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











Base de dados
Intervalo de ano de publicação
1.
Circ Res ; 132(2): 154-166, 2023 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-36575984

RESUMO

BACKGROUND: Hydrogen sulfide is a critical endogenous signaling molecule that exerts protective effects in the setting of heart failure. Cystathionine γ-lyase (CSE), 1 of 3 hydrogen-sulfide-producing enzyme, is predominantly localized in the vascular endothelium. The interaction between the endothelial CSE-hydrogen sulfide axis and endothelial-mesenchymal transition, an important pathological process contributing to the formation of fibrosis, has yet to be investigated. METHODS: Endothelial-cell-specific CSE knockout and Endothelial cell-CSE overexpressing mice were subjected to transverse aortic constriction to induce heart failure with reduced ejection fraction. Cardiac function, vascular reactivity, and treadmill exercise capacity were measured to determine the severity of heart failure. Histological and gene expression analyses were performed to investigate changes in cardiac fibrosis and the activation of endothelial-mesenchymal transition. RESULTS: Endothelial-cell-specific CSE knockout mice exhibited increased endothelial-mesenchymal transition and reduced nitric oxide bioavailability in the myocardium, which was associated with increased cardiac fibrosis, impaired cardiac and vascular function, and worsened exercise performance. In contrast, genetic overexpression of CSE in endothelial cells led to increased myocardial nitric oxide, decreased endothelial-mesenchymal transition and cardiac fibrosis, preserved cardiac and endothelial function, and improved exercise capacity. CONCLUSIONS: Our data demonstrate that endothelial CSE modulates endothelial-mesenchymal transition and ameliorate the severity of pressure-overload-induced heart failure, in part, through nitric oxide-related mechanisms. These data further suggest that endothelium-derived hydrogen sulfide is a potential therapeutic for the treatment of heart failure with reduced ejection fraction.


Assuntos
Insuficiência Cardíaca , Sulfeto de Hidrogênio , Disfunção Ventricular Esquerda , Camundongos , Animais , Sulfeto de Hidrogênio/metabolismo , Células Endoteliais/metabolismo , Óxido Nítrico/metabolismo , Camundongos Knockout , Endotélio Vascular/metabolismo , Fibrose
2.
Circ Res ; 131(3): 222-235, 2022 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-35701874

RESUMO

BACKGROUND: Hydrogen sulfide (H2S) exerts mitochondria-specific actions that include the preservation of oxidative phosphorylation, biogenesis, and ATP synthesis, while inhibiting cell death. 3-MST (3-mercaptopyruvate sulfurtransferase) is a mitochondrial H2S-producing enzyme whose functions in the cardiovascular disease are not fully understood. In the current study, we investigated the effects of global 3-MST deficiency in the setting of pressure overload-induced heart failure. METHODS: Human myocardial samples obtained from patients with heart failure undergoing cardiac surgeries were probed for 3-MST protein expression. 3-MST knockout mice and C57BL/6J wild-type mice were subjected to transverse aortic constriction to induce pressure overload heart failure with reduced ejection fraction. Cardiac structure and function, vascular reactivity, exercise performance, mitochondrial respiration, and ATP synthesis efficiency were assessed. In addition, untargeted metabolomics were utilized to identify key pathways altered by 3-MST deficiency. RESULTS: Myocardial 3-MST was significantly reduced in patients with heart failure compared with nonfailing controls. 3-MST KO mice exhibited increased accumulation of branched-chain amino acids in the myocardium, which was associated with reduced mitochondrial respiration and ATP synthesis, exacerbated cardiac and vascular dysfunction, and worsened exercise performance following transverse aortic constriction. Restoring myocardial branched-chain amino acid catabolism with 3,6-dichlorobenzo1[b]thiophene-2-carboxylic acid (BT2) and administration of a potent H2S donor JK-1 ameliorates the detrimental effects of 3-MST deficiency in heart failure with reduced ejection fraction. CONCLUSIONS: Our data suggest that 3-MST derived mitochondrial H2S may play a regulatory role in branched-chain amino acid catabolism and mediate critical cardiovascular protection in heart failure.


Assuntos
Insuficiência Cardíaca , Sulfeto de Hidrogênio , Disfunção Ventricular Esquerda , Trifosfato de Adenosina/metabolismo , Aminoácidos de Cadeia Ramificada/metabolismo , Animais , Insuficiência Cardíaca/metabolismo , Humanos , Sulfeto de Hidrogênio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Miocárdio/metabolismo , Disfunção Ventricular Esquerda/metabolismo
3.
J Am Heart Assoc ; 9(19): e017544, 2020 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-32990120

RESUMO

Background Hydrogen sulfide (H2S) is an important endogenous physiological signaling molecule and exerts protective properties in the cardiovascular system. Cystathionine γ-lyase (CSE), 1 of 3 H2S producing enzyme, is predominantly localized in the vascular endothelium. However, the regulation of CSE in vascular endothelium remains incompletely understood. Methods and Results We generated inducible endothelial cell-specific CSE overexpressed transgenic mice (EC-CSE Tg) and endothelial cell-specific CSE knockout mice (EC-CSE KO), and investigated vascular function in isolated thoracic aorta, treadmill exercise capacity, and myocardial injury following ischemia-reperfusion in these mice. Overexpression of CSE in endothelial cells resulted in increased circulating and myocardial H2S and NO, augmented endothelial-dependent vasorelaxation response in thoracic aorta, improved exercise capacity, and reduced myocardial-reperfusion injury. In contrast, genetic deletion of CSE in endothelial cells led to decreased circulating H2S and cardiac NO production, impaired endothelial dependent vasorelaxation response and reduced exercise capacity. However, myocardial-reperfusion injury was not affected by genetic deletion of endothelial cell CSE. Conclusions CSE-derived H2S production in endothelial cells is critical in maintaining endothelial function, exercise capacity, and protecting against myocardial ischemia/reperfusion injury. Our data suggest that the endothelial NO synthase-NO pathway is likely involved in the beneficial effects of overexpression of CSE in the endothelium.


Assuntos
Cistationina gama-Liase/metabolismo , Células Endoteliais/metabolismo , Tolerância ao Exercício/fisiologia , Sulfeto de Hidrogênio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Óxido Nítrico/metabolismo , Animais , Aorta Torácica/metabolismo , Aorta Torácica/fisiopatologia , Camundongos , Camundongos Transgênicos , Miocárdio/metabolismo , Miocárdio/patologia , Óxido Nítrico Sintase/metabolismo , Transdução de Sinais
4.
J Am Coll Cardiol ; 70(17): 2139-2153, 2017 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-29050562

RESUMO

BACKGROUND: Sustained sympathetic activation contributes to the progression of myocardial cell injury, cardiac fibrosis, and left ventricular (LV) dysfunction in heart failure (HF). OBJECTIVES: This study investigated the effects of radiofrequency renal nerve denervation (RF-RDN) on the pathobiology of HF and the interaction between the renal sympathetic nerves and natriuretic peptide (NP) metabolism. METHODS: Spontaneously hypertensive rats (SHR) and normotensive Wistar-Kyoto rats (WKY) were subjected to 45 min of coronary artery ligation and reperfusion for 12 weeks. At 4 weeks post-reperfusion, SHR and WKY underwent either bilateral RF-RDN or sham-RDN. RESULTS: Following RF-RDN in both strains, LV ejection fraction remained significantly above those levels in respective sham-RDN rats, and at the end of the 12-week study, rats in both strains had significantly reduced LV fibrosis and improved vascular function. RF-RDN therapy significantly improved vascular reactivity to endothelium-dependent and -independent vasodilators as well as vascular compliance in the setting of severe HF. Improvements in LV function were accompanied by significant elevations in circulating NP as compared to those associated with sham-RDN. Further investigation into the cause of increased circulating NP levels demonstrated that RF-RDN significantly inhibited renal neprilysin activity in SHR and WKY with HF. Likewise, chronic treatment with the beta1 antagonist bisoprolol inhibited renal neprilysin activity and increased circulation NP levels in WKY with HF. CONCLUSIONS: This study identifies a novel endogenous pathway by which the renal nerves participate in the degradation of cardioprotective NP. Furthermore, removal of the influence of the renal nerves on kidney function attenuates renal neprilysin activity, augments circulating NP levels, reduces myocardial fibrosis, and improves LV function in the setting of HF.


Assuntos
Insuficiência Cardíaca/terapia , Rim/inervação , Neprilisina/antagonistas & inibidores , Simpatectomia , Aminobutiratos/farmacologia , Angiotensina II/sangue , Animais , Compostos de Bifenilo , Bisoprolol/farmacologia , Pressão Sanguínea , Combinação de Medicamentos , Ecocardiografia , Miocárdio/química , Miocárdio/patologia , Neprilisina/fisiologia , Nitritos/análise , Norepinefrina/sangue , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Artéria Renal/inervação , Renina/sangue , Traumatismo por Reperfusão/fisiopatologia , Tetrazóis/farmacologia , Valsartana , Função Ventricular Esquerda/fisiologia
5.
Peptides ; 95: 25-32, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28720396

RESUMO

Mitoxantrone (MXT) is an androstenedione that is used to treat cancers and progressive forms of multiple sclerosis; however, its use is limited by its cardiotoxicity. Pituitary adenylate cyclase activating polypeptide (PACAP) is a member of the secretin/growth hormone-releasing hormone/vasoactive intestinal peptide family and has many functions, including cytoprotection and immunosuppression. We tested the hypothesis that PACAP can protect against MXT-induced cardiotoxicity in mice. Female BALB/c mice were treated once weekly for 4 weeks with saline (n=14) or MXT (3mg/kg, i.p.; n=14). Half of the mice in each group received PACAP (10µg, i.p.) 1h before and 24 and 48h after MXT, while the remaining mice received injections of saline on the same schedule. Echocardiography was used to assess cardiac structure and function. In mice treated with MXT and saline, body weight was significantly reduced after the third dose of MXT. PACAP significantly attenuated the reduction in body weight; however, the weights did not return to control level. Compared to controls, MXT-treated mice had significantly increased left ventricular (LV) diameter and LV volume and decreased LV posterior wall thickness. Fractional shortening (FS) and ejection fraction (EF) were also significantly decreased. Treatment with PACAP prevented MXT-induced LV dilation and significantly attenuated the reductions in FS and EF, although FS and EF did not return to control level. PACAP38 did not prevent MXT-induced decreases in LV posterior wall thickness. MXT dose-dependently decreased the viability of cultured U937 (human leukemia) cells; PACAP did not protect cultured U937 cells from MXT-mediated cell death. In conclusion, PACAP can attenuate MXT-mediated LV dilation and dysfunction in mice.


Assuntos
Traumatismos Cardíacos/tratamento farmacológico , Mitoxantrona/efeitos adversos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Disfunção Ventricular Esquerda/tratamento farmacológico , Animais , Cardiotoxicidade/tratamento farmacológico , Cardiotoxicidade/patologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Traumatismos Cardíacos/induzido quimicamente , Traumatismos Cardíacos/patologia , Humanos , Camundongos , Mitoxantrona/uso terapêutico , Neoplasias/complicações , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Substâncias Protetoras/administração & dosagem , Disfunção Ventricular Esquerda/induzido quimicamente , Disfunção Ventricular Esquerda/patologia
6.
Circ Res ; 121(1): 43-55, 2017 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-28512108

