Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Stroke Cerebrovasc Dis ; 23(6): 1429-39, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24529602

RESUMO

BACKGROUND: Statins, or 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors, have been suggested to possess pleiotropic effects, including antioxidant and anti-inflammatory properties. We investigated the protective effects of pretreatment with rosuvastatin, a relatively hydrophilic statin, on early brain injury (EBI) after a subarachnoid hemorrhage (SAH), using the endovascular perforation SAH model. METHODS: Eighty-six male Sprague-Dawley rats were randomly divided into 3 groups: (1) sham operation, (2) SAH+vehicle, and (3) SAH+10 mg/kg rosuvastatin. Rosuvastatin or vehicle was orally administered to rats once daily from 7 days before to 1 day after the SAH operation. After SAH, we examined the effects of rosuvastatin on the neurologic score, brain water content, neuronal cell death estimated by terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate nick end labeling staining, blood-brain barrier disruption by immunoglobulin G (IgG) extravasation, oxidative stress, and proinflammatory molecules. RESULTS: Compared with the vehicle group, rosuvastatin significantly improved the neurologic score and reduced the brain water content, neuronal cell death, and IgG extravasation. Rosuvastatin inhibited brain superoxide production, nuclear factor-kappa B (NF-κB) activation, and the increase in activated microglial cells after SAH. The increased expressions of tumor necrosis factor-alpha, endothelial matrix metalloproteinase-9, and neuronal cyclooxygenase-2 induced by SAH were prevented by rosuvastatin pretreatment. CONCLUSIONS: The present study demonstrates that rosuvastatin pretreatment ameliorates EBI after SAH through the attenuation of oxidative stress and NF-κB-mediated inflammation.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Fluorbenzenos/uso terapêutico , NF-kappa B/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Pirimidinas/uso terapêutico , Hemorragia Subaracnóidea/tratamento farmacológico , Sulfonamidas/uso terapêutico , Superóxidos/metabolismo , Animais , Apoptose/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Lesões Encefálicas/etiologia , Lesões Encefálicas/metabolismo , Fluorbenzenos/farmacologia , Masculino , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Pirimidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Rosuvastatina Cálcica , Hemorragia Subaracnóidea/complicações , Hemorragia Subaracnóidea/metabolismo , Sulfonamidas/farmacologia
2.
Arterioscler Thromb Vasc Biol ; 34(3): 616-25, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24371084

RESUMO

OBJECTIVE: There are currently no specific strategies for the treatment or prevention of vascular dementia. White matter lesions, a common pathology in cerebral small vessel disease, are a major cause of vascular dementia. We investigated whether apoptosis signal-regulating kinase 1 (ASK1) might be a key molecule in cerebral hypoperfusion, associated with blood-brain barrier breakdown and white matter lesions. APPROACH AND RESULTS: A mouse model of cognitive impairment was developed by inducing chronic cerebral hypoperfusion in white matter including the corpus callosum via bilateral common carotid artery stenosis (BCAS) surgery. BCAS-induced white matter lesions caused cognitive decline in C57BL/6J (wild-type) mice but not in ASK1-deficient (ASK1(-/-)) mice. Phosphorylated ASK1 increased in wild-type mouse brains, and phosphorylated p38 and tumor necrosis factor-α expression increased in corpus callosum cerebral endothelial cells after BCAS in wild-type mice but not in ASK1(-/-) mice. BCAS decreased claudin-5 expression and disrupted blood-brain barrier in the corpus callosum of wild-type but not ASK1(-/-) mice. Cerebral nitrotyrosine was increased in wild-type and ASK1(-/-) BCAS mice. Cerebral phosphorylated ASK1 did not increase in wild-type mice treated with NADPH-oxidase inhibitor. A p38 inhibitor and NADPH-oxidase inhibitor mimicked the protective effect of ASK1 deficiency against cognitive impairment. Specific ASK1 inhibitor prevented cognitive decline in BCAS mice. In vitro oxygen-glucose deprivation and tumor necrosis factor-α stimulation caused the disruption of endothelial tight junctions from wild-type mice but not ASK1(-/-) mice. CONCLUSIONS: Oxidative stress-ASK1-p38 cascade plays a role in the pathogenesis of cognitive impairment, through blood-brain barrier breakdown via the disruption of endothelial tight junctions. ASK1 might be a promising therapeutic target for chronic cerebral hypoperfusion-induced cognitive impairment.


Assuntos
Estenose das Carótidas/complicações , Demência Vascular/etiologia , MAP Quinase Quinase Quinase 5/fisiologia , Animais , Barreira Hematoencefálica , Isquemia Encefálica/etiologia , Isquemia Encefálica/fisiopatologia , Isquemia Encefálica/prevenção & controle , Isquemia Encefálica/psicologia , Estenose das Carótidas/fisiopatologia , Estenose das Carótidas/psicologia , Circulação Cerebrovascular/efeitos dos fármacos , Condicionamento Operante/fisiologia , Corpo Caloso/irrigação sanguínea , Demência Vascular/enzimologia , Demência Vascular/fisiopatologia , Demência Vascular/prevenção & controle , Células Endoteliais/enzimologia , Comportamento Exploratório , MAP Quinase Quinase Quinase 5/antagonistas & inibidores , MAP Quinase Quinase Quinase 5/deficiência , MAP Quinase Quinase Quinase 5/genética , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Neuroglia/fisiologia , Estresse Oxidativo , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional , Reconhecimento Psicológico , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Junções Íntimas , Fator de Necrose Tumoral alfa/farmacologia
3.
Brain Res ; 1519: 87-94, 2013 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-23632378

RESUMO

This study aimed to examine the potential protective effect of rosuvastatin against cerebral ischemia/reperfusion injury and its mechanisms. Forty-eight male SD rats underwent 90 min of transient middle cerebral artery occlusion (tMCAO), followed by reperfusion. Rats were orally given (1) rosuvastatin 1mg/kg, (2) rosuvastatin 10mg/kg or (3) water (vehicle) once a day from 7 days before to 1 day after induction of tMCAO. Neurological score, infarct volume, and oxidative stress-related molecules (assessed by immunohistochemistry, dihydroethidium staining, or western blotting) were estimated at 24h after reperfusion. Rosuvastatin prevented the impairment of neurological function and decreased the infarct volume, compared with the vehicle group. The increases in activated microglia, macrophage, and superoxide levels usually caused by ischemia/reperfusion were significantly ameliorated by rosuvastatin. Rosuvastatin also inhibited the upregulation of gp91(phox) and p22phox, phosphorylation of nuclear factor-kappa B, and induction of cyclooxygenase 2 and inducible nitric oxide synthase, compared with vehicle. The results suggest that pretreatment with rosuvastatin may be a promising therapeutic strategy for cerebral ischemia/reperfusion injury, through attenuation of oxidative stress and inflammation.


Assuntos
Isquemia Encefálica/prevenção & controle , Encefalite/tratamento farmacológico , Fluorbenzenos/administração & dosagem , Inibidores de Hidroximetilglutaril-CoA Redutases/administração & dosagem , Estresse Oxidativo/efeitos dos fármacos , Pirimidinas/administração & dosagem , Traumatismo por Reperfusão/prevenção & controle , Sulfonamidas/administração & dosagem , Animais , Infarto Encefálico/etiologia , Infarto Encefálico/prevenção & controle , Proteínas de Ligação ao Cálcio/metabolismo , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Esquema de Medicação , Ectodisplasinas/metabolismo , Encefalite/etiologia , Masculino , Glicoproteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , NADPH Oxidase 2 , NADPH Oxidases/metabolismo , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Ratos , Ratos Sprague-Dawley , Rosuvastatina Cálcica , Regulação para Cima/efeitos dos fármacos
4.
Kidney Int ; 83(4): 582-92, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23325087

RESUMO

The accumulation of p-cresyl sulfate (PCS), a uremic toxin, is associated with the mortality rate of chronic kidney disease patients; however, the biological functions and the mechanism of its action remain largely unknown. Here we determine whether PCS enhances the production of reactive oxygen species (ROS) in renal tubular cells resulting in cytotoxicity. PCS exhibited pro-oxidant properties in human tubular epithelial cells by enhancing NADPH oxidase (nicotinamide adenine dinucleotide phosphate-oxidase) activity. PCS also upregulated mRNA levels of inflammatory cytokines and active TGF-ß1 protein secretion associated with renal fibrosis. Knockdown of p22(phox) or Nox4 expression suppressed the effect of PCS, underlining the importance of NADPH oxidase activation on its mechanism of action. PCS also reduced cell viability by increasing ROS production. The toxicity of PCS was largely suppressed in the presence of probenecid, an organic acid transport inhibitor. Administration of PCS for 4 weeks caused significant renal tubular damage in 5/6-nephrectomized rats by enhancing oxidative stress. Thus, the renal toxicity of PCS is attributed to its intracellular accumulation, leading to both increased NADPH oxidase activity and ROS production, which, in turn, triggers induction of inflammatory cytokines involved in renal fibrosis. This mechanism is similar to that for the renal toxicity of indoxyl sulfate.


Assuntos
Cresóis/toxicidade , Células Epiteliais/efeitos dos fármacos , Túbulos Renais Proximais/efeitos dos fármacos , NADPH Oxidases/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Insuficiência Renal Crônica/induzido quimicamente , Ésteres do Ácido Sulfúrico/toxicidade , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Citocinas/genética , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Células Epiteliais/enzimologia , Células Epiteliais/patologia , Fibrose , Humanos , Mediadores da Inflamação/metabolismo , Túbulos Renais Proximais/enzimologia , Túbulos Renais Proximais/patologia , Masculino , NADPH Oxidase 4 , NADPH Oxidases/genética , Nefrectomia , Probenecid/farmacologia , Interferência de RNA , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Insuficiência Renal Crônica/enzimologia , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/patologia , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Transfecção , Fator de Crescimento Transformador beta1/metabolismo
5.
Am J Hypertens ; 25(6): 704-10, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22421905

RESUMO

BACKGROUND: The pharmacological advantage of combination of an angiotensin receptor blocker (ARB) and a calcium-channel blocker (CCB) is not fully defined. This study was undertaken to elucidate the potential benefit of their combination in metabolic syndrome. METHODS: SHR/NDmcr-cp (SHRcp), a rat model of human metabolic syndrome, were divided into four groups, and were administered (i) vehicle, (ii) candesartan (an ARB) 0.3 mg/kg/day, (iii) amlodipine (a CCB) 3 mg/kg/day, and (iv) candesartan 0.3 mg/kg/day plus amlodipine 3 mg/kg/day, for 4 weeks. RESULTS: Candesartan, amlodipine, or their combination significantly ameliorated the impairment of vascular endothelium-dependent relaxation with acetylcholine in SHRcp. However, the impairment of insulin-induced vasodilation in SHRcp was partially improved by candesartan alone, but not by amlodipine alone. Interestingly, amlodipine added to candesartan synergistically enhanced the improvement of impaired insulin-induced vasodilation by candesartan, indicating the synergistic improvement of vascular insulin resistance by the combination of these drugs. Candesartan alone, but not amlodipine alone, significantly attenuated vascular superoxide and NADPH oxidase subunit p22phox in SHRcp. Amlodipine added to candesartan synergistically enhanced the reduction of vascular p22phox levels and superoxide by candesartan in SHRcp, suggesting the association of vascular insulin resistance with oxidative stress. Furthermore, the combination of candesartan with amlodipine synergistically decreased the increase in visceral adipocyte size, serum free-fatty acid, and tumor necrosis factor-α in SHRcp. CONCLUSIONS: ARB and CCB combination synergistically ameliorated vascular insulin resistance in metabolic syndrome, being associated with the synergistic attenuation of vascular oxidative stress and metabolic disorders.


Assuntos
Anlodipino/uso terapêutico , Benzimidazóis/uso terapêutico , Resistência à Insulina/fisiologia , Síndrome Metabólica/tratamento farmacológico , Síndrome Metabólica/fisiopatologia , Estresse Oxidativo/fisiologia , Tetrazóis/uso terapêutico , Anlodipino/farmacologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Animais , Benzimidazóis/farmacologia , Compostos de Bifenilo , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Bloqueadores dos Canais de Cálcio/farmacologia , Bloqueadores dos Canais de Cálcio/uso terapêutico , Modelos Animais de Doenças , Sinergismo Farmacológico , Quimioterapia Combinada , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Masculino , Síndrome Metabólica/metabolismo , NADPH Oxidases/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Endogâmicos WKY , Superóxidos/metabolismo , Tetrazóis/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatação/fisiologia
6.
Hypertens Res ; 35(2): 194-200, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22089532

RESUMO

This study was undertaken to elucidate a novel mechanism underlying angiotensin II-induced cardiac injury, focusing on the role of oxidative stress and myocardial capillary density. Salt-loaded Dahl salt-sensitive hypertensive rats (DS rats), a useful model for hypertensive cardiac remodeling or heart failure, were orally given irbesartan (an AT1 receptor blocker), tempol (a superoxide dismutase mimetic) or hydralazine (a vasodilator). Irbesartan significantly ameliorated left ventricular ischemia and prevented the development of cardiac hypertrophy and fibrosis in DS rats. The benefits were associated with the attenuation of oxidative stress, normalization of myocardial capillary density and inhibition of capillary endothelial apoptosis. Moreover, DS rats with significant cardiac hypertrophy and fibrosis displayed decreased myocardial vascular endothelial growth factor (VEGF) expression and increased cardiac apoptosis signal-regulating kinase 1 (ASK1) activation. Treatment with irbesartan significantly reversed these phenotypes. Tempol treatment of DS rats mimicked all the above-mentioned effects of irbesartan, indicating the critical role of oxidative stress in cardiac injury. We also investigated the role of VEGF and ASK1 in oxidative stress-induced endothelial apoptosis by using cultured endothelial cells from wild-type and ASK1-deficient mice. Oxidative stress-induced ASK1 activation led to endothelial apoptosis, and VEGF treatment prevented oxidative stress-induced endothelial apoptosis by inhibiting ASK1 activation. We obtained the first evidence that oxidative stress-induced cardiac VEGF repression and ASK1 activation caused the enhancement of endothelial apoptosis and contributed to a decrease in myocardial capillary density. These effects resulted in angiotensin II-induced progression of cardiac injury.


Assuntos
Angiotensina II/toxicidade , Cardiopatias/induzido quimicamente , Hipertensão/fisiopatologia , MAP Quinase Quinase Quinase 5/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Anti-Hipertensivos/farmacologia , Apoptose/efeitos dos fármacos , Compostos de Bifenilo/farmacologia , Pressão Sanguínea/fisiologia , Western Blotting , Capilares/efeitos dos fármacos , Células Cultivadas , Óxidos N-Cíclicos/farmacologia , Células Endoteliais/efeitos dos fármacos , Cardiopatias/diagnóstico por imagem , Cardiopatias/patologia , Hidralazina/farmacologia , Hipertensão/complicações , Hipertensão/diagnóstico por imagem , Hipertrofia Ventricular Esquerda/patologia , Imuno-Histoquímica , Irbesartana , Isquemia Miocárdica/fisiopatologia , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Endogâmicos Dahl , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Marcadores de Spin , Superóxidos/metabolismo , Tetrazóis/farmacologia , Ultrassonografia
7.
Arterioscler Thromb Vasc Biol ; 31(12): 2872-80, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21940947

RESUMO

OBJECTIVE: Whole-body periodic acceleration (WBPA) has been developed as a passive exercise technique to improve endothelial function by increasing shear stress through repetitive movements in spinal axis direction. We investigated the effects of WBPA on blood flow recovery in a mouse model of hindlimb ischemia and in patients with peripheral arterial disease. METHODS AND RESULTS: After unilateral femoral artery excision, mice were assigned to either the WBPA (n=15) or the control (n=13) group. WBPA was applied at 150 cpm for 45 minutes under anesthesia once a day. WBPA significantly increased blood flow recovery after ischemic surgery, as determined by laser Doppler perfusion imaging. Sections of ischemic adductor muscle stained with anti-CD31 antibody showed a significant increase in capillary density in WBPA mice compared with control mice. WBPA increased the phosphorylation of endothelial nitric oxide synthase (eNOS) in skeletal muscle. The proangiogenic effect of WBPA on ischemic limb was blunted in eNOS-deficient mice, suggesting that the stimulatory effects of WBPA on revascularization are eNOS dependent. Quantitative real-time polymerase chain reaction analysis showed significant increases in angiogenic growth factor expression in ischemic hindlimb by WBPA. Facilitated blood flow recovery was observed in a mouse model of diabetes despite there being no changes in glucose tolerance and insulin sensitivity. Furthermore, both a single session and 7-day repeated sessions of WBPA significantly improved blood flow in the lower extremity of patients with peripheral arterial disease. CONCLUSIONS: WBPA increased blood supply to ischemic lower extremities through activation of eNOS signaling and upregulation of proangiogenic growth factor in ischemic skeletal muscle. WBPA is a potentially suitable noninvasive intervention to facilitate therapeutic angiogenesis.


Assuntos
Aceleração , Membro Posterior/irrigação sanguínea , Isquemia/terapia , Terapia Passiva Contínua de Movimento/métodos , Neovascularização Fisiológica/fisiologia , Doença Arterial Periférica/terapia , Condicionamento Físico Animal/métodos , Idoso , Animais , Modelos Animais de Doenças , Endotélio Vascular/fisiopatologia , Feminino , Fator 2 de Crescimento de Fibroblastos/metabolismo , Seguimentos , Membro Posterior/metabolismo , Humanos , Isquemia/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Músculo Esquelético/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Doença Arterial Periférica/fisiopatologia , Proteínas Proto-Oncogênicas c-sis/metabolismo , Fluxo Sanguíneo Regional/fisiologia , Resultado do Tratamento , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
Biochem Biophys Res Commun ; 410(3): 508-13, 2011 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-21679694

RESUMO

Experimental and clinical data support the notion that peroxisome proliferator-activated receptor γ (PPARγ) activation is associated with anti-atherosclerosis as well as anti-diabetic effect. Telmisartan, an angiotensin receptor blocker (ARB), acts as a partial PPARγ agonist. We hypothesized that telmisartan protects against diabetic vascular complications, through PPARγ activation. We compared the effects of telmisartan, telmisartan combined with GW9662 (a PPARγ antagonist), and losartan with no PPARγ activity on vascular injury in obese type 2 diabetic db/db mice. Compared to losartan, telmisartan significantly ameliorated vascular endothelial dysfunction, downregulation of phospho-eNOS, and coronary arterial remodeling in db/db mice. More vascular protective effects of telmisartan than losartan were associated with greater anti-inflammatory effects of telmisartan, as shown by attenuation of vascular nuclear factor kappa B (NFκB) activation and tumor necrosis factor α. Coadministration of GW9662 with telmisartan abolished the above mentioned greater protective effects of telmisartan against vascular injury than losartan in db/db mice. Thus, PPARγ activity appears to be involved in the vascular protective effects of telmisartan in db/db mice. Moreover, telmisartan, but not losartan, prevented the downregulation of vascular PPARγ in db/db mice and this effect of telmisartan was cancelled by the coadministration of GW9662. Our data provided the first evidence indicating that PPARγ activity of telmisartan contributed to the protective effects of telmisartan against diabetic vascular complication. PPARγ activity of telmisartan was involved in the normalization of vascular PPARγ downregulation in diabetic mice. Thus, telmisartan seems to exert vascular protective effects in hypertensive patients with diabetes.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Benzimidazóis/uso terapêutico , Benzoatos/uso terapêutico , Diabetes Mellitus Tipo 2/complicações , Angiopatias Diabéticas/prevenção & controle , Obesidade/complicações , PPAR gama/metabolismo , Anilidas/administração & dosagem , Animais , Angiopatias Diabéticas/etiologia , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Telmisartan
9.
Hypertens Res ; 34(7): 817-24, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21471977

RESUMO

The clinical value of the combination of amlodipine and eplerenone is unclear. This study was undertaken to test whether eplerenone potentiates the protective effects of amlodipine against hypertensive cardiovascular injury. Salt-loaded Dahl salt-sensitive hypertensive rats (DS rats) were given (1) vehicle, (2) an antihypertensive dose of amlodipine, (3) a non-antihypertensive dose of eplerenone or (4) combined amlodipine and eplerenone for 6 weeks, and the effects on cardiovascular injuries were compared. There was no significant difference among the four groups regarding plasma aldosterone, urine volume or urinary electrolytes. A subpressor dose of eplerenone markedly ameliorated vascular endothelial dysfunction, cardiac inflammation and fibrosis in DS rats to a similar degree as an antihypertensive dose of amlodipine. Addition of eplerenone to amlodipine, without affecting blood pressure, enhanced the improvement by amlodipine of vascular endothelial function, cardiac inflammation, fibrosis and diastolic dysfunction in DS rats. Additive beneficial effects of eplerenone were attributed to additive potentiation of eNOS and Akt phosphorylation and additive reduction of oxidative stress. Eplerenone significantly attenuated cardiovascular NADPH oxidase activity by reducing gp91(phox) upregulation and attenuated the upregulation of cardiovascular AT1 receptor, but amlodipine failed to affect them. Thus, the normalization by eplerenone of gp91(phox) and AT1 receptor upregulation seems to be at least partially responsible for the additive benefits of eplerenone in the prevention of hypertensive cardiovascular injury. The combination of amlodipine and eplerenone may be a promising therapeutic strategy for cardiovascular disease in salt-sensitive hypertension.


Assuntos
Anlodipino/uso terapêutico , Anti-Hipertensivos/uso terapêutico , Coração/efeitos dos fármacos , Hipertensão/tratamento farmacológico , Antagonistas de Receptores de Mineralocorticoides/uso terapêutico , Espironolactona/análogos & derivados , Aldosterona/sangue , Anlodipino/administração & dosagem , Animais , Anti-Hipertensivos/administração & dosagem , Pressão Sanguínea/efeitos dos fármacos , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/metabolismo , Quimioterapia Combinada , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiopatologia , Eplerenona , Hipertensão/fisiopatologia , Masculino , Antagonistas de Receptores de Mineralocorticoides/administração & dosagem , Miocárdio/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Endogâmicos Dahl , Receptor Tipo 1 de Angiotensina/metabolismo , Cloreto de Sódio na Dieta/farmacologia , Espironolactona/administração & dosagem , Espironolactona/uso terapêutico , Superóxidos/metabolismo , Resultado do Tratamento
10.
Biochem Biophys Res Commun ; 403(3-4): 258-63, 2010 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-21059340

RESUMO

The effect of calcium channel blockers (CCBs) on type 2 diabetes is still unclear. The present study was undertaken to examine the efficacy of nifedipine, a dihydropyridine CCB, on obesity, glucose intolerance and vascular endothelial dysfunction in db/db mice (a mouse model of obesity and type 2 diabetes). db/db mice, fed high-fat diet (HFD) were treated with vehicle, nifedipine (10 mg kg(-1) day(-1)) or hydralazine (5 mg kg(-1) day(-1)) for 4 weeks, and the protective effects were compared. Although nifedipine and hydralazine exerted similar blood pressure lowering in db/db mice, neither affected body weight, fat weight, and glucose intolerance of db/db mice. However, nifedipine, but not hydralazine, significantly improved vascular endothelial function in db/db mice, being accompanied by more attenuation of vascular superoxide by nifedipine than hydralazine. These protective effects of nifedipine were attributed to the attenuation of eNOS uncoupling as shown by the prevention of vascular endothelial nitric oxide synthase (eNOS) dimer disruption, and the prevention of dihydrofolate reductase (DHFR) downregulation, the key enzyme responsible for eNOS uncoupling. Moreover, nifedipine, but not hydralazine, significantly prevented the decreases in phosphorylation of vascular akt and eNOS in db/db mice. Our work provided the first evidence that nifedipine prevents vascular endothelial dysfunction, through the inhibition of eNOS uncoupling and the enhancement of eNOS phosphorylation, independently of blood pressure-lowering effect. We propose that nifedipine may be a promising therapeutic agent for cardiovascular complications in type 2 diabetes.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Diabetes Mellitus Experimental/enzimologia , Diabetes Mellitus Tipo 2/enzimologia , Endotélio Vascular/efeitos dos fármacos , Nifedipino/farmacologia , Óxido Nítrico Sintase Tipo III/metabolismo , Obesidade/enzimologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Modelos Animais de Doenças , Endotélio Vascular/enzimologia , Endotélio Vascular/fisiopatologia , Teste de Tolerância a Glucose , Camundongos , Camundongos Endogâmicos , Tamanho do Órgão/efeitos dos fármacos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Superóxidos/metabolismo
11.
J Hypertens ; 28(2): 340-52, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19864959

RESUMO

OBJECTIVES: The efficacy of renin-angiotensin system (RAS) blockers on type 2 diabetes and its complications remains to be defined. This study was undertaken to test the hypothesis that candesartan may enhance the protective effects of pioglitazone against type 2 diabetes. METHODS: We compared the effects of pioglitazone, candesartan, and their combination on cardiorenal and vascular injury, diabetes, and tissue oxidative stress in obese and type 2 diabetic db/db mice, and also examined the effects of tempol, a superoxide dismutase (SOD) mimetic, on db/db mice to define the role of oxidative stress. RESULTS: The addition of candesartan to pioglitazone significantly potentiated the suppressive effects of pioglitazone on cardiac macrophage infiltration and interstitial fibrosis, and glomerular macrophage infiltration and sclerosis in db/db mice. These benefits of the combination of pioglitazone and candesartan in db/db mice were attributed to additive attenuation of cardiorenal oxidative stress, through the attenuation of NADPH oxidase or the restoration of Cu/Zn-SOD and EC-SOD. The combination of these drugs reversed vascular endothelial dysfunction in db/db mice more than either monotherapy, by causing more phosphorylation of eNOS. Candesartan slightly augmented the improvement of glucose tolerance by pioglitazone in db/db mice, and this additive effect was mediated by more attenuation of oxidative stress. CONCLUSIONS: Our work demonstrated that candesartan significantly potentiated the protective effects of pioglitazone against cardiorenal and vascular injury, and diabetes in obese type 2 diabetic mice. Thus, the combination of pioglitazone with candesartan is potentially a promising therapeutic strategy for type 2 diabetes.


Assuntos
Benzimidazóis/administração & dosagem , Diabetes Mellitus Tipo 2/tratamento farmacológico , Angiopatias Diabéticas/prevenção & controle , Nefropatias Diabéticas/prevenção & controle , Tetrazóis/administração & dosagem , Tiazolidinedionas/administração & dosagem , Bloqueadores do Receptor Tipo 1 de Angiotensina II/administração & dosagem , Animais , Antioxidantes/farmacologia , Compostos de Bifenilo , Óxidos N-Cíclicos/farmacologia , Grupo dos Citocromos b/metabolismo , Diabetes Mellitus Tipo 2/fisiopatologia , Sinergismo Farmacológico , Hipoglicemiantes/administração & dosagem , Rim/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Miocárdio/metabolismo , NADPH Oxidase 2 , NADPH Oxidases/metabolismo , Obesidade/complicações , Obesidade/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Pioglitazona , Marcadores de Spin , Superóxido Dismutase/metabolismo
12.
Hypertension ; 54(3): 544-51, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19581502

RESUMO

The molecular mechanism underlying aldosterone/salt-induced cardiovascular injury remains to be defined. This work was undertaken to determine the role of apoptosis signal-regulating kinase 1 (ASK1) in the mechanism underlying aldosterone-induced cardiac injury in vivo. We compared the in vivo effects of 4 weeks of aldosterone/salt treatment on wild-type and ASK1-deficient mice. Aldosterone infusion plus high salt intake in wild-type mice significantly increased blood pressure and urinary albumin excretion and decreased plasma potassium concentrations, and these effects of aldosterone/salt were not affected by ASK1 deficiency. Thus, ASK1 seems to play a minor role in aldosterone-induced hypertension and renal injury. ASK1 deficiency also failed to affect aldosterone-induced cardiac hypertrophy. However, ASK1 deficiency markedly ameliorated aldosterone-induced cardiac injury, eg, the enhancement of cardiac macrophage infiltration, monocyte chemotactic protein 1 expression, interstitial fibrosis, perivascular fibrosis, and transforming growth factor-beta1 and collagen type I expressions. Thus, ASK1 participates in aldosterone-induced cardiac inflammation and fibrosis. Furthermore, the enhancement of NADPH oxidase-mediated cardiac oxidative stress caused by aldosterone infusion was markedly lessened by ASK1 deficiency, which was associated with the significant amelioration by ASK1 deficiency of aldosterone-induced cardiac Nox2 upregulation. Furthermore, aldosterone/salt treatment significantly enhanced cardiac expression of the angiotensin-converting enzyme and angiotensin II type 1 receptor in wild-type mice, whereas the enhancement of these proteins by aldosterone/salt was abolished by ASK1 deficiency. Our results demonstrate that ASK1 is implicated in aldosterone/salt-induced cardiac inflammation and fibrosis through the enhancement of NADPH oxidase-mediated oxidative stress and the upregulation of the cardiac renin-angiotensin system.


Assuntos
Aldosterona/toxicidade , MAP Quinase Quinase Quinase 5/metabolismo , Miocárdio/patologia , Cloreto de Sódio na Dieta/toxicidade , Albuminúria/urina , Aldosterona/administração & dosagem , Animais , Pressão Sanguínea/efeitos dos fármacos , Quimiocina CCL2/genética , Feminino , Fibrose/sangue , Fibrose/etiologia , Fibrose/urina , Expressão Gênica/efeitos dos fármacos , Inflamação/sangue , Inflamação/etiologia , Inflamação/urina , MAP Quinase Quinase Quinase 5/genética , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Miocárdio/metabolismo , NADPH Oxidases/metabolismo , Peptidil Dipeptidase A/genética , Potássio/sangue , Receptor Tipo 1 de Angiotensina/genética , Sódio/sangue , Cloreto de Sódio na Dieta/administração & dosagem , Superóxidos/metabolismo , Fator de Crescimento Transformador beta1/genética
13.
Stroke ; 39(11): 3049-56, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18688015

RESUMO

BACKGROUND AND PURPOSE: The precise mechanism of salt-induced brain injury is unclear. We examined the detailed causative role of angiotensin II and NADPH oxidase in salt-accelerated brain injury of stroke-prone spontaneously hypertensive rats (SHRSP). METHODS: We examined the effect of salt loading on brain reactive oxygen species (ROS), inflammation, and apoptosis in SHRSP. Salt-loaded SHRSP were given vehicle, valsartan (an angiotensin AT1 receptor blocker), or hydralazine to compare their efficacy on brain injury. We also examined the efficacy of apocynin (a NADPH oxidase inhibitor) on brain injury of salt-loaded SHRSP. RESULTS: Cerebral NADPH oxidase activity and ROS in SHRSP were already increased at 1 week after salt loading followed by the significant increase in ED-1-positive cells and neuronal apoptosis. Thus, cerebral NADPH oxidase activation preceded cerebral inflammation and neuronal apoptosis. Despite comparable hypotensive effects between valsartan and hydralazine in salt-loaded SHRSP, valsartan reduced cerebral NADPH oxidase activity and ROS more than hydralazine being accompanied by more prevention of stroke by valsartan than hydralazine. Valsartan, but not hydralazine, prevented neuronal apoptosis, being associated with the suppression of apoptosis signal-regulating kinase 1 activation by valsartan. Moreover, cerebral inflammation was also prevented by valsartan more than hydralazine, being associated with more suppression of monocyte chemotactic protein-1 and tumor necrosis factor-alpha expressions by valsartan. Thus, angiotensin II was directly involved in salt-induced neuronal NADPH oxidase activation, ROS, apoptosis, and inflammation in SHRSP. Apocynin attenuated the enhancement of ROS, cerebral inflammation, neuronal apoptosis, and apoptosis signal-regulating kinase 1 activation and prevented stroke in salt-loaded SHRSP, indicating the causative role of cerebral NADPH oxidase in salt-induced brain injury. CONCLUSIONS: We obtained the evidence that excess salt, through ROS produced by angiotensin II-activated NADPH oxidase, caused cerebral neuronal apoptosis and inflammation as well as stroke in SHRSP.


Assuntos
Angiotensina II/metabolismo , Apoptose/fisiologia , Córtex Cerebral/citologia , Córtex Cerebral/patologia , Inflamação/metabolismo , NADPH Oxidases/metabolismo , Neurônios/metabolismo , Cloreto de Sódio/efeitos adversos , Acetofenonas/metabolismo , Animais , Anti-Hipertensivos/farmacologia , Astrócitos/citologia , Astrócitos/metabolismo , Pressão Sanguínea/fisiologia , Córtex Cerebral/efeitos dos fármacos , Ativação Enzimática , Inibidores Enzimáticos/metabolismo , Humanos , Hidralazina/farmacologia , Hipertensão/patologia , Hipertensão/fisiopatologia , Masculino , NADPH Oxidases/genética , Neurônios/citologia , Ratos , Ratos Endogâmicos SHR , Espécies Reativas de Oxigênio/metabolismo , Cloreto de Sódio/administração & dosagem , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia , Tetrazóis/farmacologia , Valina/análogos & derivados , Valina/farmacologia , Valsartana
14.
Stroke ; 38(11): 3016-22, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17885259

RESUMO

BACKGROUND AND PURPOSE: Very recent subgroup analysis from the PROspective pioglitAzone Clinical Trial In macroVascular Events has shown that pioglitazone reduces the risk of recurrent stroke in type 2 diabetic patients. However, the underlying mechanism of stroke prevention by pioglitazone is unknown. Our aim was to examine the effect of pioglitazone on hypertension-based stroke in rats. METHODS: Pioglitazone (1 mg x kg(-1) x d(-1)) was orally administered to stroke-prone spontaneously hypertensive rats (SHRSP) to examine the effect on incidental stroke, cerebrovascular injury, brain inflammation, oxidative stress, and vascular endothelial dysfunction induced by hypertension. RESULTS: Treatment of SHRSP with pioglitazone for 4 weeks, without affecting blood pressure and blood glucose values, improved vascular endothelial dysfunction (P<0.05), suppressed remodeling of the middle cerebral artery (P<0.05) and brain microvessels (P<0.05), and inhibited brain macrophage infiltration (P<0.05) and the upregulation of brain monocyte chemoattractant protein-1 and tumor necrosis factor-alpha expression (P<0.01). Furthermore, pioglitazone treatment significantly delayed the onset of stroke signs and death in SHRSP (P<0.05). These beneficial effects of pioglitazone on cerebrovascular injury and stroke in SHRSP were associated with a reduction of brain and vascular superoxide via the inhibition of NADPH oxidase activity. CONCLUSIONS: Our work provides the first evidence that pioglitazone significantly protects against hypertension-induced cerebrovascular injury and stroke by improving vascular endothelial dysfunction, inhibiting brain inflammation, and reducing oxidative stress. These beneficial effects of pioglitazone were independent of blood pressure or blood sugar values. Thus, pioglitazone appears to be a potential therapeutic agent for stroke in type 2 diabetes with hypertension.


Assuntos
Hipertensão/complicações , Fármacos Neuroprotetores/farmacologia , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/fisiopatologia , Tiazolidinedionas/farmacologia , Animais , Glicemia/efeitos dos fármacos , Glicemia/fisiologia , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Artérias Cerebrais/efeitos dos fármacos , Artérias Cerebrais/metabolismo , Artérias Cerebrais/fisiopatologia , Citocinas/efeitos dos fármacos , Citocinas/metabolismo , Complicações do Diabetes/fisiopatologia , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Hipertensão/fisiopatologia , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Microcirculação/efeitos dos fármacos , Microcirculação/metabolismo , Microcirculação/fisiopatologia , NADH NADPH Oxirredutases/antagonistas & inibidores , NADH NADPH Oxirredutases/metabolismo , NADPH Oxidase 1 , Fármacos Neuroprotetores/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Pioglitazona , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Acidente Vascular Cerebral/etiologia , Superóxidos/antagonistas & inibidores , Superóxidos/metabolismo , Tiazolidinedionas/uso terapêutico , Resultado do Tratamento
15.
Hypertension ; 50(3): 519-24, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17646574

RESUMO

Long-term treatment with N(omega)-nitro-l-arginine methylester (l-NAME), an NO synthase inhibitor, induces hypertension and cardiovascular injury. However, its precise mechanism is unknown. Using apoptosis signal-regulating kinase-1 (ASK1)-deficient mice, we investigated the role of ASK1 in cardiovascular injury caused by l-NAME treatment. l-NAME was orally administered to ASK1-deficient and C57BL/6J (wild) mice for 8 weeks. l-NAME treatment increased blood pressure of wild and ASK1-deficient mice to a similar extent, indicating no role of ASK1 in NO-deficient hypertension. l-NAME treatment significantly impaired acetylcholine-induced carotid arterial relaxation in wild mice (P<0.01), being associated with the decreased endothelial NO synthase (eNOS) activity (P<0.01) and the increased disruption of eNOS dimer (P<0.01), whereas these changes by l-NAME were substantially attenuated in ASK1-deficient mice. Thus, ASK1 is involved in the impairment of vascular endothelial function by reducing eNOS activity and disrupting eNOS dimer. l-NAME treatment increased vascular reduced nicotinamide-adenine dinucleotide phosphate oxidase activity and superoxide in wild mice to a greater extent than in ASK1 deficient mice. l-NAME treatment in wild mice caused cardiac hypertrophy, myocyte apoptosis, macrophage infiltration, coronary arterial remodeling, interstitial fibrosis, and the expression of monocyte chemoattractant protein-1 and transforming growth factor-beta1, whereas these cardiac changes by l-NAME were absent in ASK1-deficient mice. Cardiac reduced nicotinamide-adenine dinucleotide phosphate oxidase activation and superoxide elevation by l-NAME were much less in ASK1-deficient mice than in wild mice. Our work provided the first evidence that ASK1 is implicated in vascular endothelial dysfunction and cardiovascular remodeling induced by NO deficiency by regulating eNOS and reduced nicotinamide-adenine dinucleotide phosphate oxidase.


Assuntos
Endotélio Vascular/fisiopatologia , MAP Quinase Quinase Quinase 5/metabolismo , Óxido Nítrico/deficiência , Remodelação Ventricular , Animais , Apoptose/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Cardiomegalia/induzido quimicamente , Cardiomegalia/patologia , Quimiocina CCL2/metabolismo , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/patologia , Vasos Coronários/fisiopatologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Inibidores Enzimáticos/farmacologia , MAP Quinase Quinase Quinase 5/deficiência , Macrófagos/patologia , Camundongos , Camundongos Knockout , Células Musculares , NADPH Oxidases/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/metabolismo , Óxidos de Nitrogênio/sangue , Superóxidos/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Remodelação Ventricular/efeitos dos fármacos
16.
Eur J Biochem ; 271(16): 3330-9, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15291810

RESUMO

HAND2/dHAND is a basic helix-loop-helix transcription factor expressed in the heart and neural crest derivatives during embryogenesis. Although dHAND is essential for branchial arch, cardiovascular and limb development, its target genes have not been identified. The regulatory mechanisms of dHAND function also remain relatively unknown. Here we report that Akt/PKB, a serine/threonine protein kinase involved in cell survival, growth and differentiation, phosphorylates dHAND and inhibits dHAND-mediated transcription. AU5-dHAND expressed in 293T cells became phosphorylated, possibly at its Akt phosphorylation motif, in the absence of kinase inhibitors, whereas the phosphatidylinositol 3-kinase inhibitor wortmannin and the Akt inhibitor NL-71-101, but not the p70 S6 kinase inhibitor rapamycin, significantly reduced dHAND phosphorylation. Coexpression of HA-Akt augmented dHAND phosphorylation at multiple serine and threonine residues mainly located in the bHLH domain and, as a result, decreased the transcriptional activity of dHAND. Consistently, alanine mutation mimicking the nonphosphorylation state abolished the inhibitory effect of Akt on dHAND, whereas aspartate mutation mimicking the phosphorylation state resulted in a loss of dHAND transcriptional activity. These changes in dHAND transcriptional activity were in parallel with changes in the DNA binding activity rather than in dimerization activity. These results suggest that Akt-mediated signaling may regulate dHAND transcriptional activity through the modulation of its DNA binding activity during embryogenesis.


Assuntos
Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Linhagem Celular , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Ensaio de Desvio de Mobilidade Eletroforética , Regulação da Expressão Gênica , Humanos , Camundongos , Dados de Sequência Molecular , Mutação/genética , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-akt , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/genética , Ativação Transcricional , Proteínas de Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA