenómenos Biomecánicos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Inhibidores Enzimáticos/farmacología , Masculino , Óxido Nítrico/biosíntesis , Oxipurinol/farmacología , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo , Irradiación Corporal Total/efectos adversos , Xantina Oxidasa/antagonistas & inhibidores
5.
Circ Res ; 107(1): 117-25, 2010 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-20489165

RESUMEN

RATIONALE: Although an age-related decrease in NO bioavailability contributes to vascular stiffness, the underlying molecular mechanisms remain incompletely understood. We hypothesize that NO constrains the activity of the matrix crosslinking enzyme tissue transglutaminase (TG2) via S-nitrosylation in young vessels, a process that is reversed in aging. OBJECTIVE: We sought to determine whether endothelium-dependent NO regulates TG2 activity by S-nitrosylation and whether this contributes to age-related vascular stiffness. METHODS AND RESULTS: We first demonstrate that NO suppresses activity and increases S-nitrosylation of TG2 in cellular models. Next, we show that nitric oxide synthase (NOS) inhibition leads to increased surface and extracellular matrix-associated TG2. We then demonstrate that endothelium-derived bioactive NO primarily mediates its effects through TG2, using TG2(-/-) mice chronically treated with the NOS inhibitor l-N(G)-nitroarginine methyl ester (L-NAME). We confirm that TG2 activity is modulated by endothelium-derived bioactive NO in young rat aorta. In aging rat aorta, although TG2 expression remains unaltered, its activity increases and S-nitrosylation decreases. Furthermore, TG2 inhibition decreases vascular stiffness in aging rats. Finally, TG2 activity and matrix crosslinks are augmented with age in human aorta, whereas abundance remains unchanged. CONCLUSIONS: Decreased S-nitrosylation of TG2 and increased TG activity lead to enhanced matrix crosslinking and contribute to vascular stiffening in aging. TG2 appears to be the member of the transglutaminase family primarily contributing to this phenotype. Inhibition of TG2 could thus represent a therapeutic target for age-associated vascular stiffness and isolated systolic hypertension.


Asunto(s)
Envejecimiento/metabolismo , Endotelio Vascular/enzimología , Proteínas de Unión al GTP/metabolismo , Óxido Nítrico/fisiología , Transglutaminasas/metabolismo , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Envejecimiento/patología , Animales , Células Cultivadas , Endotelio Vascular/patología , Proteínas de Unión al GTP/antagonistas & inhibidores , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Células 3T3 NIH , Óxido Nítrico/antagonistas & inhibidores , Proteína Glutamina Gamma Glutamiltransferasa 2 , Ratas , Ratas Endogámicas F344 , Transglutaminasas/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...