RESUMO

RATIONALE: Neurogenic hypertension is characterized by an increase in sympathetic activity and often resistance to drug treatments. We previously reported that it is also associated with a reduction of angiotensin-converting enzyme type 2 (ACE2) and an increase in a disintegrin and metalloprotease 17 (ADAM17) activity in experimental hypertension. In addition, while multiple cells within the central nervous system have been involved in the development of neurogenic hypertension, the contribution of ADAM17 has not been investigated. OBJECTIVE: To assess the clinical relevance of this ADAM17-mediated ACE2 shedding in hypertensive patients and further identify the cell types and signaling pathways involved in this process. METHODS AND RESULTS: Using a mass spectrometry-based assay, we identified ACE2 as the main enzyme converting angiotensin II into angiotensin-(1-7) in human cerebrospinal fluid. We also observed an increase in ACE2 activity in the cerebrospinal fluid of hypertensive patients, which was correlated with systolic blood pressure. Moreover, the increased level of tumor necrosis factor-α in those cerebrospinal fluid samples confirmed that ADAM17 was upregulated in the brain of hypertensive patients. To further assess the interaction between brain renin-angiotensin system and ADAM17, we generated mice lacking angiotensin II type 1 receptors specifically on neurons. Our data reveal that despite expression on astrocytes and other cells types in the brain, ADAM17 upregulation during deoxycorticosterone acetate-salt hypertension occurs selectively on neurons, and neuronal angiotensin II type 1 receptors are indispensable to this process. Mechanistically, reactive oxygen species and extracellular signal-regulated kinase were found to mediate ADAM17 activation. CONCLUSIONS: Our data demonstrate that angiotensin II type 1 receptors promote ADAM17-mediated ACE2 shedding in the brain of hypertensive patients, leading to a loss in compensatory activity during neurogenic hypertension.


Assuntos
Proteína ADAM17/fisiologia , Hipertensão/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Peptidil Dipeptidase A/metabolismo , Receptor Tipo 1 de Angiotensina/fisiologia , Adulto , Angiotensina II/metabolismo , Enzima de Conversão de Angiotensina 2 , Animais , Animais Recém-Nascidos , Células Cultivadas , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos
7.
Methods Mol Biol ; 1527: 117-126, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28116711

RESUMO

Angiotensin-converting enzyme 2 (ACE2) is a component of the renin-angiotensin system (RAS) which plays an important role in the regulation of blood pressure and volume homeostasis. Accumulating evidence shows alterations in ACE2 expression and activity in several hypertensive animal models, as well as in patients with hypertension. In order to assess the role of brain ACE2 in hypertension, a specific ACE2 assay is required. Based on a quenched fluorescent substrate, we describe an easy-to-use method for determining ACE2 activity in brain tissue and cerebrospinal fluid. The method can further be adapted for other tissues, plasma, cell extracts, and cell culture supernatants.


Assuntos
Encéfalo/metabolismo , Ensaios Enzimáticos/métodos , Peptidil Dipeptidase A/metabolismo , Enzima de Conversão de Angiotensina 2 , Animais , Pressão Sanguínea/genética , Pressão Sanguínea/fisiologia , Humanos , Hipertensão/líquido cefalorraquidiano , Hipertensão/metabolismo , Peptídeos , Peptidil Dipeptidase A/líquido cefalorraquidiano , Sistema Renina-Angiotensina/fisiologia
8.
Am J Physiol Heart Circ Physiol ; 309(5): H926-34, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26254330

RESUMO

We previously reported that type 2 angiotensin-converting enzyme (ACE2) compensatory activity is impaired by the disintegrin and metalloprotease 17 (ADAM17), and lack of ACE2 is associated with oxidative stress in neurogenic hypertension. To investigate the relationship between ADAM17 and oxidative stress, Neuro2A cells were treated with ANG II (100 nM) 24 h after vehicle or α-lipoic acid (LA, 500 µM). ADAM17 expression was increased by ANG II (120.5 ± 9.1 vs. 100.2 ± 0.8%, P < 0.05) and decreased after LA (69.0 ± 0.3 vs. 120.5 ± 9.1%, P < 0.05). In another set of experiments, LA reduced ADAM17 (92.9 ± 5.3 vs. 100.0 ± 11.2%, P < 0.05) following its overexpression. Moreover, ADAM17 activity was reduced by LA in ADAM17-overexpressing cells [109.5 ± 19.8 vs. 158.0 ± 20.0 fluorescence units (FU)·min(-1)·µg protein(-1), P < 0.05], in which ADAM17 overexpression increased oxidative stress (114.1 ± 2.5 vs. 101.0 ± 1.0%, P < 0.05). Conversely, LA-treated cells attenuated ADAM17 overexpression-induced oxidative stress (76.0 ± 9.1 vs. 114.1 ± 2.5%, P < 0.05). In deoxycorticosterone acetate (DOCA)-salt hypertensive mice, a model in which ADAM17 expression and activity are increased, hypertension was blunted by pretreatment with LA (119.0 ± 2.4 vs. 131.4 ± 2.2 mmHg, P < 0.05). In addition, LA improved dysautonomia and baroreflex sensitivity. Furthermore, LA blunted the increase in NADPH oxidase subunit expression, as well as the increase in ADAM17 and decrease in ACE2 activity in the hypothalamus of DOCA-salt hypertensive mice. Taken together, these data suggest that LA might preserve ACE2 compensatory activity by breaking the feedforward cycle between ADAM17 and oxidative stress, resulting in a reduction of neurogenic hypertension.


Assuntos
Proteínas ADAM/metabolismo , Antioxidantes/farmacologia , Hipertensão/metabolismo , Estresse Oxidativo , Ácido Tióctico/farmacologia , Proteínas ADAM/genética , Proteína ADAM17 , Angiotensina II/farmacologia , Enzima de Conversão de Angiotensina 2 , Animais , Antioxidantes/uso terapêutico , Barorreflexo , Linhagem Celular Tumoral , Hipertensão/tratamento farmacológico , Hipotálamo/citologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NADPH Oxidases/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Peptidil Dipeptidase A/metabolismo , Ácido Tióctico/uso terapêutico
9.
Hypertension ; 65(3): 577-86, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25489058

RESUMO

Overactivity of the renin-angiotensin system, oxidative stress, and cyclooxygenases (COX) in the brain are implicated in the pathogenesis of hypertension. We previously reported that angiotensin-converting enzyme 2 (ACE2) overexpression in the brain attenuates the development of deoxycorticosterone acetate-salt hypertension, a neurogenic hypertension model with enhanced brain renin-angiotensin system and sympathetic activity. To elucidate the mechanisms involved, we investigated whether oxidative stress, mitogen-activated protein kinase signaling and cyclooxygenase (COX) activation in the brain are modulated by ACE2 in neurogenic hypertension. Deoxycorticosterone acetate-salt hypertension significantly increased expression of Nox-2 (+61±5%), Nox-4 (+50±13%), and nitrotyrosine (+89±32%) and reduced activity of the antioxidant enzymes, catalase (-29±4%) and superoxide dismutase (-31±7%), indicating increased oxidative stress in the brain of nontransgenic mice. This increased oxidative stress was attenuated in transgenic mice overexpressing ACE2 in the brain. Deoxycorticosterone acetate-salt-induced reduction of neuronal nitric oxide synthase expression (-26±7%) and phosphorylated endothelial nitric oxide synthase/total endothelial nitric oxide synthase (-30±3%), and enhanced phosphorylation of protein kinase B and extracellular signal-regulated kinase 1/2 in the paraventricular nucleus, were reversed by ACE2 overexpression. In addition, ACE2 overexpression blunted the hypertension-mediated increase in gene and protein expression of COX-1 and COX-2 in the paraventricular nucleus. Furthermore, gene silencing of either COX-1 or COX-2 in the brain, reduced microglial activation and accompanied neuroinflammation, ultimately attenuating Deoxycorticosterone acetate-salt hypertension. Together, these data provide evidence that brain ACE2 overexpression reduces oxidative stress and COX-mediated neuroinflammation, improves antioxidant and nitric oxide signaling, and thereby attenuates the development of neurogenic hypertension.


Assuntos
Encéfalo/metabolismo , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Encefalite/prevenção & controle , Hipertensão/prevenção & controle , Proteínas de Membrana/metabolismo , Peptidil Dipeptidase A/metabolismo , Regulação para Cima , Enzima de Conversão de Angiotensina 2 , Animais , Antioxidantes/metabolismo , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 2/genética , Acetato de Desoxicorticosterona/efeitos adversos , Modelos Animais de Doenças , Encefalite/metabolismo , Inativação Gênica , Hipertensão/induzido quimicamente , Hipertensão/metabolismo , Isoenzimas/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Óxido Nítrico Sintase/metabolismo , Estresse Oxidativo/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo
10.
Am J Physiol Regul Integr Comp Physiol ; 308(5): R370-8, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25519733

RESUMO

Endoplasmic reticulum (ER) stress was previously reported to contribute to neurogenic hypertension while neuronal angiotensin-converting enzyme type 2 (ACE2) overexpression blunts the disease. To assess which brain regions are important for ACE2 beneficial effects and the contribution of ER stress to neurogenic hypertension, we first used transgenic mice harboring a floxed neuronal hACE2 transgene (SL) and tested the impact of hACE2 knockdown in the subfornical organ (SFO) and paraventricular nucleus (PVN) on deoxycorticosterone acetate (DOCA)-salt hypertension. SL and nontransgenic (NT) mice underwent DOCA-salt or sham treatment while infected with an adenoassociated virus (AAV) encoding Cre recombinase (AAV-Cre) or a control virus (AAV-green fluorescent protein) to the SFO or PVN. DOCA-salt-induced hypertension was reduced in SL mice, with hACE2 overexpression in the brain. This reduction was only partially blunted by knockdown of hACE2 in the SFO or PVN, suggesting that both regions are involved but not essential for ACE2 regulation of blood pressure (BP). DOCA-salt treatment did not increase the protein levels of ER stress and autophagy markers in NT mice, despite a significant increase in BP. In addition, these markers were not affected by hACE2 overexpression in the brain, despite a significant reduction of hypertension in SL mice. To further assess the role of ER stress in neurogenic hypertension, NT mice were infused intracerebroventricularlly with tauroursodeoxycholic acid (TUDCA), an ER stress inhibitor, during DOCA-salt treatment. However, TUDCA infusion failed to blunt the development of hypertension in NT mice. Our data suggest that brain ER stress does not contribute to DOCA-salt hypertension and that ACE2 blunts neurogenic hypertension independently of ER stress.


Assuntos
Encéfalo/enzimologia , Acetato de Desoxicorticosterona , Estresse do Retículo Endoplasmático , Retículo Endoplasmático/enzimologia , Hipertensão/prevenção & controle , Peptidil Dipeptidase A/metabolismo , Cloreto de Sódio na Dieta , Enzima de Conversão de Angiotensina 2 , Animais , Biomarcadores/metabolismo , Pressão Sanguínea , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Retículo Endoplasmático/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Hipertensão/enzimologia , Hipertensão/genética , Hipertensão/fisiopatologia , Infusões Intraventriculares , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Núcleo Hipotalâmico Paraventricular/enzimologia , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Peptidil Dipeptidase A/genética , Órgão Subfornical/enzimologia , Órgão Subfornical/fisiopatologia , Ácido Tauroquenodesoxicólico/administração & dosagem , Fatores de Tempo , Regulação para Cima
11.
CNS Neurosci Ther ; 20(5): 452-9, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24581232

RESUMO

BACKGROUND: The angiotensin (Ang) converting enzyme 2 (ACE2)/Ang-(1-7)/Mas receptor pathway is an important component of the renin-angiotensin system and has been suggested to exert beneficial effects in ischemic stroke. AIMS: This study explored whether the ACE2/Ang-(1-7)/Mas pathway has a protective effect on cerebral ischemic injury and whether this effect is affected by age. METHODS: We used three-month and eight-month transgenic mice with neural over-expression of ACE2 (SA) and their age-matched nontransgenic (NT) controls. Neurological deficits and ischemic stroke volume were determined following middle cerebral artery occlusion (MCAO). In oxygen and glucose deprivation (OGD) experiments on brain slices, the effects of the Mas receptor agonist (Ang1-7) or antagonist (A779) on tissue swelling, Nox2/Nox4 expression reactive oxygen species (ROS) production and cell death were measured. RESULTS: (1) Middle cerebral artery occlusion -induced ischemic injury and neurological deficit were reduced in SA mice, especially in eight-month animals; (2) OGD-induced tissue swelling and cell death were decreased in SA mice with a greater reduction seen in eight-month mice; (3) Ang-(1-7) and A779 had opposite effects on OGD-induced responses, which correlated with changes in Nox2/Nox4 expression and ROS production. CONCLUSIONS: Angiotensin converting enzyme 2/Ang-(1-7)/Mas axis protects brain from ischemic injury via the Nox/ROS signaling pathway, with a greater effect in older animals.


Assuntos
Angiotensina II/análogos & derivados , Angiotensina I/metabolismo , Isquemia Encefálica/fisiopatologia , Encéfalo/fisiopatologia , Neurônios/fisiologia , Fragmentos de Peptídeos/metabolismo , Peptidil Dipeptidase A/metabolismo , Fatores Etários , Angiotensina II/metabolismo , Enzima de Conversão de Angiotensina 2 , Animais , Encéfalo/patologia , Edema Encefálico/etiologia , Edema Encefálico/patologia , Edema Encefálico/fisiopatologia , Isquemia Encefálica/etiologia , Isquemia Encefálica/patologia , Morte Celular/fisiologia , Feminino , Glucose/deficiência , Hipóxia Encefálica/patologia , Hipóxia Encefálica/fisiopatologia , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Masculino , Glicoproteínas de Membrana/metabolismo , Camundongos Transgênicos , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/metabolismo , Neurônios/patologia , Espécies Reativas de Oxigênio/metabolismo , Acidente Vascular Cerebral/etiologia , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia , Técnicas de Cultura de Tecidos
12.
Neuropharmacology ; 79: 550-8, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24440367

RESUMO

Angiotensin (Ang) II exaggerates cerebral injury in ischemic damage. Angiotensin-converting enzyme type 2 (ACE2) converts Ang II into Ang (1-7) and thus, may protect against the effects of Ang II. We hypothesized that neuronal ACE2 over-expression decreases ischemic stroke in mice with Ang II overproduction. Human renin and angiotensinogen double transgenic (RA) mice and RA mice with neuronal over-expression of ACE2 (SARA) were used for the study. The mean arterial pressure (MAP) was calculated from telemetry-recorded blood pressure (BP). SARA mice were infused peripherally with Norepinephrine to "clamp" the BP, or intracerebroventricularly-infused with a Mas receptor antagonist (A-779). Middle cerebral artery occlusion (MCAO) surgery was performed to induce permanent focal ischemic stroke. Cerebral blood flow (CBF) and neurological function were determined. Two days after surgery, brain samples were collected for various analyses. Results showed: 1) When compared to chronically hypertensive RA mice, SARA mice had lower basal MAP, less MCAO-induced infarct volume, and increased CBF, neurological function and cerebral microvascular density in the peri-infarct area; 2) These changes in SARA mice were not altered after MAP "clamping", but partially reversed by brain infusion of A-779; 3) Ang (1-7)/Ang II ratio, angiogenic factors, endothelial nitric oxide synthase (eNOS) expression and nitric oxide production were increased, whereas, NADPH oxidase subunits and reactive oxygen species were decreased in the brain of SARA mice. ACE2 protects brain from ischemic injury via the regulation of NADPH oxidase/eNOS pathways by changing Ang (1-7)/Ang II ratio, independently of MAP changes.


Assuntos
Isquemia Encefálica/fisiopatologia , Encéfalo/fisiopatologia , Infarto da Artéria Cerebral Média/fisiopatologia , Neurônios/fisiologia , Peptidil Dipeptidase A/metabolismo , Angiotensina II/análogos & derivados , Angiotensina II/farmacologia , Enzima de Conversão de Angiotensina 2 , Angiotensinogênio/genética , Angiotensinogênio/metabolismo , Animais , Pressão Arterial/efeitos dos fármacos , Pressão Arterial/fisiologia , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/patologia , Circulação Cerebrovascular/efeitos dos fármacos , Circulação Cerebrovascular/fisiologia , Humanos , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/patologia , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/farmacologia , Peptidil Dipeptidase A/genética , Renina/genética , Renina/metabolismo
13.
Biochim Biophys Acta ; 1829(11): 1225-35, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24100303

RESUMO

Pancreatic angiotensin-converting enzyme 2 (ACE2) has previously been shown to be critical for maintaining glycemia and ß-cell function. Efforts to maintain or increase ACE2 expression in pancreatic ß-cells might therefore have therapeutic potential for treating diabetes. In our study, we investigated the transcriptional role of hepatocyte nuclear factor 1α (HNF1α) and hepatocyte nuclear factor 1ß (HNF1ß) in induction of ACE2 expression in insulin-secreting cells. A deficient allele of HNF1α or HNF1ß causes maturity-onset diabetes of the young (MODY) types 3 and 5, respectively, in humans. We found that ACE2 is primarily transcribed from the proximal part of the ACE2 promoter in the pancreas. In the proximal part of the human ACE2 promoter, we further identified three functional HNF1 binding sites, as they have binding affinity for HNF1α and HNF1ß and are required for induction of promoter activity by HNF1ß in insulinoma cells. These three sites are well-conserved among mammalian species. Both HNF1α and HNF1ß induce expression of ACE2 mRNA and lead to elevated levels of ACE2 protein and ACE2 enzymatic activity in insulinoma cells. Furthermore, HNF1α dose-dependently increases ACE2 expression in primary pancreatic islet cells. We conclude that HNF1α can induce the expression of ACE2 in pancreatic islet cells via evolutionarily conserved HNF1 binding sites in the ACE2 promoter. Potential therapeutics aimed at counteracting functional HNF1α depletion in diabetes and MODY3 will thus have ACE2 induction in pancreatic islets as a likely beneficial effect.


Assuntos
Evolução Molecular , Fator 1-alfa Nuclear de Hepatócito/fisiologia , Ilhotas Pancreáticas/enzimologia , Peptidil Dipeptidase A/metabolismo , Enzima de Conversão de Angiotensina 2 , Animais , Sequência de Bases , Linhagem Celular Tumoral , Primers do DNA , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Peptidil Dipeptidase A/genética , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Circ Res ; 113(9): 1087-1096, 2013 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-24014829

RESUMO

RATIONALE: Overactivity of the brain renin-angiotensin system is a major contributor to neurogenic hypertension. Although overexpression of angiotensin-converting enzyme type 2 (ACE2) has been shown to be beneficial in reducing hypertension by transforming angiotensin II into angiotensin-(1-7), several groups have reported decreased brain ACE2 expression and activity during the development of hypertension. OBJECTIVE: We hypothesized that ADAM17-mediated ACE2 shedding results in decreased membrane-bound ACE2 in the brain, thus promoting the development of neurogenic hypertension. METHODS AND RESULTS: To test this hypothesis, we used the deoxycorticosterone acetate-salt model of neurogenic hypertension in nontransgenic and syn-hACE2 mice overexpressing ACE2 in neurons. Deoxycorticosterone acetate-salt treatment in nontransgenic mice led to significant increases in blood pressure, hypothalamic angiotensin II levels, inflammation, impaired baroreflex sensitivity, and autonomic dysfunction, as well as decreased hypothalamic ACE2 activity and expression, although these changes were blunted or prevented in syn-hACE2 mice. In addition, reduction of ACE2 expression and activity in the brain paralleled an increase in ACE2 activity in the cerebrospinal fluid of nontransgenic mice after deoxycorticosterone acetate-salt treatment and were accompanied by enhanced ADAM17 expression and activity in the hypothalamus. Chronic knockdown of ADAM17 in the brain blunted the development of hypertension and restored ACE2 activity and baroreflex function. CONCLUSIONS: Our data provide the first evidence that ADAM17-mediated shedding impairs brain ACE2 compensatory activity, thus contributing to the development of neurogenic hypertension.


Assuntos
Pressão Sanguínea , Encéfalo/enzimologia , Hipertensão/enzimologia , Peptidil Dipeptidase A/metabolismo , Sistema Renina-Angiotensina , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM17 , Angiotensina II/metabolismo , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Enzima de Conversão de Angiotensina 2 , Animais , Anti-Hipertensivos/farmacologia , Sistema Nervoso Autônomo/metabolismo , Sistema Nervoso Autônomo/fisiopatologia , Barorreflexo , Pressão Sanguínea/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Acetato de Desoxicorticosterona , Modelos Animais de Doenças , Humanos , Hipertensão/induzido quimicamente , Hipertensão/tratamento farmacológico , Hipertensão/genética , Hipertensão/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Peptidil Dipeptidase A/genética , Interferência de RNA , Sistema Renina-Angiotensina/efeitos dos fármacos , Fatores de Tempo
15.
Am J Physiol Endocrinol Metab ; 304(8): E874-84, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23462816

RESUMO

An overactive renin-angiotensin system (RAS) is known to contribute to type 2 diabetes mellitus (T2DM). Although ACE2 overexpression has been shown to be protective against the overactive RAS, a role for pancreatic ACE2, particularly in the islets of Langerhans, in regulating glycemia in response to elevated angiotensin II (Ang II) levels remains to be elucidated. This study examined the role of endogenous pancreatic ACE2 and the impact of elevated Ang II levels on the enzyme's ability to alleviate hyperglycemia in an Ang II infusion mouse model. Male C57bl/6J mice were infused with Ang II or saline for a period of 14 days. On the 7th day of infusion, either an adenovirus encoding human ACE2 (Ad-hACE2) or a control adenovirus (Ad-eGFP) was injected into the mouse pancreas. After an additional 7-8 days, glycemia and plasma insulin levels as well as RAS components expression and oxidative stress were assessed. Ang II-infused mice exhibited hyperglycemia, hyperinsulinemia, and impaired glucose-stimulated insulin secretion from pancreatic islets compared with control mice. This phenotype was associated with decreased ACE2 expression and activity, increased Ang II type 1 receptor (AT1R) expression, and increased oxidative stress in the mouse pancreas. Ad-hACE2 treatment restored pancreatic ACE2 expression and compensatory activity against Ang II-mediated impaired glycemia, thus improving ß-cell function. Our data suggest that decreased pancreatic ACE2 is a link between overactive RAS and impaired glycemia in T2DM. Moreover, maintenance of a normal endogenous ACE2 compensatory activity in the pancreas appears critical to avoid ß-cell dysfunction, supporting a therapeutic potential for ACE2 in controlling diabetes resulting from an overactive RAS.


Assuntos
Angiotensina II/farmacologia , Diabetes Mellitus Tipo 2/terapia , Terapia Genética/métodos , Hiperglicemia/terapia , Células Secretoras de Insulina/fisiologia , Peptidil Dipeptidase A/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adenoviridae/genética , Enzima de Conversão de Angiotensina 2 , Animais , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Peptídeo C/sangue , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Hiperglicemia/metabolismo , Insulina/sangue , Resistência à Insulina/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peptidil Dipeptidase A/metabolismo , Peptidil Dipeptidase A/farmacologia , Sistema Renina-Angiotensina/efeitos dos fármacos , Sistema Renina-Angiotensina/fisiologia , Vasoconstritores/farmacologia
16.
Am J Physiol Regul Integr Comp Physiol ; 301(5): R1293-9, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21880865

RESUMO

Angiotensin-converting enzyme 2 (ACE2) is a component of the renin-angiotensin system, and its expression and activity have been shown to be reduced in cardiovascular diseases. Enzymatic activity of ACE2 is commonly measured by hydrolysis of quenched fluorescent substrates in the absence or presence of an ACE2-specific inhibitor, such as the commercially available inhibitor DX600. Whereas recombinant human ACE2 is readily detected in mouse tissues using 1 µM DX600 at pH 7.5, the endogenous ACE2 activity in mouse tissues is barely detectable. We compared human, mouse, and rat ACE2 overexpressed in cell lines for their sensitivity to inhibition by DX600. ACE2 from all three species could be inhibited by DX600, but the half maximal inhibitory concentration (IC(50)) for human ACE2 was much lower (78-fold) than for rodent ACE2. Following optimization of pH, substrate concentration, and antagonist concentration, rat and mouse ACE2 expressed in a cell line could be accurately quantified with 10 µM DX600 (>95% inhibition) but not with 1 µM DX600 (<75% inhibition). Validation that the optimized method robustly quantifies ACE2 in mouse tissues (kidney, brain, heart, and plasma) was performed using wild-type and ACE2 knockout mice. This study provides a reliable method for measuring human, as well as endogenous ACE2 activity in rodents. Our data underscore the importance of validating the effect of DX600 on ACE2 from each particular species at the experimental conditions employed.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Peptídeos/farmacologia , Peptidil Dipeptidase A/metabolismo , Enzima de Conversão de Angiotensina 2 , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Humanos , Concentração de Íons de Hidrogênio , Hidrólise , Rim/efeitos dos fármacos , Rim/enzimologia , Cinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/enzimologia , Peptidil Dipeptidase A/deficiência , Peptidil Dipeptidase A/genética , Ratos , Reprodutibilidade dos Testes , Especificidade da Espécie , Espectrometria de Fluorescência , Especificidade por Substrato , Transfecção
17.
PLoS One ; 6(7): e22682, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21818366

RESUMO

Oxidative stress in the central nervous system mediates the increase in sympathetic tone that precedes the development of hypertension. We hypothesized that by transforming Angiotensin-II (AngII) into Ang-(1-7), ACE2 might reduce AngII-mediated oxidative stress in the brain and prevent autonomic dysfunction. To test this hypothesis, a relationship between ACE2 and oxidative stress was first confirmed in a mouse neuroblastoma cell line (Neuro2A cells) treated with AngII and infected with Ad-hACE2. ACE2 overexpression resulted in a reduction of reactive oxygen species (ROS) formation. In vivo, ACE2 knockout (ACE2(-/y)) mice and non-transgenic (NT) littermates were infused with AngII (10 days) and infected with Ad-hACE2 in the paraventricular nucleus (PVN). Baseline blood pressure (BP), AngII and brain ROS levels were not different between young mice (12 weeks). However, cardiac sympathetic tone, brain NADPH oxidase and SOD activities were significantly increased in ACE2(-/y). Post infusion, plasma and brain AngII levels were also significantly higher in ACE2(-/y), although BP was similarly increased in both genotypes. ROS formation in the PVN and RVLM was significantly higher in ACE2(-/y) mice following AngII infusion. Similar phenotypes, i.e. increased oxidative stress, exacerbated dysautonomia and hypertension, were also observed on baseline in mature ACE2(-/y) mice (48 weeks). ACE2 gene therapy to the PVN reduced AngII-mediated increase in NADPH oxidase activity and normalized cardiac dysautonomia in ACE2(-/y) mice. Altogether, these data indicate that ACE2 gene deletion promotes age-dependent oxidative stress, autonomic dysfunction and hypertension, while PVN-targeted ACE2 gene therapy decreases ROS formation via NADPH oxidase inhibition and improves autonomic function. Accordingly, ACE2 could represent a new target for the treatment of hypertension-associated dysautonomia and oxidative stress.


Assuntos
Sistema Nervoso Autônomo/fisiopatologia , Sistema Nervoso Central/fisiopatologia , Estresse Oxidativo , Peptidil Dipeptidase A/metabolismo , Envelhecimento/patologia , Angiotensina II/farmacologia , Enzima de Conversão de Angiotensina 2 , Animais , Sistema Nervoso Autônomo/efeitos dos fármacos , Sistema Nervoso Autônomo/patologia , Pressão Sanguínea/efeitos dos fármacos , Encéfalo/enzimologia , Encéfalo/patologia , Encéfalo/fisiopatologia , Linhagem Celular Tumoral , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/patologia , Deleção de Genes , Terapia Genética , Masculino , Camundongos , Miocárdio/patologia , NADPH Oxidases/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Disautonomias Primárias/patologia , Disautonomias Primárias/fisiopatologia , Espécies Reativas de Oxigênio/metabolismo , Sistema Nervoso Simpático/efeitos dos fármacos , Sistema Nervoso Simpático/patologia , Sistema Nervoso Simpático/fisiopatologia , Nervo Vago/efeitos dos fármacos , Nervo Vago/patologia , Nervo Vago/fisiopatologia
18.
Diabetes ; 59(10): 2540-8, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20660625

RESUMO

OBJECTIVE: Several clinical studies have shown the benefits of renin-angiotensin system (RAS) blockade in the development of diabetes, and a local RAS has been identified in pancreatic islets. Angiotensin I-converting enzyme (ACE)2, a new component of the RAS, has been identified in the pancreas, but its role in ß-cell function remains unknown. Using 8- and 16-week-old obese db/db mice, we examined the ability of ACE2 to alter pancreatic ß-cell function and thereby modulate hyperglycemia. RESEARCH DESIGN AND METHODS: Both db/db and nondiabetic lean control (db/m) mice were infected with an adenovirus expressing human ACE2 (Ad-hACE2-eGFP) or the control virus (Ad-eGFP) via injection into the pancreas. Glycemia and ß-cell function were assessed 1 week later at the peak of viral expression. RESULTS: In 8-week-old db/db mice, Ad-hACE2-eGFP significantly improved fasting glycemia, enhanced intraperitoneal glucose tolerance, increased islet insulin content and ß-cell proliferation, and reduced ß-cell apoptosis compared with Ad-eGFP. ACE2 overexpression had no effect on insulin sensitivity in comparison with Ad-eGFP treatment in diabetic mice. Angiotensin-(1-7) receptor blockade by D-Ala(7)-Ang-(1-7) prevented the ACE2-mediated improvements in intraperitoneal glucose tolerance, glycemia, and islet function and also impaired insulin sensitivity in both Ad-hACE2-eGFP- and Ad-eGFP-treated db/db mice. D-Ala(7)-Ang-(1-7) had no effect on db/m mice. In 16-week-old diabetic mice, Ad-hACE2-eGFP treatment improved fasting blood glucose but had no effect on any of the other parameters. CONCLUSIONS: These findings identify ACE2 as a novel target for the prevention of ß-cell dysfunction and apoptosis occurring in type 2 diabetes.


Assuntos
Glicemia/metabolismo , Diabetes Mellitus Experimental/sangue , Terapia Genética/métodos , Peptidil Dipeptidase A/genética , Enzima de Conversão de Angiotensina 2 , Animais , Apoptose , Divisão Celular , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/terapia , Regulação Enzimológica da Expressão Gênica , Genes Reporter , Teste de Tolerância a Glucose , Proteínas de Fluorescência Verde/genética , Homeostase , Humanos , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/fisiopatologia , Camundongos , Camundongos Transgênicos , Obesidade/complicações , Pâncreas/enzimologia , Pâncreas/patologia , Pâncreas/fisiopatologia , Peptidil Dipeptidase A/metabolismo
19.
Circ Res ; 102(6): 729-36, 2008 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-18258853

RESUMO

We recently reported the presence of angiotensin-converting enzyme (ACE)2 in brain regions controlling cardiovascular function; however, the role of ACE2 in blood pressure regulation remains unclear because of the lack of specific tools to investigate its function. We hypothesized that ACE2 could play a pivotal role in the central regulation of cardiovascular function by regulating other renin-angiotensin system components. To test this hypothesis, we generated an adenovirus expressing the human ACE2 cDNA upstream of an enhanced green fluorescent protein (eGFP) reporter gene (Ad-hACE2-eGFP). In vitro characterization shows that neuronal cells infected with Ad-hACE2-eGFP (10 to 100 multiplicities of infection), but not Ad-eGFP (100 multiplicities of infection), exhibit dose-dependent ACE2 expression and activity. In addition, an active secreted form was detected in the conditioned medium. In vivo, Ad-hACE2-eGFP infection (2x10(6) plaque-forming units intracerebroventricularly) produced time-dependent expression and activity (with a peak at 7 days) in the mouse subfornical organ. More importantly, 7 days after virus infection, the pressor response to angiotensin (Ang) II (200 pmol intracerebroventricularly) was significantly reduced in Ad-hACE2-eGFP-treated mice compared with controls. Furthermore, subfornical organ-targeted ACE2 overexpression dramatically reduced the Ang II-mediated drinking response. Interestingly, ACE2 overexpression was associated with downregulation of the Ang II type 1 receptor expression both in vitro and in vivo. These data suggest that ACE2 overexpression in the subfornical organ impairs Ang II-mediated pressor and drinking responses at least by inhibiting the Ang II type 1 receptor expression. Taken together, our results show that ACE2 plays a pivotal role in the central regulation of blood pressure and volume homeostasis, offering a new target for the treatment of hypertension and other cardiovascular diseases.


Assuntos
Angiotensina II/metabolismo , Barorreflexo , Comportamento de Ingestão de Líquido , Neurônios/metabolismo , Peptidil Dipeptidase A/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Órgão Subfornical/metabolismo , Adenoviridae/efeitos dos fármacos , Angiotensina II/administração & dosagem , Enzima de Conversão de Angiotensina 2 , Animais , Barorreflexo/efeitos dos fármacos , Pressão Sanguínea , Linhagem Celular Tumoral , Meios de Cultura/metabolismo , Regulação para Baixo , Comportamento de Ingestão de Líquido/efeitos dos fármacos , Genes Reporter , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Frequência Cardíaca , Humanos , Injeções Intraventriculares , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/enzimologia , Peptidil Dipeptidase A/genética , Receptor Tipo 1 de Angiotensina/agonistas , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Órgão Subfornical/efeitos dos fármacos , Órgão Subfornical/enzimologia , Fatores de Tempo , Transdução Genética , Regulação para Cima
20.
Life Sci ; 79(19): 1812-9, 2006 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-16822527

RESUMO

The exaggerated cardiac arrhythmias in cardiomyopathy induced by L-thyroxine treatment are related to ion channelopathies and to an abnormal endothelin (ET) pathway. It was hypothesized that an increased incidence of ventricular fibrillation (VF) could be mediated by inflammatory factors including the ET pathway, nuclear factor kappa B (NFkappaB), tumor necrosis factor-alpha (TNFalpha) and inducible nitric oxide synthase (iNOS). Abnormal expression of NFkappaB, TNFalpha, iNOS and enhanced VF are linked with the activated ET pathway and a significant reversion could be achieved by the selective endothelin A receptor antagonist darusentan. Cardiomyopathy in rats was produced by L-thyroxine treatment (0.3 mg kg(-1) d(-1), sc) for 10 days. The mRNA expression of the ET pathway, NFkappaB, TNFalpha, iNOS and the activity of the redox system were assayed in association with the incidence of VF produced by coronary ligation/reperfusion. Darusentan was administered on days 6-10 of L-thyroxine treatment. The VF incidence, which was higher in the l-thyroxine cardiomyopathy group, was suppressed by darusentan. The mRNA levels of preproET-1, endothelin converting enzyme, endothelin receptor A (ET(A)R), endothelin receptor B (ET(B)R), NFkappaB, TNFalpha and iNOS in left ventricle were up-regulated in the cardiomyopathic heart. There was significant oxidative stress in this cardiomyopathy model. Darusentan suppressed the up-regulated mRNA levels of ET(A)R, ET(B)R, NFkappaB, TNFalpha, and iNOS. These results indicate that the high incidence of VF which is related to up-regulation of inflammatory factors in the cardiomyopathic myocardium is significantly suppressed by selective ET(A)R blockade.


Assuntos
Cardiomiopatias/tratamento farmacológico , Antagonistas do Receptor de Endotelina A , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , NF-kappa B/biossíntese , Óxido Nítrico Sintase Tipo II/biossíntese , Fenilpropionatos/uso terapêutico , Pirimidinas/uso terapêutico , Fibrilação Ventricular/prevenção & controle , Animais , Cardiomiopatias/complicações , Cardiomiopatias/metabolismo , Modelos Animais de Doenças , Endotelinas/biossíntese , Masculino , Oxirredução , Ratos , Ratos Sprague-Dawley , Tiroxina , Fator de Necrose Tumoral alfa/biossíntese , Regulação para Cima , Fibrilação Ventricular/etiologia , Fibrilação Ventricular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA