Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Int J Mol Sci ; 23(10)2022 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-35628350

RESUMEN

Hypoxia and inflammation play a major role in revascularization following ischemia. Sildenafil inhibits phosphodiesterase-5, increases intracellular cGMP and induces revascularization through a pathway which remains incompletely understood. Thus, we investigated the effect of sildenafil on post-ischemic revascularization. The left femoral artery was ligated in control and sildenafil-treated (25 mg/kg per day) rats. Vascular density was evaluated and expressed as the left/right leg (L/R) ratio. In control rats, L/R ratio was 33 ± 2% and 54 ± 9%, at 7- and 21-days post-ligation, respectively, and was significantly increased in sildenafil-treated rats to 47 ± 4% and 128 ± 11%, respectively. A neutralizing anti-VEGF antibody significantly decreased vascular density (by 0.48-fold) in control without effect in sildenafil-treated animals. Blood flow and arteriolar density followed the same pattern. In the ischemic leg, HIF-1α and VEGF expression levels increased in control, but not in sildenafil-treated rats, suggesting that sildenafil did not induce angiogenesis. PI3-kinase, Akt and eNOS increased after 7 days, with down-regulation after 21 days. Sildenafil induced outward remodeling or arteriogenesis in mesenteric resistance arteries in association with eNOS protein activation. We conclude that sildenafil treatment increased tissue blood flow and arteriogenesis independently of VEGF, but in association with PI3-kinase, Akt and eNOS activation.


Asunto(s)
Miembro Posterior , Isquemia , Óxido Nítrico Sintasa de Tipo III , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Citrato de Sildenafil , Animales , Miembro Posterior/irrigación sanguínea , Miembro Posterior/efectos de los fármacos , Miembro Posterior/metabolismo , Isquemia/tratamiento farmacológico , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Transducción de Señal , Citrato de Sildenafil/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo
2.
Oxid Med Cell Longev ; 2019: 2567198, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31214278

RESUMEN

Vascular aging is characterized by functional and structural changes of the vessel wall, including endothelial dysfunction, with decreased endothelial NO· bioavailability and elevated vasoconstrictor and inflammatory mediator production, vascular rigidity, and tone impairment. Moringa oleifera (MOI) is a little tree, and different parts of which are used in traditional medicine in tropical Africa, America, and Asia for therapeutic applications in several disorders including cardiovascular disease. The present study is aimed at assessing the effect of MOI on aging-associated alteration of the endothelial function in Wistar rats. Middle-aged Wistar rats (46-week-old males) have been fed with food containing or not 750 mg/kg/day of MOI seed powder for 4 weeks. A group of young Wistar rats (16-week-old) was used as control. Measurement of isometric contraction, western blot analysis, and immunostaining has then been performed in the aortas and mesenteric arteries to assess the endothelium function. MOI treatment improved carbachol-induced relaxation in both aortas and mesenteric arteries of middle-aged rats. In the aortas, this was associated with an increased Akt signalling and endothelial NO synthase activation and a downregulation of arginase-1. In the mesenteric arteries, the improvement of the endothelial-dependent relaxation was related to an EDHF-dependent mechanism. These results suggest a vascular protective effect of MOI seeds against the vascular dysfunction that develops during aging through different mechanisms in conductance and resistance arteries.


Asunto(s)
Envejecimiento/fisiología , Aorta/patología , Enfermedades Cardiovasculares/terapia , Endotelio/fisiología , Moringa oleifera , Animales , Células Cultivadas , Suplementos Dietéticos , Humanos , Masculino , Contracción Miocárdica , Óxido Nítrico Sintasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Semillas , Transducción de Señal , Vasodilatación
3.
Oxid Med Cell Longev ; 2017: 4129459, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28713487

RESUMEN

Moringa oleifera (MOI) is a tree currently used in traditional medicine in tropical Africa, America, and Asia for therapeutic applications in several disorders including arterial hypertension. We previously described a cardiac protective role of a treatment with MOI seeds in spontaneously hypertensive rats (SHR). Here, we investigated the effects of this treatment on oxidative and nitrosative vascular stresses in SHR, with normotensive Wistar Kyoto rats used as controls. Oxidative and nitrosative stresses detected in SHR aortas using the fluorescent dye dihydroethidine and protein nitrotyrosine staining were reduced in MOI-treated SHR aortas. This was associated with a decrease of free 8-isoprostane circulating level, vascular p22phox and p47phox expressions, and SOD2 upregulation. Moreover, circulating nitrites and C-reactive protein, increased in SHR, were both reduced in SHR receiving MOI. This was associated to decrease iNOS and NF-κB protein expressions after MOI treatment. In functional studies, the endothelium-dependent carbachol-induced relaxation was improved in MOI-treated SHR resistance arteries. Oral administration of MOI seeds demonstrates vascular antioxidant, anti-inflammatory, and endothelial protective effects in SHR. Our data support the use of MOI seeds in diet against cardiovascular disorders associated with oxidative stress and inflammation such as hypertension, scientifically validating the use of these seeds in Malagasy traditional medicine.


Asunto(s)
Hipertensión/tratamiento farmacológico , Moringa oleifera/metabolismo , Animales , Antioxidantes/metabolismo , Masculino , Moringa oleifera/citología , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Endogámicas WKY
4.
Am J Hypertens ; 29(7): 873-81, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26864583

RESUMEN

BACKGROUND: Hypertension is characterized by a maintained high blood pressure leading to cardiac complications such as left ventricular hypertrophy and fibrosis and an increased risk of heart failure and myocardial infarction. This study investigated the cardiac effects of oral administration of Moringa oleifera (MOI) seed powder in spontaneous hypertensive rats (SHR). METHODS: SHR received food containing MOI seed powder (750mg/d, 8 weeks) or normal food. In vivo measurement of hemodynamic parameters by telemetry and cardiac structure and function analysis by echocardiography were performed. Histological studies were performed to determine fibrosis and protein expression. RESULTS: MOI treatment did not modify blood pressure in SHR but reduced nocturnal heart rate and improved cardiac diastolic function (reduction of isovolumetric relaxation time and deceleration time of the E wave, increase of ejection volume and cardiac output compared to nontreated SHR). Left ventricular anterior wall thickness, interseptal thickness on diastole, and relative wall thickness were reduced after MOI treatment. Furthermore, we found a significant reduction of fibrosis in the left ventricle of MOI-treated SHR. This antihypertrophic and antifibrotic effect of MOI was associated with increased expression of peroxisome proliferator-activated receptor (PPAR)-α and δ, reduced cardiac triglyceride level, and enhanced plasmatic prostacyclins. CONCLUSIONS: Our data show a beneficial effect of MOI on the cardiac structure and function in SHR associated with an upregulation of PPAR-α and δ signaling. This study thus provides scientific rational support for the empirical use of MOI in the traditional Malagasy medicine against cardiac diseases associated with blood pressure overload.


Asunto(s)
Cardiopatías/prevención & control , Hipertensión/complicaciones , Moringa oleifera , Fitoterapia , Preparaciones de Plantas/uso terapéutico , Animales , Presión Sanguínea , Ecocardiografía , Cardiopatías/etiología , Frecuencia Cardíaca , Metabolismo de los Lípidos , Masculino , Miocardio/metabolismo , PPAR alfa/metabolismo , PPAR delta/metabolismo , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Semillas
5.
PLoS One ; 10(9): e0137408, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26355997

RESUMEN

BACKGROUND: Hypertension is one of the main risk factors of cardiovascular diseases. In Madagascar, studies on hypertension in urban and rural communities are scarce. OBJECTIVES: The aim of this study was to determine the prevalence of hypertension and identify associated risk factors in adults living in a health and demographic system in Moramanga, Madagascar. METHODS: The study included people aged 15 years old and above living in a health and demographic system in Moramanga. A household census was performed in 2012 to enumerate the population in 3 communities in Moramanga. In addition to the questionnaire used in the initial census, a standardized questionnaire and blood pressure were taken twice after 5 and 10 minutes of rest. In urban areas, heights and weights were also measured to calculate the body mass index. RESULTS: There were 3621 and 4010 participants respectively in rural and urban areas. Prevalence of hypertension in rural population was 27.0% (IC95% [25.6-28.5]) and 29.7% (IC95% [28.3-31.1]) in urban population. Among hypertensive subjects, 1.7% (17/979) and 5.3% (64/1191) were on antihypertensive treatment for at least 1 month before the survey in rural and urban population, respectively. In rural areas, increasing age (65 years and older vs 18-25 years OR = 11.81, IC95% [7.79-18.07]), giving more than 3 positive responses to the usual risks factors of hypertension (OR = 1.67, IC95% [1.14-2.42]) and singles in comparison with married people (OR = 1.61, IC95% [1.20-2.17]) were associated to hypertension in a logistic regression model. In urban areas, increasing age (65 years and older vs 18-25 years OR = 37.54, IC95% [24.81-57.92]), more than 3 positive responses to the usual risks of hypertension (OR = 3.47, IC95% [2.58-4.67]) and obesity (OR = 2.45, IC95% [1.56-3.87]) were found as risk factors. CONCLUSION: Hypertension is highly prevalent in rural areas although it is significantly less treated. As a result, a major epidemic of cardiovascular diseases is at risk in Madagascar's progressively aging society.


Asunto(s)
Hipertensión/epidemiología , Enfermedades Desatendidas/epidemiología , Población Rural/estadística & datos numéricos , Población Urbana/estadística & datos numéricos , Adulto , Antihipertensivos/uso terapéutico , Demografía , Femenino , Humanos , Hipertensión/tratamiento farmacológico , Madagascar/epidemiología , Masculino , Prevalencia , Factores de Riesgo
6.
Hypertension ; 65(6): 1273-8, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25870189

RESUMEN

Although a causative role for RhoA-Rho kinase has been recognized in the development of human hypertension, the molecular mechanism(s) and the RhoA guanine exchange factor(s) responsible for the overactivation of RhoA remain unknown. Arhgef1 was identified as a RhoA guanine exchange factor involved in angiotensin II (Ang II)-mediated regulation of vascular tone and hypertension in mice. The aim of this study was to determine whether Arhgef1 is activated and involved in the activation of RhoA-Rho kinase signaling by Ang II in humans. In vitro stimulation of human coronary artery smooth muscle cells and human peripheral blood mononuclear cells by Ang II (0.1 µmol/L) induced activation of Arhgef1 attested by its increased tyrosine phosphorylation. Silencing of Arhgef1 expression by siRNA inhibited Ang II-induced activation of RhoA-Rho kinase signaling. In normotensive subjects, activation of the renin-angiotensin system by a low-salt diet for 7 days increased RhoA-Rho kinase signaling and stimulated Arhgef1 activity in peripheral blood mononuclear cells. In conclusion, our results strongly suggest that Arhgef1 mediates Ang II-induced RhoA activation in humans. Moreover, they show that measurement of RhoA guanine exchange factor activity in peripheral blood mononuclear cells might be a useful method to evaluate RhoA guanine exchange factor activity in humans.


Asunto(s)
Angiotensina II/farmacología , Leucocitos Mononucleares/metabolismo , Músculo Liso Vascular/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Western Blotting , Células Cultivadas , Humanos , Hipertensión/tratamiento farmacológico , Hipertensión/fisiopatología , Leucocitos Mononucleares/efectos de los fármacos , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , ARN Mensajero/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/efectos de los fármacos , Transducción de Señal , Estadísticas no Paramétricas , Proteína de Unión al GTP rhoA/efectos de los fármacos
7.
Small GTPases ; 5(4): 1-10, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25496262

RESUMEN

Proper regulation of arterial blood pressure is essential to allow permanent adjustment of nutrient and oxygen supply to organs and tissues according to their need. This is achieved through highly coordinated regulation processes controlling vascular resistance through modulation of arterial smooth muscle contraction, cardiac output, and kidney function. Members of the Rho family of small GTPases, in particular RhoA and Rac1, have been identified as key signaling molecules playing important roles in several different steps of these regulatory processes. Here, we review the current state of knowledge regarding the involvement of Rho GTPase signaling in the control of blood pressure and the pathogenesis of hypertension. We describe how knockout models in mouse, genetic, and pharmacological studies in human have been useful to address this question.


Asunto(s)
Hipertensión/patología , Proteínas de Unión al GTP rho/metabolismo , Animales , Presión Sanguínea , Sistema Nervioso Central/metabolismo , Humanos , Riñón/metabolismo , Músculo Liso Vascular/fisiología , Óxido Nítrico/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
8.
Fitoterapia ; 96: 65-75, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24731922

RESUMEN

Advanced glycation end-products (AGEs) are associated with many pathogenic disorders such as pathogenesis of diabetes or endothelial dysfunction leading to cardiovascular events. Therefore, the identification of new anti-AGE molecules or extracts aims at preventing such pathologies. Many Clusiaceae and Calophyllaceae species are used in traditional medicines to treat arterial hypertension as well as diabetes. Focusing on these plant families, an anti-AGE plant screening allowed us to select Mammea neurophylla for further phytochemical and biological studies. Indeed, both DCM and MeOH stem bark extracts demonstrated in vitro their ability to prevent inflammation in endothelial cells and to reduce vasoconstriction. A bioguided fractionation of these extracts allowed us to point out 4-phenyl- and 4-(1-acetoxypropyl)coumarins and procyanidins as potent inhibitors of AGE formation, potentially preventing endothelial dysfunction. The fractionation steps also led to the isolation of two new compounds, namely neurophyllols A and B from the DCM bark extract together with thirteen known mammea A and E coumarins (mammea A/AA, mammea A/AB, mammea A/BA, mammea A/BB, mammea A/AA cycloD, mammea A/AB cycloD, disparinol B, mammea A/AB cycloE, ochrocarpin A, mammea A/AA cycloF, mammea A/AB cycloF, mammea E/BA, mammea E/BB) as well as δ-tocotrienol, xanthones (1-hydroxy-7-methoxyxanthone, 2-hydroxyxanthone) and triterpenes (friedelin and betulinic acid). During this study, R,S-asperphenamate, previously described from fungal origin was also purified.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Biflavonoides/farmacología , Catequina/farmacología , Cumarinas/farmacología , Productos Finales de Glicación Avanzada/efectos de los fármacos , Mammea/química , Extractos Vegetales/farmacología , Proantocianidinas/farmacología , Animales , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/aislamiento & purificación , Biflavonoides/química , Biflavonoides/aislamiento & purificación , Catequina/química , Catequina/aislamiento & purificación , Supervivencia Celular/efectos de los fármacos , Cumarinas/química , Cumarinas/aislamiento & purificación , Células Endoteliales , Frutas/química , Masculino , Estructura Molecular , Triterpenos Pentacíclicos , Corteza de la Planta/química , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Hojas de la Planta/química , Proantocianidinas/química , Proantocianidinas/aislamiento & purificación , Ratas , Ratas Wistar , Triterpenos/química , Triterpenos/aislamiento & purificación , Triterpenos/farmacología , Xantonas/química , Xantonas/aislamiento & purificación , Xantonas/farmacología , Ácido Betulínico
9.
Physiol Rev ; 93(4): 1659-720, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24137019

RESUMEN

Small G proteins exist in eukaryotes from yeast to human and constitute the Ras superfamily comprising more than 100 members. This superfamily is structurally classified into five families: the Ras, Rho, Rab, Arf, and Ran families that control a wide variety of cell and biological functions through highly coordinated regulation processes. Increasing evidence has accumulated to identify small G proteins and their regulators as key players of the cardiovascular physiology that control a large panel of cardiac (heart rhythm, contraction, hypertrophy) and vascular functions (angiogenesis, vascular permeability, vasoconstriction). Indeed, basal Ras protein activity is required for homeostatic functions in physiological conditions, but sustained overactivation of Ras proteins or spatiotemporal dysregulation of Ras signaling pathways has pathological consequences in the cardiovascular system. The primary object of this review is to provide a comprehensive overview of the current progress in our understanding of the role of small G proteins and their regulators in cardiovascular physiology and pathologies.


Asunto(s)
Enfermedades Cardiovasculares/fisiopatología , Fenómenos Fisiológicos Cardiovasculares , Proteínas de Unión al GTP Monoméricas/fisiología , Animales , Humanos , Modelos Animales , Proteínas de Unión al GTP Monoméricas/química , Transducción de Señal/fisiología , Proteínas ras/fisiología
10.
Am J Physiol Cell Physiol ; 302(9): C1394-404, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22322975

RESUMEN

Sustained overactivation of RhoA is a common component for the pathogenesis of several cardiovascular disorders, including hypertension. Although activity of Rho proteins depends on Rho exchange factors (Rho-GEFs), the identity of Rho-GEFs expressed in vascular smooth muscle cells (VSMC) and participating in the control of Rho protein activity and Rho-dependent functions remains unknown. To address this question, we analyzed by quantitative RT-PCR the expression profile of 28 RhoA-GEFs in arteries of normotensive (saline-treated) and hypertensive (ANG II-treated) rats. Sixteen RhoA-GEFs were downregulated in mesenteric arteries of hypertensive rats, among which nine are also downregulated in cultured VSMC stimulated by ANG II (100 nM, 48 h), suggesting a direct effect of ANG II. Inhibition of type 1 ANG II receptors (losartan, 1 µM) or Rho kinase (fasudil, 10 µM) prevented ANG II-induced RhoA-GEF downregulation. Functionally, ANG II-induced downregulation of RhoA-GEFs is associated with decreased Rho kinase activation in response to endothelin-1, norepinephrine, and U-46619. This work thus identifies a group of RhoA-GEFs that controls RhoA and RhoA-dependent functions in VSMC, and a negative feedback of RhoA/Rho kinase activity on the expression of these RhoA-GEFs that may play an adaptative role to limit RhoA/Rho kinase activation.


Asunto(s)
Retroalimentación Fisiológica/fisiología , Factores de Intercambio de Guanina Nucleótido/biosíntesis , Hipertensión/fisiopatología , Músculo Liso Vascular/metabolismo , Quinasas Asociadas a rho/metabolismo , Angiotensina II/metabolismo , Angiotensina II/toxicidad , Animales , Arterias/metabolismo , Western Blotting , Perfilación de la Expresión Génica , Hipertensión/inducido químicamente , Masculino , Músculo Liso Vascular/fisiopatología , ARN Interferente Pequeño , Ratas , Ratas Endogámicas WKY , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología , Transfección
11.
Arterioscler Thromb Vasc Biol ; 31(11): 2634-42, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21852563

RESUMEN

OBJECTIVE: Estradiol (E2) mediates numerous beneficial effects assigned to estrogens, but whereas mechanisms have been described at the endothelial level, direct effects on vascular smooth muscle cells (VSMC) are poorly documented. As evidence accumulates regarding the role of RhoA in vascular pathophysiology and the benefit of RhoA-Rho associated protein kinase (Rock) pathway inhibition, we analyzed if E2 could inhibit it in VSMC. METHODS AND RESULTS: We show that in VSMC, E2 inhibits the RhoA-Rock pathway in a time- and concentration-dependent manner. The inhibition of RhoA-Rock pathway results from E2-induced phosphorylation of the Ser188 of RhoA. Using pharmacological, transfection, and in vitro phosphorylation experiments, we demonstrate that AMP-activated protein kinase subunit alpha 1 (AMPKα1) is activated by estrogen receptor stimulation and catalyzes RhoA phosphorylation induced by E2. Ex vivo, ovariectomy leads to an increase in the amplitude of phenylephrine- or serotonine-induced contractions of aortic rings in wild-type mice but not in AMPKα1-knock-out mice or E2-supplemented animals. These functional effects were correlated with a reduced level of RhoA phosphorylation in the aorta of ovariectomized female, male, and AMPKα1 knock-out mice. CONCLUSION: Our work thus defines AMPKα1 as (1) a new kinase for RhoA and (2) a new mediator of the vasoprotective effects of estrogen.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Estradiol/farmacología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Ratones , Ratones Noqueados , Modelos Animales , Músculo Liso Vascular/citología , Ovariectomía , Fosforilación/efectos de los fármacos , Receptores de Estrógenos/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Vasoconstricción/fisiología , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA
12.
Circulation ; 122(19): 1937-47, 2010 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-20974998

RESUMEN

BACKGROUND: Pulmonary hypertension (PH) is among the complications of HIV infection. Combination antiretroviral therapy may influence the progression of HIV-related PH. Because Akt signaling is a potential molecular target of HIV protease inhibitors (HPIs), we hypothesized that these drugs altered monocrotaline- and hypoxia-induced PH in rats by downregulating the Akt pathway, thereby inhibiting pulmonary artery smooth muscle cell proliferation. METHODS AND RESULTS: Daily treatment with each of 3 first-generation HPIs (ritonavir 30 mg/kg, amprenavir 100 mg/kg, and nelfinavir 500 mg/kg) started 3 weeks after a subcutaneous monocrotaline injection (60 mg/kg) substantially diminished pulmonary artery pressure, right ventricular hypertrophy, number of muscularized pulmonary vessels, pulmonary arterial wall thickness, and proliferating pulmonary vascular Ki67-labeled cells without affecting vessel caspase 3 staining. HPI treatment partially prevented the development of hypoxia- and monocrotaline-induced PH. Monocrotaline-induced PH was associated with marked activation of Akt signaling in the lungs and proximal pulmonary arteries, with increases in phosphorylated Akt, phosphorylated glycogen-synthase-kinase-3ß (GSK3), and phosphorylated endothelial nitric oxide synthase, all of which decreased markedly after treatment with each HPI. In contrast, PH-associated increases in phosphorylated extracellular signal-related kinase 1/2 and myosin light-chain phosphatase were unaltered by the HPIs. The 3 HPIs and the phosphatidylinositol 3-kinase inhibitor LY294002 inhibited platelet-derived growth factor-induced phosphorylation of Akt and GSK3 in cultured pulmonary artery smooth muscle cells and blocked cell proliferation; this last effect was abolished by the GSK3 inhibitor SB216763. CONCLUSION: These results support an effect of HPIs on pulmonary vascular remodeling mediated by inhibition of Akt phosphorylation and consequently of pulmonary artery smooth muscle cell proliferation.


Asunto(s)
Inhibidores de la Proteasa del VIH/farmacología , Hipertensión Pulmonar/fisiopatología , Animales , Animales Recién Nacidos , Terapia Antirretroviral Altamente Activa , Antivirales/farmacología , Presión Sanguínea/efectos de los fármacos , Carbamatos/farmacología , División Celular/efectos de los fármacos , Furanos , Hemodinámica/efectos de los fármacos , Hemodinámica/fisiología , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/mortalidad , Hipoxia , Masculino , Monocrotalina , Nelfinavir/farmacología , Arteria Pulmonar/citología , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/fisiopatología , Ratas , Ratas Wistar , Ritonavir/farmacología , Sulfonamidas/farmacología
13.
Nat Rev Cardiol ; 7(11): 637-47, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20808285

RESUMEN

Arterial hypertension is a common health problem that affects 25% of the adult population in industrialized societies, and is a major risk factor for myocardial infarction and stroke. However, the pathogenesis of hypertension, as well as the basic mechanisms of blood-pressure control, are insufficiently understood. Although the development of hypertension is complex, involving many different mechanisms, including dysregulation of the autonomic nervous system, renal function, and the balance between water and electrolytes, and increased vascular tone and the resulting rise in peripheral vascular resistance are major determinants of the elevated arterial pressure in hypertension. Since the discovery of the essential role of RhoA and its downstream target, Rho kinase, in the regulation of vascular tone, as well as the antihypertensive effect of a Rho kinase inhibitor, much evidence has accumulated to implicate activation of Rho family proteins in the pathogenesis of hypertension. RhoA remains the most-analyzed member of the Rho proteins in the context of vascular physiology and hypertension, but evidence is accumulating that also points to a role of Rac1 in arterial pathophysiology. In this Review, we discuss progress in our understanding of the role of Rho proteins and their regulators in the pathogenesis of high blood pressure.


Asunto(s)
Hipertensión/fisiopatología , Músculo Liso Vascular/patología , Transducción de Señal/fisiología , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/metabolismo , Endotelio Vascular/enzimología , Endotelio Vascular/patología , Humanos , Hipertensión/enzimología , Hipertensión Pulmonar/enzimología , Hipertensión Pulmonar/fisiopatología , Músculo Liso Vascular/enzimología , Factores de Riesgo , Proteína de Unión al GTP rac1/metabolismo
14.
Mol Cell Biol ; 30(20): 4786-96, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20696841

RESUMEN

Although overactivation of RhoA is recognized as a common component of vascular disorders, the molecular mechanisms regulating RhoA activity in vascular smooth muscle cells (VSMC) are still unclear. We have previously shown that in VSMC, RhoA is phosphorylated on Ser188 by nitric oxide (NO)-stimulated cGMP-dependent kinase (PKG), which leads to RhoA-Rho kinase pathway inhibition. In this study, we showed that expression of phosphoresistant RhoA mutants prevented the stimulation of VSMC migration and adhesion induced by NO-PKG pathway activation. In contrast, under basal conditions, phosphomimetic RhoA mutants stimulated VSMC adhesion and migration through a signaling pathway requiring Rac1 and the Rho exchange factor Vav3. RhoA phosphorylation or phosphomimetic RhoA mutants induced Rac1 activation but did not activate Vav3. Indeed, phosphorylated RhoA or phosphomimetic mutants trapped guanine dissociation inhibitor α (GDIα), leading to the release of Rac1 and its translocation to the membrane, where it was then activated by the basal Vav3 nucleotide exchange activity. In vivo, RhoA phosphorylation induced by PKG activation in the aortas of rats treated with sildenafil induced dissociation of Rac1 from GDIα and activation of the Rac1 signaling pathway. These results suggest that the phosphorylation of RhoA represents a novel potent and physiological GDIα displacement factor that leads to Rac1 activation and regulation of Rac1-dependent VSMC functions.


Asunto(s)
Inhibidores de Disociación de Guanina Nucleótido/metabolismo , Miocitos del Músculo Liso/fisiología , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Secuencia de Bases , Adhesión Celular/fisiología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Cultivadas , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacología , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Óxido Nítrico/metabolismo , Fosforilación , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-vav/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-vav/genética , Proteínas Proto-Oncogénicas c-vav/metabolismo , ARN Interferente Pequeño/genética , Ratas , Transducción de Señal , Tionucleótidos/farmacología , Cicatrización de Heridas/fisiología , Proteína de Unión al GTP rhoA/química , Proteína de Unión al GTP rhoA/genética
16.
Circ Cardiovasc Interv ; 3(2): 120-6, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20197512

RESUMEN

BACKGROUND: The use of a drug-eluting stent (DES) has strongly limited the incidence of in-stent restenosis in bifurcation lesions; nevertheless, restenosis still remains a problem at the origin of the bifurcation side branch. The aim of this study is to analyze the consequences of the kissing postdilatation technique on 5 DESs, using microfocus x-ray computerized tomography and scanning electron microscopy. METHODS AND RESULTS: Five different DESs (Cypher, Cypher Select, Endeavor, Taxus Express, and Taxus Liberté) were deployed using kissing postdilatation protocols in a bench-top model. For all types of DES, microfocus x-ray computerized tomography analysis showed that (1) kissing postdilatation of the stent by 2 coaxial balloons caused elliptic deformation in the proximal segment and (2) kissing postdilatation technique reduced the ratio of potential metal to artery (manufacturer's data/calculated ratio [%]: Cypher, 12.7/8.8; Cypher Select, 13.5/10.2; Endeavor, 19.0/13.3; Taxus Express, 20.5/4.7; Taxus Liberté, 17.9/12.5) and the potential drug application to area in the proximal segment, including the ostial struts (struts adjacent to and lying around the side branch ostium) (manufacturer's data/calculated drug application [microg/mm(2)]: Cypher, 1.4/1.0; Cypher Select, 1.4/1.1; Endeavor, 1.6/1.1; Taxus Express, 1.0/0.7; Taxus Liberté, 1.0/0.7). Scanning electron microscopy analysis showed a significantly greater coating damage to the ostial struts in all stents evaluated (P<0.05). CONCLUSIONS: Commercially available DESs subjected to simultaneous kissing balloon postdilatation in an unconstrained model may contribute to side branch ostial restenosis by proximal segment elliptic deformation and damage to the polymer coating.


Asunto(s)
Angioplastia Coronaria con Balón , Estenosis Coronaria/terapia , Vasos Coronarios/patología , Stents Liberadores de Fármacos/efectos adversos , Angioplastia Coronaria con Balón/efectos adversos , Angioplastia Coronaria con Balón/instrumentación , Angioplastia Coronaria con Balón/métodos , Reestenosis Coronaria/etiología , Estenosis Coronaria/diagnóstico por imagen , Estenosis Coronaria/patología , Estenosis Coronaria/fisiopatología , Vasos Coronarios/crecimiento & desarrollo , Vasos Coronarios/cirugía , Humanos , Microscopía Electrónica de Rastreo , Neovascularización Patológica , Polímeros , Tomografía Computarizada por Rayos X
17.
Nat Med ; 16(2): 183-90, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20098430

RESUMEN

Hypertension is one of the most frequent pathologies in the industrialized world. Although recognized to be dependent on a combination of genetic and environmental factors, its molecular basis remains elusive. Increased activity of the monomeric G protein RhoA in arteries is a common feature of hypertension. However, how RhoA is activated and whether it has a causative role in hypertension remains unclear. Here we provide evidence that Arhgef1 is the RhoA guanine exchange factor specifically responsible for angiotensin II-induced activation of RhoA signaling in arterial smooth muscle cells. We found that angiotensin II activates Arhgef1 through a previously undescribed mechanism in which Jak2 phosphorylates Tyr738 of Arhgef1. Arhgef1 inactivation in smooth muscle induced resistance to angiotensin II-dependent hypertension in mice, but did not affect normal blood pressure regulation. Our results show that control of RhoA signaling through Arhgef1 is central to the development of angiotensin II-dependent hypertension and identify Arhgef1 as a potential target for the treatment of hypertension.


Asunto(s)
Angiotensina II/fisiología , Presión Sanguínea/fisiología , Factores de Intercambio de Guanina Nucleótido/fisiología , Proteínas Proto-Oncogénicas/fisiología , Animales , Factores de Intercambio de Guanina Nucleótido/química , Factores de Intercambio de Guanina Nucleótido/metabolismo , Hipertensión/etiología , Hipertensión/fisiopatología , Janus Quinasa 2/metabolismo , Ratones , Ratones Noqueados , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiología , Fosforilación , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho , Transducción de Señal , Tirosina/metabolismo
18.
Cardiovasc Res ; 86(1): 131-40, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19969623

RESUMEN

AIMS: Rho guanine nucleotide exchange factors (Rho GEFs) are responsible for Rho protein activation by catalyzing the exchange of GDP for GTP. Although overactivation of Rho proteins is a common component of the pathogenesis of vascular disorders, the molecular mechanisms and the Rho GEFs regulating Rho protein activity and Rho-dependent functions in vascular smooth muscle cells (VSMC) are still unknown. The aim of this study was thus to identify Rho GEFs involved in the regulation of VSMC functions. METHODS AND RESULTS: By a functional screening based on small interfering RNA (siRNA)-mediated silencing of 27 Rho GEFs, we found that only silencing of the Rho GEF Vav3 inhibited rat VSMC proliferation. Conversely, overexpression of Vav3 potentiated VSMC proliferation, whereas the catalytically inactive Vav3 mutant had no effect. The stimulatory effect of Vav3 on VSMC proliferation was inhibited by the Src tyrosine kinase inhibitor SU6656 and by co-expression of the dominant-negative Rac1-N17 mutant. In agreement with this observation, expression of Vav3 induced enrichment of Rac1 to the membrane, activation of its effector PAK, and stimulated VSMC migration. Increased levels of Vav3 transcripts were found in stented arteries and arteries from hypertensive rats. Furthermore, siRNA targeting Vav3 decreased arterial wall cell proliferation. CONCLUSION: The Rho GEF Vav3 controls VSMC proliferation and migration through activation of Rac1/PAK signalling. Vav3 is a convergent point mediating Rac1 activation in response to different upstream mediators that promote VSMC proliferation and migration and thus appears to be a new potential therapeutic target that could be used to limit vascular proliferative diseases.


Asunto(s)
Movimiento Celular/fisiología , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiología , Proteínas Proto-Oncogénicas c-vav/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Aorta/citología , Aorta/fisiología , División Celular/fisiología , Células Cultivadas , Inhibidores Enzimáticos/farmacología , NG-Nitroarginina Metil Éster/farmacología , Proteínas Proto-Oncogénicas c-vav/genética , ARN Interferente Pequeño , Ratas , Ratas Endogámicas WKY , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Quinasas p21 Activadas/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteínas de Unión al GTP rho/genética
19.
Am J Physiol Cell Physiol ; 297(5): C1062-70, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19692654

RESUMEN

Angiotensin II (ANG II) is a major regulator of blood pressure that essentially acts through activation of ANG II type 1 receptor (AT1R) of vascular smooth muscle cells (VSMC). AT1R activates numerous intracellular signaling pathways, including the small G protein RhoA known to control several VSMC functions. Nevertheless, the mechanisms leading to RhoA activation by AT1R are unknown. RhoA activation can result from activation of RhoA exchange factor and/or inhibition of Rho GTPase-activating protein (GAP). Here we hypothesize that a RhoGAP could participate to RhoA activation induced by ANG II in rat aortic VSMC. The knockdown of the RhoGAP p190A by small interfering RNA (siRNA) abolishes the activation of RhoA-Rho kinase pathway induced after 5 min of ANG II (0.1 microM) stimulation in rat aortic VSMC. We then show that AT1R activation induces p190A dephosphorylation and inactivation. In addition, expression of catalytically inactive or phosphoresistant p190A mutants increases the basal activity of RhoA-Rho kinase pathway, whereas phosphomimetic mutant inhibits early RhoA activation by ANG II. Using siRNA and mutant overexpression, we then demonstrate that the tyrosine phosphatase SHP2 is necessary for 1) maintaining p190A basally phosphorylated and activated by the tyrosine kinase c-Abl, and 2) inducing p190A dephosphorylation and RhoA activation in response to AT1R activation. Our work then defines p190A as a new mediator of RhoA activation by ANG II in VSMC.


Asunto(s)
Angiotensina II/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Músculo Liso Vascular/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Western Blotting , Activación Enzimática/fisiología , Inmunoprecipitación , Miocitos del Músculo Liso/metabolismo , Fosforilación , ARN Interferente Pequeño , Ratas , Receptor de Angiotensina Tipo 1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología , Quinasas Asociadas a rho/metabolismo
20.
Am J Respir Crit Care Med ; 179(12): 1151-8, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19299501

RESUMEN

RATIONALE: The complex and multifactorial pathogenesis of pulmonary hypertension (PH) involves constriction, remodeling, and in situ thrombosis of pulmonary vessels. Both serotonin (5-HT) and Rho kinase signaling may contribute to these alterations. OBJECTIVES: To investigate possible links between the 5-HT transporter (5-HTT) and RhoA/Rho kinase pathways, as well as their involvement in the progression of human and experimental PH. METHODS: Biochemical and functional analyses of lungs, platelets, and pulmonary artery smooth muscle cells (PA-SMCs) from patients with idiopathic PH (iPH) and 5-HTT overexpressing mice. MEASUREMENTS AND MAIN RESULTS: Lungs, platelets, and PA-SMCs from patients with iPH were characterized by marked elevation in RhoA and Rho kinase activities and a strong increase in 5-HT binding to RhoA indicating RhoA serotonylation. The 5-HTT inhibitor fluoxetine and the type 2 transglutaminase inhibitor monodansylcadaverin prevented 5-HT-induced RhoA serotonylation and RhoA/Rho kinase activation, as well as 5-HT-induced proliferation of PA-SMCs from iPH patients that was also inhibited by the Rho kinase inhibitor fasudil. Increased Rho kinase activity, RhoA activation, and RhoA serotonylation were also observed in lungs from SM22-5-HTT(+)mice, which overexpress 5-HTT in smooth muscle and spontaneously develop PH. Treatment of SM22-5-HTT(+) mice with either fasudil or fluoxetine limited PH progression and RhoA/Rho kinase activation. CONCLUSIONS: RhoA and Rho kinase activities are increased in iPH, in association with enhanced RhoA serotonylation. Direct involvement of the 5-HTT/RhoA/Rho kinase signaling pathway in 5-HT-mediated PA-SMC proliferation and platelet activation during PH progression identify RhoA/Rho kinase signaling as a promising target for new treatments against PH.


Asunto(s)
Hipertensión Pulmonar/enzimología , Serotonina/metabolismo , Transducción de Señal/fisiología , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Adolescente , Adulto , Animales , Plaquetas/metabolismo , Western Blotting , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Activación Enzimática , Femenino , Humanos , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/fisiopatología , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Vasoconstricción/fisiología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...