Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(10)2022 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-35628350

RESUMO

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.


Assuntos
Membro Posterior , Isquemia , Óxido Nítrico Sintase Tipo III , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Citrato de Sildenafila , Animais , Membro Posterior/irrigação sanguínea , Membro Posterior/efeitos dos fármacos , Membro Posterior/metabolismo , Isquemia/tratamento farmacológico , Óxido Nítrico Sintase Tipo III/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Transdução de Sinais , Citrato de Sildenafila/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
Physiol Rev ; 93(4): 1659-720, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24137019

RESUMO

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.


Assuntos
Doenças Cardiovasculares/fisiopatologia , Fenômenos Fisiológicos Cardiovasculares , Proteínas Monoméricas de Ligação ao GTP/fisiologia , Animais , Humanos , Modelos Animais , Proteínas Monoméricas de Ligação ao GTP/química , Transdução de Sinais/fisiologia , Proteínas ras/fisiologia
3.
Am J Physiol Cell Physiol ; 302(9): C1394-404, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22322975

RESUMO

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.


Assuntos
Retroalimentação Fisiológica/fisiologia , Fatores de Troca do Nucleotídeo Guanina/biossíntese , Hipertensão/fisiopatologia , Músculo Liso Vascular/metabolismo , Quinases Associadas a rho/metabolismo , Angiotensina II/metabolismo , Angiotensina II/toxicidade , Animais , Artérias/metabolismo , Western Blotting , Perfilação da Expressão Gênica , Hipertensão/induzido quimicamente , Masculino , Músculo Liso Vascular/fisiopatologia , RNA Interferente Pequeno , Ratos , Ratos Endogâmicos WKY , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Transfecção
4.
Arterioscler Thromb Vasc Biol ; 31(11): 2634-42, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21852563

RESUMO

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.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Estradiol/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Feminino , Masculino , Camundongos , Camundongos Knockout , Modelos Animais , Músculo Liso Vascular/citologia , Ovariectomia , Fosforilação/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Vasoconstrição/fisiologia , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP
5.
Circulation ; 122(19): 1937-47, 2010 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-20974998

RESUMO

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.


Assuntos
Inibidores da Protease de HIV/farmacologia , Hipertensão Pulmonar/fisiopatologia , Animais , Animais Recém-Nascidos , Terapia Antirretroviral de Alta Atividade , Antivirais/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Carbamatos/farmacologia , Divisão Celular/efeitos dos fármacos , Furanos , Hemodinâmica/efeitos dos fármacos , Hemodinâmica/fisiologia , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/mortalidade , Hipóxia , Masculino , Monocrotalina , Nelfinavir/farmacologia , Artéria Pulmonar/citologia , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/fisiopatologia , Ratos , Ratos Wistar , Ritonavir/farmacologia , Sulfonamidas/farmacologia
6.
Circ Res ; 102(10): 1265-74, 2008 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-18420945

RESUMO

The small G protein Rho signaling pathways are recognized as major regulators of cardiovascular functions, and activation of Rho proteins appears to be a common component for the pathogenesis of hypertension and vascular proliferative disorders. Recent evidence suggests that modulation of Rho protein signaling by phosphorylation of Rho proteins provides an additional simple mechanism for coordinating Rho protein functions. Phosphorylation of RhoA by cAMP- or cGMP-activated kinase on Ser188 induces cytosolic sequestration of RhoA through increased interaction with guanine dissociation inhibitor, thereby resulting in inhibition of RhoA-dependent functions. Here we show that stimulation of angiotensin II (Ang II) type 2 receptor (AT(2)R) in vascular smooth muscle cells induces Ser188 phosphorylation of RhoA independently of cAMP- or cGMP-activated kinase. We identify the Ser/Thr kinase Ste20-related kinase SLK as a new kinase phosphorylating RhoA on Ser188. Activation of the signaling cascade involving Src homology 2 domain-containing protein-tyrosine phosphatase 1, casein kinase II and SLK is responsible for RhoA phosphorylation and inhibition of RhoA-mediated arterial contraction induced by AT(2)R activation. These results thus identify the molecular mechanism linking AT(2)R to RhoA inhibition and vasodilation.


Assuntos
Hipertensão/metabolismo , Músculo Liso Vascular/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Receptor Tipo 2 de Angiotensina/metabolismo , Vasodilatação/fisiologia , Proteína rhoA de Ligação ao GTP/metabolismo , Angiotensina II/farmacologia , Animais , Aorta Torácica/citologia , Aorta Torácica/enzimologia , Caseína Quinase II/metabolismo , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Etinilestradiol/metabolismo , Masculino , Acetato de Megestrol/metabolismo , Músculo Liso Vascular/citologia , Óxido Nítrico Sintase/metabolismo , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Regulação para Cima/fisiologia , Vasoconstritores/farmacologia
7.
Am J Respir Crit Care Med ; 179(12): 1151-8, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19299501

RESUMO

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.


Assuntos
Hipertensão Pulmonar/enzimologia , Serotonina/metabolismo , Transdução de Sinais/fisiologia , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Adolescente , Adulto , Animais , Plaquetas/metabolismo , Western Blotting , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Ativação Enzimática , Feminino , Humanos , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/fisiopatologia , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Artéria Pulmonar/metabolismo , Artéria Pulmonar/patologia , Vasoconstrição/fisiologia , Adulto Jovem
8.
Am J Physiol Cell Physiol ; 297(5): C1062-70, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19692654

RESUMO

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.


Assuntos
Angiotensina II/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Músculo Liso Vascular/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Western Blotting , Ativação Enzimática/fisiologia , Imunoprecipitação , Miócitos de Músculo Liso/metabolismo , Fosforilação , RNA Interferente Pequeno , Ratos , Receptor Tipo 1 de Angiotensina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Quinases Associadas a rho/metabolismo
9.
Curr Opin Pharmacol ; 8(2): 174-80, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18222728

RESUMO

Increasing evidence has accumulated to implicate overactivation of Rho protein as a common component for the pathogenesis of several cardiovascular disorders including hypertension, coronary and cerebral vasospasm, atherosclerosis, and diabetes. Recent advances in Rho protein signaling research indicate that the Rho exchange factors (Rho GEFs) which activate Rho proteins by catalyzing the exchange of GDP for GTP are major regulators of Rho protein activity. In addition, linkage analysis and association studies have recently identified Rho GEFs as susceptibility genes for cardiovascular diseases. All of these data are converging to suggest that as upstream activators of Rho proteins, Rho GEFs expressed in cardiovascular cells are good candidate targets for the treatment of cardiovascular disorders.


Assuntos
Doenças Cardiovasculares/fisiopatologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Doenças Cardiovasculares/tratamento farmacológico , Fenômenos Fisiológicos Cardiovasculares , Sistemas de Liberação de Medicamentos , Expressão Gênica , Humanos , Polimorfismo Genético , Transdução de Sinais
10.
Oxid Med Cell Longev ; 2019: 2567198, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31214278

RESUMO

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.


Assuntos
Envelhecimento/fisiologia , Aorta/patologia , Doenças Cardiovasculares/terapia , Endotélio/fisiologia , Moringa oleifera , Animais , Células Cultivadas , Suplementos Nutricionais , Humanos , Masculino , Contração Miocárdica , Óxido Nítrico Sintase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Sementes , Transdução de Sinais , Vasodilatação
11.
Peptides ; 29(5): 778-82, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-17933432

RESUMO

Urotensin II, through its interaction with its UT receptor, is a potent vasoactive peptide in humans and in several animal models. Recent studies have demonstrated elevated plasma U-II levels in patients with atherosclerosis and coronary artery disease. U-II is expressed in endothelial cells, smooth muscle cells and infiltrating macrophages of atherosclerotic human coronary arteries. UT receptor expression is up-regulated by inflammatory stimuli. Activation of UT receptor by U-II stimulates endothelial and smooth muscle cell proliferation and monocytes chemotaxis. Therefore, in addition to its primary vasoactive effect, these observations suggest a role of U-II and UT receptor in the initiation and/or progression of atherosclerosis.


Assuntos
Aterosclerose/metabolismo , Urotensinas/metabolismo , Animais , Proliferação de Células , Fatores Quimiotáticos/metabolismo , Células Endoteliais/metabolismo , Células Espumosas/metabolismo , Humanos , Inflamação/metabolismo , Monócitos/citologia , Monócitos/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Urotensinas/genética
12.
Peptides ; 29(5): 658-73, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-17931747

RESUMO

Urotensin II (U-II) and urotensin II-related peptide (URP) are the endogenous ligands for the orphan G-protein-coupled receptor GPR14 now renamed UT. At the periphery, U-II and/or URP exert a wide range of biological effects on cardiovascular tissues, airway smooth muscles, kidney and endocrine glands, while central administration of U-II elicits various behavioral and cardiovascular responses. There is also evidence that U-II and/or URP may be involved in a number of pathological conditions including heart failure, atherosclerosis, renal dysfunction and diabetes. Because of the potential involvement of the urotensinergic system in various physiopathological processes, there is need for the rational design of potent and selective ligands for the UT receptor. Structure-activity relationship studies have shown that the minimal sequence required to retain full biological activity is the conserved U-II(4-11) domain, in particular the Cys5 and Cys10 residues involved in the disulfide bridge, and the Phe6, Lys8 and Tyr9 residues. Free alpha-amino group and C-terminal COOH group are not necessary for the biological activity, and modifications of these radicals may even increase the stability of the analogs. Punctual substitution of native amino acids, notably Phe6 and Trp7, by particular residues generates analogs with antagonistic properties. These studies, which provide crucial information regarding the structural and conformational requirements for ligand-receptor interactions, will be of considerable importance for the design of novel UT ligands with increased selectivity, potency and stability, that may eventually lead to the development of innovative drugs.


Assuntos
Hormônios Peptídicos/química , Hormônios Peptídicos/metabolismo , Relação Estrutura-Atividade , Urotensinas/química , Urotensinas/metabolismo , Alanina/química , Alanina/metabolismo , Sequência de Aminoácidos , Animais , Cisteína/química , Cisteína/metabolismo , Dissulfetos/química , Humanos , Dados de Sequência Molecular , Estrutura Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Hormônios Peptídicos/genética , Conformação Proteica , Alinhamento de Sequência , Tirosina/química , Tirosina/metabolismo , Urotensinas/genética
13.
Circ Res ; 98(3): 322-34, 2006 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-16484628

RESUMO

Rho kinases (ROCKs) are the first and the best-characterized effectors of the small G-protein RhoA. In addition to their effect on actin organization, or through this effect, ROCKs have been found to regulate a wide range of fundamental cell functions such as contraction, motility, proliferation, and apoptosis. Abnormal activation of the RhoA/ROCK pathway has been observed in major cardiovascular disorders such as atherosclerosis, restenosis, hypertension, pulmonary hypertension, and cardiac hypertrophy. This review, based on recent molecular, cellular, and animal studies, focuses on the current understanding of ROCK signaling and its roles in cardiovascular physiology and pathophysiology.


Assuntos
Doenças Cardiovasculares/fisiopatologia , Fenômenos Fisiológicos Cardiovasculares , Sistema Cardiovascular/fisiopatologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Aterosclerose/enzimologia , Aterosclerose/fisiopatologia , Doenças Cardiovasculares/enzimologia , Reestenose Coronária , Endotélio Vascular/fisiologia , Humanos , Hipertensão/enzimologia , Hipertensão/fisiopatologia , Inflamação/enzimologia , Inflamação/fisiopatologia , Músculo Liso Vascular/fisiologia , Quinases Associadas a rho
14.
Cardiovasc Res ; 73(1): 190-7, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17150200

RESUMO

OBJECTIVE: Myogenic tone, which has a major role in the regulation of local blood flow, refers to the ability of vascular smooth muscle to adapt its contractility to changes in transmural pressure. Although Rho-kinase is involved in myogenic tone, the pathway involved remains unclear, especially concerning translocation to the plasma membrane and activation of RhoA. As caveolae have a key role in the signal transduction of membrane-bound proteins, we tested the hypothesis that RhoA might be activated by pressure and that its activation might involve caveolin-1, which has been shown to be involved in vascular functions. METHODS: Myogenic tone was studied in isolated rat mesenteric resistance arteries (118+/-15 microm internal diameter with a pressure of 75 mmHg) submitted to pressure steps (25, 75, and 150 mmHg). Pharmacological blockade of caveolae or RhoA-Rho-kinase pathway was assessed by confocal microscopy in pressurized arteries to analyze protein co-localization and by co-immunoprecipitation in order to confirm protein interactions. Caveolin-1-deficient mice were used to confirm the role of the protein in myogenic tone. RESULTS: Pressure-induced myogenic tone was significantly reduced by RhoA inactivation with TAT-C3 (90.5% inhibition at 150 mmHg) and by the Rho-kinase inhibitor Y27632 (91.8% inhibition at 150 mmHg). In arteries pressurized at 150 mmHg, RhoA was localized to the plasma membrane (localization by confocal microscopy and increased quantity of RhoA in the membrane fraction after protein extraction). Thus, translocation of RhoA to the plasma membrane was associated with pressure-induced tone. In addition, caveolae disruption with methyl-beta-cyclodextrin reduced myogenic tone by 66% at 150 mmHg. Further, myogenic tone was significantly reduced to 24% of control in caveolin-1-deficient mice (active tone was 32.3+/-2.8 microm and 9.1+/-3.7 microm in +/+ and -/- mice, respectively, n = 5 per group), suggesting a key role of caveolin-1 in myogenic tone. Finally, RhoA and caveolin-1 co-immunoprecipitation and co-localization significantly increased when myogenic tone developed at 150 mmHg (co-localization showed 26+/-13% merging at 25 mmHg versus 97+/-21% at 150 mmHg, n = 5). Co-immunoprecipitation was prevented by TAT-C3 and by methyl beta-cyclodextrin. CONCLUSION: RhoA activation is critical for the development of myogenic tone in resistance arteries. This activation induced translocation of RhoA to the plasma membrane within caveolae, where the interaction of RhoA with caveolin-1 leads selectively to the activation of a Rho-kinase-dependent force development.


Assuntos
Caveolina 1/metabolismo , Artérias Mesentéricas/metabolismo , Músculo Liso Vascular/metabolismo , Vasoconstrição/efeitos dos fármacos , Proteína rhoA de Ligação ao GTP/metabolismo , Actinas/análise , Amidas/farmacologia , Animais , Transporte Biológico Ativo , Western Blotting/métodos , Caveolina 1/análise , Membrana Celular/metabolismo , Colesterol/farmacologia , Citosol/metabolismo , Inibidores Enzimáticos/farmacologia , Imuno-Histoquímica/métodos , Imunoprecipitação/métodos , Técnicas In Vitro , Camundongos , Camundongos Knockout , Microscopia Confocal , Piridinas/farmacologia , Ratos , Ratos Wistar , Resistência Vascular , beta-Ciclodextrinas/farmacologia , Proteína rhoA de Ligação ao GTP/antagonistas & inibidores , Proteína rhoA de Ligação ao GTP/genética
15.
J Vasc Res ; 44(6): 444-59, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17643059

RESUMO

This study was designed to identify the global pattern of differentially expressed genes in human varicose veins. Using suppressive subtractive hybridization, we identified overexpression of genes known to be associated with extracellular matrix remodeling, including collagen III, tissue inhibitor of metalloproteinases I, dermatopontin, matrix Gla protein (MGP) and tenascin C. Real-time polymerase chain reaction analysis confirmed the differential expression of these genes. The overexpression of MGP transcript was associated with increased MGP level in varicose veins, in particular the undercarboxylated form of the protein. Smooth muscle cells from varicose veins showed increased proliferation rate and enhanced matrix mineralization. This observation correlated with the presence of ectopic mineralization areas in the varicose vein walls. The use of warfarin, to inhibit MGP activity, or siRNA targeting MGP transcript induced a reduction in the exacerbated proliferation of varicose vein smooth muscle cells. Our results suggest that high expression of MGP in varicose veins may contribute to venous wall remodeling by affecting proliferation and mineralization processes probably through impaired carboxylation of MGP. In addition, suppressive subtractive hybridization results also produce a profile of differentially expressed genes in varicose veins, in particular extracellular matrix components. Further study of these genes will provide insights into their specific roles in the etiology of venous disease.


Assuntos
Calcinose/genética , Proteínas de Ligação ao Cálcio/genética , Proteínas da Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Regulação da Expressão Gênica , Músculo Liso Vascular/metabolismo , Processamento de Proteína Pós-Traducional , Varizes/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Calcinose/metabolismo , Calcinose/patologia , Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Proteínas de Ligação ao Cálcio/metabolismo , Estudos de Casos e Controles , Proliferação de Células , Células Cultivadas , Relação Dose-Resposta a Droga , Proteínas da Matriz Extracelular/antagonistas & inibidores , Proteínas da Matriz Extracelular/metabolismo , Perfilação da Expressão Gênica/métodos , Glicerofosfatos/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Veia Safena/metabolismo , Regulação para Cima , Varizes/metabolismo , Varizes/patologia , Varfarina/farmacologia , Proteína de Matriz Gla
16.
Circ Res ; 96(11): 1152-60, 2005 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-15890975

RESUMO

cAMP and cyclic GMP-dependent kinases (PKA and PKG) phosphorylate the small G protein RhoA on Ser188. We have previously demonstrated that phosphorylation of Ser188 inhibits RhoA-dependent functions and positively regulates RhoA expression, and that the nitric oxide (NO)/cGMP-dependent protein kinase pathway plays an essential role, both in vitro and in vivo, in the regulation of RhoA protein expression and functions in vascular smooth muscle cells. Here we analyze the consequences of Ser188 phosphorylation on RhoA protein degradation. By expressing Ser188 phosphomimetic wild-type (WT-RhoA-S188E) and active RhoA proteins (Q63L-RhoA-S188E), we show that phosphorylation of Ser188 of RhoA protects RhoA, particularly its active form, from ubiquitin-mediated proteasomal degradation. Coimmunoprecipitation experiments indicate that the resistance of the phosphorylated active form of RhoA to proteasome-mediated degradation is because of its cytoplasmic sequestration through enhanced RhoGDI interaction. In rat aortic smooth muscle cells, stimulation of PKG and inhibition of proteasome by lactacystin, induce nonadditive increases in RhoA protein expression. In addition, stimulation of PKG leads to the accumulation of GTP-bound RhoA in the cytoplasm. In vivo stimulation of the NO/PKG signaling by treating rats with sildenafil increased RhoA level and RhoA phosphorylation, and enhanced its association to RhoGDI in the pulmonary artery, whereas opposite effects are induced by chronic inhibition of NO synthesis in N-omega-nitro-L-arginine-treated rats. Our results thus suggest that Ser188 phosphorylation-mediated protection against degradation is a physiological process regulating the level of endogenous RhoA and define a novel function for RhoGDI, as an inhibitor of Rho protein degradation.


Assuntos
Músculo Liso Vascular/metabolismo , Complexo de Endopeptidases do Proteassoma/fisiologia , Serina/metabolismo , Ubiquitina/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Células Cultivadas , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Citosol/metabolismo , Inibidores de Dissociação do Nucleotídeo Guanina/metabolismo , Humanos , Masculino , Camundongos , Óxido Nítrico/fisiologia , Fosforilação , Ratos , Ratos Wistar , Transdução de Sinais , Células Swiss 3T3 , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho , Inibidores da Dissociação do Nucleotídeo Guanina rho-Específico
17.
Trends Cardiovasc Med ; 16(6): 199-204, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16839863

RESUMO

The small G protein Rho signaling pathways are recognized as major regulators of cardiovascular functions, and activation of Rho proteins appears to be a common component for the pathogenesis of hypertension and vascular proliferative disorders. Rho proteins are tightly regulated, and recent evidence suggests that modulation of Rho protein signaling by phosphorylation of Rho proteins provides an additional simple mechanism for coordinating Rho protein functions. This regulation by phosphorylation is particularly important in the arterial wall, where RhoA protein expressed in vascular smooth muscle cells is controlled by the endothelium through the nitric oxide/cGMP-dependent kinase pathway.


Assuntos
Sistema Cardiovascular/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/fisiopatologia , Sistema Cardiovascular/patologia , Sistema Cardiovascular/fisiopatologia , Humanos , Fosforilação , Transdução de Sinais , Proteínas rho de Ligação ao GTP/classificação , Proteínas rho de Ligação ao GTP/fisiologia
18.
Cardiovasc Res ; 72(2): 339-48, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16934786

RESUMO

OBJECTIVE: Hyaluronan (HA) is an important constituent of the extracellular matrix and is known to regulate cellular events through binding to CD44 and the receptor for HA-mediated motility (RHAMM). Here we investigated the role of these receptors and the signaling pathways involved in HA-mediated effects in arterial smooth muscle cells (ASMC). METHODS: Effects of high-molecular weight HA (1 to 5 mg/ml) were analyzed in cultured ASMC from rat aorta. RESULTS: HA promoted actin stress fiber and lamellipodia formation and dose-dependently induced ASMC migration without effect on proliferation. Pull-down assay of Rho protein activity indicated that HA activated RhoA and Rac. HA-induced ASMC migration was not affected by the RhoA inhibitor Tat-C3 (10 microg/ml), the Rho kinase inhibitor Y-27632 (10 microM) and blocking anti-CD44 antibody ,but was reduced by the non-selective Rho protein inhibitor simvastatin (10 microM), the Rac inhibitor LT-toxin (1 mug/ml), small interfering RNA (siRNA) targeting Rac and the phosphatidyl inositol 3-kinase (PI3K) inhibitor LY294002 (25 microM), which also blocked HA-induced Rac activation. CD44 knockdown by siRNA inhibited HA-mediated RhoA activation without effect on ASMC migration. In contrast, siRNA targeting RHAMM inhibited both HA-induced migration and Rac activation. CONCLUSIONS: High-molecular weight HA independently activates RhoA and Rac through CD44 and RHAMM, respectively. HA-induced migration depends exclusively on RHAMM-mediated PI3K-dependent Rac activation.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/farmacologia , Músculo Liso Vascular/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Actinas/metabolismo , Amidas/farmacologia , Animais , Anticorpos Monoclonais/farmacologia , Aorta , Toxinas Bacterianas/farmacologia , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Cromonas/farmacologia , Complemento C3/genética , Citoesqueleto/metabolismo , Relação Dose-Resposta a Droga , Enterotoxinas/farmacologia , Proteínas de Escherichia coli/farmacologia , Genes tat , Receptores de Hialuronatos/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Morfolinas/farmacologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Piridinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Ratos , Proteínas Recombinantes de Fusão/farmacologia , Transdução de Sinais , Sinvastatina/farmacologia , Estimulação Química , Proteínas rac de Ligação ao GTP/antagonistas & inibidores , Proteínas rac de Ligação ao GTP/genética , Proteínas rac de Ligação ao GTP/metabolismo , Quinases Associadas a rho , Proteína rhoA de Ligação ao GTP/antagonistas & inibidores , Proteína rhoA de Ligação ao GTP/metabolismo
19.
Oxid Med Cell Longev ; 2017: 4129459, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28713487

RESUMO

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.


Assuntos
Hipertensão/tratamento farmacológico , Moringa oleifera/metabolismo , Animais , Antioxidantes/metabolismo , Masculino , Moringa oleifera/citologia , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Endogâmicos WKY
20.
J Med Chem ; 49(24): 7234-8, 2006 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-17125276

RESUMO

Urotensin II (U-II) is a potent vasoconstrictor peptide which has been identified as the endogenous ligand for the orphan G protein-coupled receptor GPR14 now renamed UT receptor. As the C-terminal cyclic hexapeptide of U-II (U-II(4-11), H-Asp-Cys-Phe-Trp-Lys-Tyr-Cys-Val-OH) possesses full biological activity, we have synthesized a series of U-II(4-11) analogues and measured their binding affinity on hGPR14-transfected CHO cells and their contractile activity on de-endothelialized rat aortic rings. The data indicate that a free amino group and a functionalized side-chain at the N-terminal extremity of the peptide are not required for biological activity. In addition, the minimal chemical requirement at position 9 of U-II(4-11) is the presence of an aromatic moiety. Most importantly, replacement of the Phe6 residue by cyclohexyl-Ala (Cha) led to an analogue, [Cha6]U-II(4-11), that was devoid of agonistic activity but was able to dose-dependently suppress the vasoconstrictor effect of U-II on rat aortic rings. These new pharmacological data, by providing further information regarding the structure-activity relationships of U-II analogues, should prove useful for the rational design of potent and nonpeptidic UT receptor agonists and antagonists.


Assuntos
Fragmentos de Peptídeos/síntese química , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Urotensinas/antagonistas & inibidores , Urotensinas/síntese química , Vasodilatadores/síntese química , Animais , Células CHO , Cricetinae , Cricetulus , Humanos , Técnicas In Vitro , Fragmentos de Peptídeos/farmacologia , Ensaio Radioligante , Ratos , Receptores Acoplados a Proteínas G/genética , Relação Estrutura-Atividade , Urotensinas/farmacologia , Vasodilatadores/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA