Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
Mais filtros

País/Região como assunto
Intervalo de ano de publicação
1.
J Mol Cell Cardiol ; 66: 18-26, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24445059

RESUMO

Fat1 is an atypical cadherin that controls vascular smooth muscle cell (VSMC) proliferation and migration. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 1 (Nox1) is an important source of reactive oxygen species (ROS) in VSMCs. Angiotensin II (Ang II) induces the expression and/or activation of both Fat1 and Nox1 proteins. This study tested the hypothesis that Ang II-induced Fat1 activation and VSMC migration are mediated by Nox1-dependent ROS generation and redox signaling. Studies were performed in cultured VSMCs from Sprague­Dawley rats. Cells were treated with Ang II (1 µmol/L) for short (5 to 30 min) or long term stimulations (3 to 12 h) in the absence or presence of the antioxidant apocynin (10 µmol/L), extracellular-signal-regulated kinases 1/2 (Erk1/2) inhibitor PD98059 (1 µmol/L), or Ang II type 1 receptor (AT1R) valsartan (1 µmol/L). siRNA was used to knockdown Nox1 or Fat1. Cell migration was determined by Boyden chamber assay. Ang II increased Fat1 mRNA and protein levels and promoted Fat1 translocation to the cell membrane, responses that were inhibited by AT1R antagonist and antioxidant treatment. Downregulation of Nox1 inhibited the effects of Ang II on Fat1 protein expression. Nox1 protein induction, ROS generation, and p44/p42 MAPK phosphorylation in response to Ang II were prevented by valsartan and apocynin, and Nox1 siRNA inhibited Ang II-induced ROS generation. Knockdown of Fat1 did not affect Ang II-mediated increases in Nox1 expression or ROS. Inhibition of p44/p42 MAPK phosphorylation by PD98059 abrogated the Ang II-induced increase in Fat1 expression and membrane translocation. Knockdown of Fat1 inhibited Ang II-induced VSMC migration, which was also prevented by valsartan, apocynin, PD98059, and Nox1 siRNA. Our findings indicate that Ang II regulates Fat1 expression and activity and induces Fat1-dependent VSMC migration via activation of AT1R, ERK1/2, and Nox1-derived ROS, suggesting a role for Fat1 downstream of Ang II signaling that leads to vascular remodeling.


Assuntos
Angiotensina II/farmacologia , Caderinas/genética , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , NADH NADPH Oxirredutases/genética , Acetofenonas/farmacologia , Angiotensina II/metabolismo , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Antioxidantes/farmacologia , Caderinas/agonistas , Caderinas/antagonistas & inibidores , Caderinas/metabolismo , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Flavonoides/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , NADH NADPH Oxirredutases/metabolismo , NADPH Oxidase 1 , Oxirredução , Inibidores de Proteínas Quinases/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Tetrazóis/farmacologia , Valina/análogos & derivados , Valina/farmacologia , Valsartana
2.
Diabetologia ; 57(3): 633-42, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24292634

RESUMO

AIMS/HYPOTHESIS: Enhanced vascular inflammation, immune cell infiltration and elevated production of reactive oxygen species (ROS) contribute significantly to pro-atherogenic responses in diabetes. We assessed the immunomodulatory role of NADPH oxidase (NOX)-derived ROS in diabetes-accelerated atherosclerosis. METHODS: Diabetes was induced in male Apoe(-/-) mice with five daily doses of streptozotocin (55 mg kg(-1) day(-1)). Atherosclerotic plaque size, markers of ROS and immune cell accumulation were assessed in addition to flow cytometric analyses of cells isolated from the adjacent mediastinal lymph nodes (meLNs). The role of NOX-derived ROS was investigated using the NOX inhibitor, GKT137831 (60 mg/kg per day; gavage) administered to diabetic and non-diabetic Apoe(-/-) mice for 10 weeks. RESULTS: Diabetes increased atherosclerotic plaque development in the aortic sinus and this correlated with increased lesional accumulation of T cells and CD11c(+) cells and altered T cell activation in the adjacent meLNs. Diabetic Apoe(-/-) mice demonstrated an elevation in vascular ROS production and expression of the proinflammatory markers monocyte chemoattractant protein 1, vascular adhesion molecule 1 and IFNγ. Blockade of NOX-derived ROS using GKT137831 prevented the diabetes-mediated increase in atherosclerotic plaque area and associated vascular T cell infiltration and also significantly reduced vascular ROS as well as markers of inflammation and plaque necrotic core area. CONCLUSIONS/INTERPRETATION: Diabetes promotes pro-inflammatory immune responses in the aortic sinus and its associated lymphoid tissue. These changes are associated with increased ROS production by NOX. Blockade of NOX-derived ROS using the NOX inhibitor GKT137831 is associated with attenuation of these changes in the immune response and reduces the diabetes-accelerated development of atherosclerotic plaques in Apoe(-/-) mice.


Assuntos
Aorta Torácica/patologia , Diabetes Mellitus Experimental/tratamento farmacológico , Angiopatias Diabéticas/tratamento farmacológico , Inflamação/tratamento farmacológico , NADPH Oxidases/efeitos dos fármacos , Placa Aterosclerótica/tratamento farmacológico , Pirazóis/farmacologia , Piridinas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Animais , Aorta Torácica/efeitos dos fármacos , Apolipoproteínas E/deficiência , Aterosclerose , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/patologia , Angiopatias Diabéticas/imunologia , Angiopatias Diabéticas/patologia , Imuno-Histoquímica , Inflamação/imunologia , Inflamação/patologia , Masculino , Camundongos , NADPH Oxidases/biossíntese , Oxirredução , Placa Aterosclerótica/imunologia , Placa Aterosclerótica/patologia , Pirazolonas , Piridonas
3.
Physiol Res ; 72(Suppl 2): S73-S90, 2023 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-37565414

RESUMO

The worldwide increase in the incidence of obesity and cardiovascular and neurodegenerative diseases, e.g. Alzheimer's disease, is related to many factors, including an unhealthy lifestyle and aging populations. However, the interconnection between these diseases is not entirely clear, and it is unknown whether common mechanisms underlie these conditions. Moreover, there are currently no fully effective therapies for obesity and neurodegeneration. While there has been extensive research in preclinical models addressing these issues, the experimental findings have not been translated to the clinic. Another challenge relates to the time of onset of individual diseases, which may not be easily identified, since there are no specific indicators or biomarkers that define disease onset. Hence knowing when to commence preventive treatment is unclear. This is especially pertinent in neurodegenerative diseases, where the onset of the disease may be subtle and occur decades before the signs and symptoms manifest. In metabolic and cardiovascular disorders, the risk may occur in-utero, in line with the concept of fetal programming. This review provides a brief overview of the link between obesity, cardiovascular and neurodegenerative diseases and discusses potential common mechanisms including the role of the gut microbiome.


Assuntos
Doença de Alzheimer , Doenças Neurodegenerativas , Humanos , Doenças Neurodegenerativas/metabolismo , Doença de Alzheimer/metabolismo , Obesidade/complicações , Obesidade/diagnóstico , Obesidade/epidemiologia
4.
Am J Physiol Renal Physiol ; 299(6): F1348-58, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20630933

RESUMO

Molecular mechanisms underlying renal complications of diabetes remain unclear. We tested whether renal NADPH oxidase (Nox) 4 contributes to increased reactive oxygen species (ROS) generation and hyperactivation of redox-sensitive signaling pathways in diabetic nephropathy. Diabetic mice (db/db) (20 wk) and cultured mouse proximal tubule (MPT) cells exposed to high glucose (25 mmol/l, D-glucose) were studied. Expression (gene and protein) of Nox4, p22(phox), and p47(phox), but not Nox1 or Nox2, was increased in kidney cortex, but not medulla, from db/db vs. control mice (db/m) (P < 0.05). ROS generation, p38 mitogen-activated protein (MAP) kinase phosphorylation, and content of fibronectin and transforming growth factor (TGF)-ß1/2 were increased in db/db vs. db/m (P < 0.01). High glucose increased expression of Nox4, but not other Noxes vs. normal glucose (P < 0.05). This was associated with increased NADPH oxidase activation and enhanced ROS production. Nox4 downregulation by small-interfering RNA and inhibition of Nox4 activity by GK-136901 (Nox1/4 inhibitor) attenuated d-glucose-induced NADPH oxidase-derived ROS generation. High d-glucose, but not l-glucose, stimulated phosphorylation of p38MAP kinase and increased expression of TGF-ß1/2 and fibronectin, effects that were inhibited by SB-203580 (p38MAP kinase inhibitor). GK-136901 inhibited d-glucose-induced actions. Our data indicate that, in diabetic conditions: 1) renal Nox4 is upregulated in a cortex-specific manner, 2) MPT cells possess functionally active Nox4-based NADPH, 3) Nox4 is a major source of renal ROS, and 4) activation of profibrotic processes is mediated via Nox4-sensitive, p38MAP kinase-dependent pathways. These findings implicate Nox4-based NADPH oxidase in molecular mechanisms underlying fibrosis in type 2 diabetic nephropathy.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Nefropatias Diabéticas/metabolismo , Rim/metabolismo , NADPH Oxidases/fisiologia , Animais , Células Cultivadas , Grupo dos Citocromos b/biossíntese , Diabetes Mellitus Experimental/patologia , Nefropatias Diabéticas/patologia , Fibrose , Glucose/farmacologia , Masculino , Camundongos , NADPH Oxidase 4 , NADPH Oxidases/biossíntese , Estresse Oxidativo/efeitos dos fármacos , Pirazóis/farmacologia , Piridonas/farmacologia , RNA Interferente Pequeno/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/efeitos dos fármacos
5.
Arterioscler Thromb Vasc Biol ; 28(8): 1511-8, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18467645

RESUMO

OBJECTIVE: Synergistic interactions between aldosterone (Aldo) and angiotensin II (Ang II) have been implicated in vascular inflammation, fibrosis, and remodeling. Molecular mechanisms underlying this are unclear. We tested the hypothesis that c-Src activation, through receptor tyrosine kinase transactivation, is critically involved in synergistic interactions between Aldo and Ang II and that it is upstream of promigratory signaling pathways in vascular smooth muscle cells (VSMCs). METHODS AND RESULTS: VSMCs from WKY rats were studied. At low concentrations (10(-10) mol/L) Aldo and Ang II alone did not influence c-Src activation, whereas in combination they rapidly increased phosphorylation (P<0.01), an effect blocked by eplerenone (Aldo receptor antagonist) and irbesartan (AT1R blocker). This synergism was attenuated by AG1478 and AG1296 (inhibitors of EGFR and PDGFR, respectively), but not by AG1024 (IGFR inhibitor). Aldo and Ang II costimulation induced c-Src-dependent activation of NAD(P)H oxidase and c-Src-independent activation of ERK1/2 (P<0.05), without effect on ERK5, p38MAPK, or JNK. Aldo/Ang II synergistically activated RhoA/Rho kinase and VSMC migration, effects blocked by PP2, apocynin, and fasudil, inhibitors of c-Src, NADPH oxidase, and Rho kinase, respectively. CONCLUSIONS: Aldo/Ang II synergistically activate c-Src, an immediate signaling response, through EGFR and PDGFR, but not IGFR transactivation. This is associated with activation of redox-regulated RhoA/Rho kinase, which controls VSMC migration. Although Aldo and Ang II interact to stimulate ERK1/2, such effects are c-Src-independent. These findings indicate differential signaling in Aldo-Ang II crosstalk and highlight the importance of c-Src in redox-sensitive RhoA, but not ERK1/2 signaling. Blockade of Aldo/Ang II may be therapeutically useful in vascular remodeling associated with abnormal VSMC migration.


Assuntos
Aldosterona/fisiologia , Angiotensina II/fisiologia , Movimento Celular/fisiologia , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/fisiologia , Animais , Células Cultivadas , Masculino , Ratos , Transdução de Sinais/fisiologia , Proteína rhoA de Ligação ao GTP/fisiologia , Quinases da Família src/fisiologia
7.
Arterioscler Thromb Vasc Biol ; 27(9): 1960-7, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17569879

RESUMO

OBJECTIVE: Endothelin-1 (ET-1) and angiotensin II (Ang II) activate common signaling pathways to promote changes in vascular reactivity, remodeling, inflammation, and oxidative stress. Here we sought to determine whether upstream regulators of mitogen-activated protein kinases (MAPKs) are differentially regulated by ET-1 and Ang II focusing on the role of c-Src and the small GTPase Ras. METHODS AND RESULTS: Mesenteric vascular smooth muscle cells (VSMCs) from mice with different disruption levels in the c-Src gene (c-Src(+/-) and c-Src(-/-)) and wild-type (c-Src(+/+)) were used. ET-1 and Ang II induced extracellular signal-regulated kinase (ERK) 1/2, SAPK/JNK, and p38MAPK phosphorylation in c-Src(+/+) VSMCs. In VSMCs from c-Src(+/-) and c-Src(-/-), Ang II effects were blunted, whereas c-Src deficiency had no effect in ET-1-induced MAPK activation. Ang II but not ET-1 induced c-Src phosphorylation in c-Src(+/+) VSMCs. Activation of c-Raf, an effector of Ras, was significantly increased by ET-1 and Ang II in c-Src(+/+) VSMCs. Ang II but not ET-1-mediated c-Raf phosphorylation was inhibited by c-Src deficiency. Knockdown of Ras by siRNA inhibited both ET-1 and Ang II-induced MAPK phosphorylation. CONCLUSIONS: Our data indicate differential regulation of MAPKs by distinct G protein-coupled receptors. Whereas Ang II has an obligatory need for c-Src, ET-1 mediates its actions through a c-Src-independent Ras-Raf-dependent pathway for MAPK activation. These findings suggest that Ang II and ET-1 can activate similar signaling pathways through unrelated mechanisms. MAP kinases are an important point of convergence for Ang II and ET-1.


Assuntos
Angiotensina II/fisiologia , Endotelina-1/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Monoméricas de Ligação ao GTP/fisiologia , Músculo Liso Vascular/enzimologia , Animais , Proteína Tirosina Quinase CSK , Células Cultivadas , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Proteínas Tirosina Quinases/fisiologia , Quinases da Família src
9.
Br J Pharmacol ; 151(1): 45-53, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17351653

RESUMO

BACKGROUND AND PURPOSE: Protective cardiovascular effects of peroxisome proliferator activated receptor (PPAR)alpha and PPARgamma activators have been demonstrated. If used as vasoprotective agents in high risk vascular patients rather than for their metabolic benefits, these agents could be associated with unwanted side effects. As a proof of concept to support the use of combined low doses of PPARalpha and PPARgamma as vascular protective agents in high risk vascular patients, we tested the hypothesis that combined low doses of PPARalpha (fenofibrate) and PPARgamma (rosiglitazone) activators would provide vascular protective benefits similar to full individual doses of these PPAR agonists. EXPERIMENTAL APPROACH: Male Sprague-Dawley rats infused with Ang II (120 ng kg(-1) min(-1)) were treated with rosiglitazone (1 or 2 mg kg(-1) day(-1)) alone or concomitantly with fenofibrate (30 mg kg(-1) day(-1)) for 7 days. Thereafter, vessels was assessed on a pressurized myograph, while NAD(P)H oxidase activity was determined by lucigenin chemiluminescence. Inflammation was evaluated using ELISA for NFkappaB and Western blotting for adhesion molecules. KEY RESULTS: Ang II-induced blood pressure increase, impaired acetylcholine-induced vasorelaxation, altered vascular structure, and enhanced vascular NAD(P)H oxidase activity and inflammation were significantly reduced by low dose rosiglitazone+fenofibrate. CONCLUSIONS AND IMPLICATIONS: Combined low doses of PPARalpha and PPARgamma activators attenuated development of hypertension, corrected vascular structural abnormalities, improved endothelial function, oxidative stress, and vascular inflammation. These agents used in low-dose combination have synergistic vascular protective effects. The clinical effects of combined low-dose PPARalpha and PPARgamma activators as vascular protective therapy, potentially with reduced side-effects and drug interactions, should be assessed.


Assuntos
Angiotensina II/farmacologia , Vasos Sanguíneos/efeitos dos fármacos , Hipertensão/tratamento farmacológico , PPAR alfa/efeitos dos fármacos , PPAR gama/efeitos dos fármacos , Animais , Pressão Sanguínea/efeitos dos fármacos , Vasos Sanguíneos/patologia , Sinergismo Farmacológico , Hipertensão/patologia , Masculino , NADPH Oxidases/sangue , PPAR alfa/fisiologia , PPAR gama/fisiologia , Ratos , Ratos Sprague-Dawley
10.
Methods Mol Biol ; 1527: 251-269, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28116722

RESUMO

This chapter outlines protocols to evaluate protein localization, recruitment or phosphorylation levels in cholesterol/sphingolipids-enriched cell membrane domains and recommends experimental designs with pharmacological tolls to evaluate potential cell functions associated with these domains. We emphasize the need for the combination of several approaches towards understanding the protein components and cellular functions attributed to these distinct microdomains.


Assuntos
Cavéolas/metabolismo , Microdomínios da Membrana/metabolismo , Animais , Carbonatos/metabolismo , Membrana Celular/metabolismo , Colesterol/química , Colesterol/metabolismo , Eletroforese em Gel de Poliacrilamida , Concentração de Íons de Hidrogênio , Microdomínios da Membrana/química , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Ratos , Esfingolipídeos/química , Esfingolipídeos/metabolismo , beta-Ciclodextrinas/química , beta-Ciclodextrinas/metabolismo
11.
Br J Pharmacol ; 174(22): 4055-4069, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27935022

RESUMO

BACKGROUND AND PURPOSE: Inflammation plays a key role in atherosclerosis. The protective role of angiotensin 1-7 (Ang-(1-7)) in vascular pathologies suggested the therapeutic use of low MW, non-peptide Ang-(1-7) mimetics, such as AVE0991. The mechanisms underlying the vaso-protective effects of AVE0991, a Mas receptor agonist, remain to be explored. EXPERIMENTAL APPROACH: We investigated the effects of AVE0991 on the spontaneous atherosclerosis in apolipoprotein E (ApoE)-/- mice, in the context of vascular inflammation and plaque stability. KEY RESULTS: AVE0991 has significant anti-atherosclerotic properties in ApoE-/- mice and increases plaque stability, by reducing plaque macrophage content, without effects on collagen. Using the descending aorta of chow-fed ApoE-/- mice, before significant atherosclerotic plaque develops, we gained insight to early events in atherosclerosis. Interestingly, perivascular adipose tissue (PVAT) and adventitial infiltration with macrophages and T-cells precedes atherosclerotic plaque or the impairment of endothelium-dependent NO bioavailability (a measure of endothelial function). AVE0991 inhibited perivascular inflammation, by reducing chemokine expression in PVAT and through direct actions on monocytes/macrophages inhibiting their activation, characterized by production of IL-1ß, TNF-α, CCL2 and CXCL10, and differentiation to M1 phenotype. Pretreatment with AVE0991 inhibited migration of THP-1 monocytes towards supernatants of activated adipocytes (SW872). Mas receptors were expressed in PVAT and in THP-1 cells in vitro, and the anti-inflammatory effects of AVE0991 were partly Mas dependent. CONCLUSIONS AND IMPLICATIONS: The selective Mas receptor agonist AVE0991 exhibited anti-atherosclerotic and anti-inflammatory actions, affecting monocyte/macrophage differentiation and recruitment to the perivascular space during early stages of atherosclerosis in ApoE-/- mice. LINKED ARTICLES: This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.


Assuntos
Anti-Inflamatórios/uso terapêutico , Aterosclerose/tratamento farmacológico , Imidazóis/uso terapêutico , Angiotensina I , Animais , Aorta/efeitos dos fármacos , Aorta/imunologia , Aorta/patologia , Aterosclerose/imunologia , Aterosclerose/patologia , Linhagem Celular , Linhagem Celular Tumoral , Citocinas/genética , Feminino , Humanos , Leucócitos/efeitos dos fármacos , Leucócitos/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Fragmentos de Peptídeos , Placa Aterosclerótica , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/agonistas , Receptores Acoplados a Proteínas G/agonistas
12.
Arterioscler Thromb Vasc Biol ; 25(3): 512-8, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15618548

RESUMO

OBJECTIVE: We tested the hypothesis that p47phox associates with the actin cytoskeleton, enabling site-directed activation of NAD(P)H oxidase, and assessed whether these actions influence reactive oxygen species (ROS) generation and signaling by angiotensin II (Ang II) in vascular smooth muscle cells (VSMCs) from human resistance and coronary arteries. METHODS AND RESULTS: Electroporation of anti-p47phox antibody into VSMCs abrogated Ang II-mediated O2 generation, establishing the requirement for p47phox in this response. Immunfluorescence confocal microscopy demonstrated a cytosolic distribution of p47phox in basal conditions. After Ang II stimulation, p47phox rearranged in a linear fashion, colocalizing with F-actin. Co-immunoprecipitation studies confirmed an association between p47phox and actin and demonstrated an interaction with the actin-binding protein cortactin. Cytoskeletal disruption with cytochalasin prevented p47phox:actin interaction and attenuated ROS formation and p38MAP kinase and Akt phosphorylation by Ang II. Intracellular ROS generation in response to LY83583 (O2 generator) or exogenous H2O2 and Ang II-induced ERK1/2 activation were unaltered by cytochalasin. CONCLUSIONS: The p47phox:actin interaction, through cortactin, plays an important role in Ang II-mediated site-directed assembly of functionally active NAD(P)H oxidase, ROS generation, and activation of redox-sensitive p38MAP kinase and Akt, but not ERK1/2. These findings demonstrate the importance of an intact actin-cytoskeleton in NAD(P)H oxidase regulation and redox signaling by Ang II in human VSMCs.


Assuntos
Citoesqueleto/metabolismo , Proteínas dos Microfilamentos/metabolismo , Músculo Liso Vascular/metabolismo , NADPH Oxidases/metabolismo , Fosfoproteínas/metabolismo , Actinas/metabolismo , Aminoquinolinas/farmacologia , Angiotensina II/farmacologia , Células Cultivadas , Vasos Coronários/citologia , Cortactina , Citocalasina B/farmacologia , Inibidores Enzimáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Músculo Esquelético/irrigação sanguínea , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , NADPH Oxidase 2 , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Superóxidos/metabolismo , Vasoconstritores/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
Can J Cardiol ; 22(7): 565-71, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16755311

RESUMO

The present paper summarizes and highlights key messages of the 2006 Canadian Hypertension Education Program recommendations for the management and diagnosis of hypertension. An important message in the 2006 Canadian Hypertension Education Program recommendations is to improve patient adherence to antihypertensive therapy by incorporating a number of techniques. These new recommendations still need to be incorporated into what remain as the older but still important considerations for the diagnosis, management and treatment of the patient with hypertension, namely, to assess blood pressure in all adults at all appropriate visits, to expedite the diagnosis of hypertension, to assess and manage global cardiovascular risk, to emphasize that lifestyle modifications are the cornerstone of antihypertensive therapy, to treat to target, and to use combinations of antihypertensive medications and lifestyles to achieve recommended targets. Minor changes in pharmacological therapies are discussed, and potentially important aspects related to home and self-monitoring, particularly with respect to patients with masked hypertension (blood pressure controlled in the office but not at home), are introduced.


Assuntos
Anti-Hipertensivos/uso terapêutico , Hipertensão/diagnóstico , Hipertensão/prevenção & controle , Antagonistas Adrenérgicos beta/uso terapêutico , Comitês Consultivos , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Determinação da Pressão Arterial , Bloqueadores dos Canais de Cálcio/uso terapêutico , Canadá , Quimioterapia Combinada , Insuficiência Cardíaca/tratamento farmacológico , Humanos , Estilo de Vida , Infarto do Miocárdio/tratamento farmacológico , Cooperação do Paciente
14.
Circulation ; 101(14): 1653-9, 2000 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-10758046

RESUMO

BACKGROUND: Structural and functional alterations of the vasculature may contribute to complications of hypertension. Because angiotensin II may be pivotal in some of these vascular abnormalities, we tested the hypothesis that the angiotensin type 1 (AT(1)) receptor antagonist losartan, in contrast to the beta-blocker atenolol, would correct resistance artery abnormalities in patients with essential hypertension. METHODS AND RESULTS: Nineteen untreated patients with mild essential hypertension (47+/-2 years, range 30 to 65 years; 57% male) were randomly assigned in double-blind fashion to losartan or atenolol treatment for 1 year. Nine age/sex-matched normotensive subjects were also studied. Both treatments reduced blood pressure to a comparable degree (losartan, from 149+/-4.1/101+/-1.6 to 128+/-3.6/86+/-2.2 mm Hg, P<0.01; atenolol, from 150+/-4.0/99+/-1.2 to 130+/-3.2/84+/-1.4 mm Hg, P<0.01). Resistance arteries (luminal diameter 150 to 350 microm) dissected from gluteal subcutaneous biopsies were studied on a pressurized myograph. After 1 year of treatment, the ratio of the media width to lumen diameter of arteries from losartan-treated patients was significantly reduced (from 8.4+/-0.4% to 6.7+/-0.3%, P<0.01). Arteries from atenolol-treated patients exhibited no significant change (from 8. 3+/-0.3% to 8.8+/-0.5% after treatment). Endothelium-dependent relaxation (acetylcholine-induced) was normalized by losartan (from 82.1+/-4.9% to 94.7+/-1.1%, P<0.01) but not by atenolol (from 80. 4+/-2.7% to 81.7+/-4.6%). Endothelium-independent relaxation (by sodium nitroprusside) was unchanged after treatment. CONCLUSIONS: The AT(1) antagonist losartan corrected the altered structure and endothelial dysfunction of resistance arteries from patients with essential hypertension, whereas the beta-blocker atenolol had no effect.


Assuntos
Antagonistas de Receptores de Angiotensina , Anti-Hipertensivos/uso terapêutico , Artérias/patologia , Endotélio Vascular/fisiopatologia , Hipertensão/patologia , Hipertensão/fisiopatologia , Losartan/uso terapêutico , Acetilcolina/farmacologia , Adulto , Artérias/efeitos dos fármacos , Artérias/fisiopatologia , Atenolol/uso terapêutico , Nádegas/irrigação sanguínea , Método Duplo-Cego , Feminino , Humanos , Hipertensão/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Receptor Tipo 1 de Angiotensina , Receptor Tipo 2 de Angiotensina , Resistência Vascular , Vasodilatação
15.
Arterioscler Thromb Vasc Biol ; 23(6): 981-7, 2003 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-12663375

RESUMO

OBJECTIVE: The aim of this study was to determine molecular mechanisms whereby c-Src regulates angiotensin II (Ang II)-mediated NAD(P)H oxidase-derived *O2- in human vascular smooth muscle cells (VSMCs). METHODS AND RESULTS: VSMCs from human small arteries were studied. Ang II increased NAD(P)H oxidase-mediated generation of *O2- and H2O2 (P<0.01). PP2, c-Src inhibitor, attenuated these effects by 70% to 80%. Immunoprecipitation of p47phox, followed by immunoblotting with antiphosphoserine antibody, demonstrated a rapid increase (1.5- to 2-fold) in p47phox phosphorylation in Ang II-stimulated cells. This was associated with p47phox translocation from cytosol to membrane, as assessed by immunoblotting and immunofluorescence. PP2 abrogated these effects. Long-term Ang II stimulation (6 to 24 hours) increased NAD(P)H oxidase subunit expression. c-Src inhibition decreased abundance of gp91phox, p22phox, and p47phox. Confirmation of c-Src-dependent regulation of NAD(P)H oxidase was tested in VSMCs from c-Src-/- mice. Ang II-induced *O2- generation was lower in c-Src-/- than c-Src+/+ counterparts. This was associated with decreased p47phox phosphorylation, blunted Ang II-stimulated NAD(P)H oxidase activation, and failure of Ang II to increase subunit expression. CONCLUSIONS: c-Src regulates NAD(P)H oxidase-derived *O2- generation acutely by stimulating p47phox phosphorylation and translocation and chronically by increasing protein content of gp91phox, p22phox, and p47phox in Ang II-stimulated cells. These novel findings identify NAD(P)H oxidase subunits, particularly p47phox, as downstream targets of c-Src.


Assuntos
Angiotensina II/farmacologia , Músculo Liso Vascular/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/fisiologia , Superóxidos/metabolismo , Animais , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Cortactina , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Camundongos , Proteínas dos Microfilamentos/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , NADPH Oxidases , Fosforilação , Subunidades Proteicas , Transporte Proteico , Proteínas Proto-Oncogênicas pp60(c-src)/antagonistas & inibidores , Pirimidinas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Receptor Tipo 1 de Angiotensina/biossíntese , Receptor Tipo 1 de Angiotensina/genética
16.
Auton Autacoid Pharmacol ; 35(3): 17-31, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26845248

RESUMO

The in vivo effect of continuous angiotensin II (Ang II) infusion on arterial blood pressure, vascular hypertrophy and α1 -adrenoceptors (α1 -ARs) expression was explored. Alzet(®) minipumps filled with Ang II (200 ng kg(-1)  min(-1) ) were subcutaneously implanted in male Wistar rats (3 months-old). Groups of rats were also treated with losartan, an AT1 R antagonist, or with BMY 7378, a selective α1D -AR antagonist. Blood pressure was measured by tail-cuff; after 2 or 4 weeks of treatment, vessels were isolated for functional and structural analyses. Angiotensin II increased systolic blood pressure. Phenylephrine-induced contraction in aorta was greater (40% higher) in Ang II-treated rats than in the controls, and similar effect occurred with KCl 80 mm. Responses in tail arteries were not significantly different among the different groups. Angiotensin II decreased α1D -ARs without modifying the other α1 -ARs and induced an increase in media thickness (hypertrophy) in aorta, while no structural change occurred in tail artery. Losartan prevented and reversed hypertension and hypertrophy, while BMY 7378 prevented and reversed the aorta's hypertrophic response, without preventing or reversing hypertension. Findings indicate that Ang II-induced aortic hypertrophic response involves Ang II-AT1 Rs and α1D -ARs. Angiotensin II-induced α1D -AR-mediated vascular remodeling occurs independently of hypertension. Findings identify a α1D -AR-mediated process whereby Ang II influences aortic hypertrophy independently of blood pressure elevation.


Assuntos
Angiotensina II/toxicidade , Hipertensão/induzido quimicamente , Hipertensão/fisiopatologia , Músculo Liso Vascular/fisiologia , Receptores Adrenérgicos alfa 1/fisiologia , Bloqueadores do Receptor Tipo 2 de Angiotensina II/farmacologia , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/fisiologia , Relação Dose-Resposta a Droga , Hipertrofia/induzido quimicamente , Hipertrofia/metabolismo , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar
17.
Hypertension ; 30(6): 1440-7, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9403565

RESUMO

Intracellular Ca2+ and pH are potent modulators of growth factor-induced mitogenesis and contraction. This study examined platelet-derived growth factor-(PDGF-BB) and insulin-like growth factor (IGF-1)-mediated signal transduction in primary cultured unpassaged vascular smooth muscle cells (VSMC) from mesenteric arteries of Sprague-Dawley rats. Intracellular free Ca2+ concentration ([Ca2+]i) and intracellular pH (pHi) were measured by fluorescence digital imaging using fura-2 AM and 2'7'-bis(2-carboxyethyl)-5(6)-carboxyfluorescein, respectively. Characteristics of [Ca2+]i transients were determined by pre-exposing cells to Ca2+-free buffer, and involvement of the Na+/Ca2+ exchanger was assessed by withdrawal of extracellular Na+ and by exposure to dimethylbenzamil (Na+/Ca2+ exchange blocker). To determine whether pHi responses were mediated via the Na+/H+ exchanger, cells were preincubated with 10(-5) mol/L 5-(N-ethyl-N-isopropyl)amiloride (a selective Na+/H+ exchange blocker). The role of protein kinase C (PKC) and tyrosine kinases in growth factor signaling was assessed by pre-exposing cells to calphostin C and chelerythrine chloride (selective PKC inhibitors; 10(-5) mol/L) and tyrphostin A23 (a selective tyrosine kinase inhibitor; 10(-5) mol/L). PDGF-BB and IGF-1 (1 to 10 ng/mL) increased [Ca2+]i and pHi in a dose-dependent manner. At concentrations greater than 1 ng/mL both growth factors induced a biphasic [Ca2+]i response with an initial transient peak followed by a sustained elevation. At 5 ng/mL PDGF-BB and IGF-1 significantly increased [Ca2+]i from 95+/-3 nmol/L to 328+/-28 and 251+/-18 nmol/L, respectively. Ca2+ withdrawal abolished the second phase of [Ca2+]i elevation. Agonist-induced [Ca2+]i responses were similarly altered by Na+ withdrawal, by Na+/ Ca2+ exchange blockade, and by PKC inhibition; latency, the period from stimulus application to the first [Ca2+]i peak, was increased, the initial [Ca2+]i peak was attenuated, and the sustained phase was prolonged. PDGF-BB and IGF-1 (10 ng/mL) significantly increased pHi from 6.89+/-0.04 nmol/L to 7.11+/-0.01 and 7.09+/-0.02 nmol/L, respectively. EIPA and calphostin C completely inhibited agonist-elicited alkalinization. Tyrphostin A-23 abolished second-messenger responses to PDGF-BB and IGF-1, whose receptors have tyrosine kinase activity. In conclusion, PDGF-BB and IGF-1 elicit significant [Ca2+]i and pHi responses in VSMC. The underlying pathways that mediate these responses are partially dependent on Na+/ Ca2+ transporters and the Na+/H+ exchanger, both of which are linked to PKC activation.


Assuntos
Cálcio/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Músculo Liso Vascular/fisiologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteína Quinase C/metabolismo , Transdução de Sinais/efeitos dos fármacos , Alcaloides , Animais , Becaplermina , Benzofenantridinas , Cálcio/farmacologia , Células Cultivadas , Concentração de Íons de Hidrogênio , Cinética , Masculino , Artérias Mesentéricas/efeitos dos fármacos , Artérias Mesentéricas/fisiologia , Músculo Liso Vascular/efeitos dos fármacos , Naftalenos/farmacologia , Fenantridinas/farmacologia , Proteínas Proto-Oncogênicas c-sis , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia , Sódio/farmacologia
18.
Hypertension ; 22(6): 853-62, 1993 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8244517

RESUMO

The aims of this study were to determine the relations between platelet free calcium concentrations ([Ca2+]i), intracellular pH (pHi), and aggregation and to assess the effects of angiotensin II (Ang II) and endothelin-1 on these platelet parameters in normotensive subjects and hypertensive patients. Seventeen normotensive subjects, 25 untreated hypertensive patients, and 34 treated hypertensive patients were studied. Platelet cytosolic free [Ca2+]i and pHi were measured spectrofluorometrically using specific fluorescent probes (fura 2-AM and BCECF-AM, respectively) in unstimulated and Ang II- and endothelin-1-stimulated platelets. Aggregation was measured by a turbidometric technique. Basal [Ca2+]i (141 +/- 11 nmol/L) and pH (7.16 +/- 0.01) were higher (P < .05) in the untreated hypertensive group compared with the normotensive (118 +/- 9 nmol/L, 7.11 +/- 0.01, respectively) and treated hypertensive (121 +/- 11 nmol/L, 7.12 +/- 0.01, respectively) groups. In the combined normotensive and hypertensive groups, there were significant correlations between [Ca2+]i and mean arterial pressure (r = .75, P < .01), pHi and mean arterial pressure (r = .72, P < .01), [Ca2+]i and pHi (r = .71, P < .01), [Ca2+]i and aggregation (r = .69, P < .02), and pHi and aggregation (r = .56, P < .05). Ang II stimulation significantly increased [Ca2+]i and pHi in the untreated hypertensive and normotensive groups. The net change in [Ca2+]i induced by Ang II was significantly higher (P < .05) in the untreated hypertensive group compared with the other groups (67 +/- 6 nmol/L for the untreated hypertensive group versus 54 +/- 5 and 29 +/- 8 nmol/L for the normotensive and treated hypertensive groups, respectively). In the presence of Ang II, thrombin-induced aggregatory responses were increased in all three groups, but the maximal response was significantly higher in the untreated hypertensive group compared with the other groups (P < .05). Endothelin-1 increased pHi through endothelin A-receptors (effect blocked by the specific antagonist BQ-123) but had no significant effect on [Ca2+]i or aggregation. However, endothelin-1 blunted thrombin-induced platelet aggregation in normotensive subjects but not in hypertensive patients. In conclusion, increased Ang II-stimulated [Ca2+]i and pHi in platelets of essential hypertensive patients may be associated with increased aggregatory responses. The stimulatory effect of endothelin-1 on pHi but not on [Ca2+]i or aggregation suggests that in platelets endothelin-induced signaling pathways other than phospholipase C may be involved.(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Angiotensina II/farmacologia , Plaquetas/efeitos dos fármacos , Endotelinas/farmacologia , Hipertensão/sangue , Agregação Plaquetária/efeitos dos fármacos , Adulto , Idoso , Amilorida/farmacologia , Análise de Variância , Plaquetas/química , Plaquetas/ultraestrutura , Cálcio/análise , Citosol/química , Citosol/efeitos dos fármacos , Relação Dose-Resposta a Droga , Antagonistas dos Receptores de Endotelina , Feminino , Humanos , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Masculino , Pessoa de Meia-Idade , Peptídeos Cíclicos/farmacologia , Receptor de Endotelina A , Receptores de Endotelina/metabolismo , Análise de Regressão , Saralasina/farmacologia
19.
Hypertension ; 27(5): 1097-103, 1996 May.
Artigo em Inglês | MEDLINE | ID: mdl-8621202

RESUMO

Tyrosine kinases have been implicated in vascular smooth muscle cell proliferation and contraction. Underlying mechanisms may involve C(a2+) -dependent pathways. This study assesses relationships between angiotensin II (Ang II)-stimulated phospholipase C-mediated Ca2+ transients and tyrosine kinase-dependent pathways in vascular smooth muscle cells. Intracellular free Ca2+ concentration ([Ca2+]i) was measured in primary cultured unpassaged vascular smooth muscle cells derived from mesenteric resistance vessels of Wistar-Kyoto rats with the use of fura 2 methodology. [Ca2+]i effects of Ang II (1 nmol/L) were determined in vascular smooth muscle cells in which tyrosine kinase pathways were stimulated by insulin (70 muU/mL; 0.5 nmol/L), insulin-like growth factor-I (1 ng/mL; 0.13 nmol/L), or platelet-derived growth factor-BB (1 ng/mL; 0.04 nmol/L) and in cells in which tyrosine kinase was inhibited by specific inhibitors (1 mumol/L tyrphostin A-23 and genistein). Ang II elicited a rapid and transient [Ca2+]i response (from 94 +/- 8 to 239 +/- 5.8 nmol/L). Activation of the receptor tyrosine kinase by insulin, platelet-derived growth factor, and insulin-like growth factor-I significantly reduced (P < .01) Ang II-induced [Ca2+]i to 161 +/- 7, 189 +/- 3.7, and 183 +/- 5 nmol/L, respectively. In the presence of tyrphostin A-23 and genistein, Ang II-stimulated [Ca2+]i remained persistently elevated and failed to return to basal levels. Tyrphostin A-1, the inactive tyrphostin analogue, had not significant effect on Ang II-induced [Ca2+]i. This study demonstrates that activation of tyrosine kinase pathways reduces Ang II-elicited [Ca2+]i responses, whereas tyrosine kinase inhibition prevents [Ca2+]i recovery after agonist stimulation. Interaction between tyrosine kinase- and phospholipase C-dependent signaling pathways modulates vascular smooth muscle cell [Ca2+]i responses to Ang II.


Assuntos
Angiotensina II/farmacologia , Cálcio/metabolismo , Membranas Intracelulares/metabolismo , Músculo Liso Vascular/metabolismo , Proteínas Tirosina Quinases/fisiologia , Transdução de Sinais , Animais , Genisteína , Substâncias de Crescimento/farmacologia , Insulina/farmacologia , Isoflavonas/farmacologia , Masculino , Músculo Liso Vascular/citologia , Concentração Osmolar , Proteínas Tirosina Quinases/antagonistas & inibidores , Ratos , Ratos Endogâmicos WKY
20.
Hypertension ; 30(2 Pt 1): 222-9, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9260984

RESUMO

Angiotensin II (Ang II), a potent vasoactive peptide with mitogenic potential, influences vascular smooth muscle cell contraction and growth through receptor-linked pathways that increase intracellular free Ca2+ concentration ([Ca2+]i) and pH (pHi). Activation of these second messengers by Ang II may involve tyrosine kinase-dependent signaling pathways. This study determined the role of tyrosine kinases in Ang II-stimulated pHi, and in simultaneously measured contractile and [Ca2+]i responses, as well as growth in cultured vascular smooth muscle cells from mesenteric arteries of Wistar-Kyoto rats. pHi was determined by fluorescent digital imaging using 2',7'-bis(2-carboxyethyl)-5(6)-carboxyfluorescein acetoxymethyl ester (BCECF-AM). Vascular smooth muscle cell [Ca2+]i and contractile responses were assessed simultaneously by fura 2 methodology and by photomicroscopy in cells grown on rat tail collagen gels. Cell growth was determined by DNA and protein synthesis as measured by [3H]thymidine and [3H]leucine incorporation, respectively. The Ang II receptor subtypes (AT1 or AT2) through which Ang II mediates effects were assessed with [Sar1,Ile8]Ang II (a nonselective subtype antagonist), losartan (a selective AT1 antagonist), and PD 123319 (a selective AT2 antagonist). To determine whether tyrosine kinases influence Ang II-stimulated responses, cells were pretreated with 10(-5) mol/L tyrphostin A-23 (a specific tyrosine kinase inhibitor). Ang II increased pHi in a dose-dependent manner (pD2, 9.2+/-0.2) and significantly increased vascular smooth muscle cell contraction (30%) and [Ca2+]i (pD2, 7.4+/-0.1). Ang II (10(-7) mol/L) increased DNA ([3H]thymidine incorporation) and protein synthesis ([3H]leucine incorporation). [Sar1,Ile8]Ang II and losartan but not PD 123319 abolished Ang II-elicited responses. Tyrphostin A-23 significantly attenuated Ang II-stimulated pHi responses; it also inhibited [Ca2+]i and contractile responses and cell growth. The inactive analogue tyrphostin A-1 did not alter Ang II-stimulated actions. These results provide novel evidence for a role of tyrosine kinases in Ang II-mediated pHi responses in vascular smooth muscle cells and indicate that tyrosine kinases participate in the regulation of signal transduction associated with AT1 receptor subtype-mediated contraction and growth.


Assuntos
Angiotensina II/fisiologia , Hidrogênio/metabolismo , Músculo Liso Vascular/metabolismo , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais , Tirfostinas , Vasoconstrição/fisiologia , Animais , Cálcio/metabolismo , Catecóis/farmacologia , Divisão Celular , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Concentração de Íons de Hidrogênio , Membranas Intracelulares/metabolismo , Leucina/metabolismo , Masculino , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Nitrilas/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Ratos , Ratos Endogâmicos WKY , Receptores de Angiotensina/metabolismo , Timidina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA