Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Heart Circ Physiol ; 318(1): H34-H48, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31675258

RESUMO

Obstructive sleep apnea is characterized by interrupted breathing that leads to cardiovascular sequelae including chronic hypertension that can persist into the waking hours. Chronic intermittent hypoxia (CIH), which models the hypoxemia associated with sleep apnea, is sufficient to cause a sustained increase in blood pressure that involves the central nervous system. The median preoptic nucleus (MnPO) is an integrative forebrain region that contributes to blood pressure regulation and neurogenic hypertension. The MnPO projects to the paraventricular nucleus (PVN), a preautonomic region. We hypothesized that pathway-specific lesions of the projection from the MnPO to the PVN would attenuate the sustained component of chronic intermittent hypoxia-induced hypertension. Adult male Sprague-Dawley rats (250-300 g) were anesthetized with isoflurane and stereotaxically injected bilaterally in the PVN with a retrograde Cre-containing adeno-associated virus (AAV; AAV9.CMV.HI.eGFP-Cre.WPRE.SV40) and injected in the MnPO with caspase-3 (AAV5-flex-taCasp3-TEVp) or control virus (AAV5-hSyn-DIO-mCherry). Three weeks after the injections the rats were exposed to a 7-day intermittent hypoxia protocol. During chronic intermittent hypoxia, controls developed a diurnal hypertension that was blunted in rats with caspase lesions. Brain tissue processed for FosB immunohistochemistry showed decreased staining with caspase-induced lesions of MnPO and downstream autonomic-regulating nuclei. Chronic intermittent hypoxia significantly increased plasma levels of advanced oxidative protein products in controls, but this increase was blocked in caspase-lesioned rats. The results indicate that PVN-projecting MnPO neurons play a significant role in blood pressure regulation in the development of persistent chronic intermittent hypoxia hypertension.NEW & NOTEWORTHY Chronic intermittent hypoxia associated with obstructive sleep apnea increases oxidative stress and leads to chronic hypertension. Sustained hypertension may be mediated by angiotensin II-induced neural plasticity of excitatory median preoptic neurons in the forebrain that project to the paraventricular nucleus of the hypothalamus. Selective caspase lesions of these neurons interrupt the drive for sustained hypertension and cause a reduction in circulating oxidative protein products. This indicates that a functional connection between the forebrain and hypothalamus is necessary to drive diurnal hypertension associated with intermittent hypoxia. These results provide new information about central mechanisms that may contribute to neurogenic hypertension.


Assuntos
Apoptose , Pressão Arterial , Caspase 3/metabolismo , Hipertensão/prevenção & controle , Hipóxia/complicações , Núcleo Hipotalâmico Paraventricular/enzimologia , Área Pré-Óptica/enzimologia , Animais , Caspase 3/genética , Ritmo Circadiano , Modelos Animais de Doenças , Frequência Cardíaca , Hipertensão/enzimologia , Hipertensão/patologia , Hipertensão/fisiopatologia , Hipóxia/enzimologia , Hipóxia/patologia , Hipóxia/fisiopatologia , Masculino , Estresse Oxidativo , Núcleo Hipotalâmico Paraventricular/patologia , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Área Pré-Óptica/patologia , Área Pré-Óptica/fisiopatologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos Sprague-Dawley , Transdução de Sinais
2.
Am J Physiol Heart Circ Physiol ; 316(6): H1389-H1405, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30925093

RESUMO

Activation of the brain renin-angiotensin system (RAS) is a pivotal step in the pathogenesis of hypertension. The paraventricular nucleus (PVN) of the hypothalamus is a critical part of the angiotensinergic sympatho-excitatory neuronal network involved in neural control of blood pressure and hypertension. However, the importance of the PVN (pro)renin receptor (PVN-PRR)-a key component of the brain RAS-in hypertension development has not been examined. In this study, we investigated the involvement and mechanisms of the PVN-PRR in DOCA-salt-induced hypertension, a mouse model of hypertension. Using nanoinjection of adeno-associated virus-mediated Cre recombinase expression to knock down the PRR specifically in the PVN, we report here that PVN-PRR knockdown attenuated the enhanced blood pressure and sympathetic tone associated with hypertension. Mechanistically, we found that PVN-PRR knockdown was associated with reduced activation of ERK (extracellular signal-regulated kinase)-1/2 in the PVN and rostral ventrolateral medulla during hypertension. In addition, using the genetically encoded Ca2+ biosensor GCaMP6 to monitor Ca2+-signaling events in the neurons of PVN brain slices, we identified a reduction in angiotensin II type 1 receptor-mediated Ca2+ activity as part of the mechanism by which PVN-PRR knockdown attenuates hypertension. Our study demonstrates an essential role of the PRR in PVN neurons in hypertension through regulation of ERK1/2 activation and angiotensin II type 1 receptor-mediated Ca2+ activity. NEW & NOTEWORTHY PRR knockdown in PVN neurons attenuates the development of DOCA-salt hypertension and autonomic dysfunction through a decrease in ERK1/2 activation in the PVN and RVLM during hypertension. In addition, PRR knockdown reduced AT1aR expression and AT1R-mediated calcium activity during hypertension. Furthermore, we characterized the neuronal targeting specificity of AAV serotype 2 in the mouse PVN and validated the advantages of the genetically encoded calcium biosensor GCaMP6 in visualizing neuronal calcium activity in the PVN.


Assuntos
Pressão Sanguínea , Sinalização do Cálcio , Hipertensão/prevenção & controle , Neurônios/enzimologia , Núcleo Hipotalâmico Paraventricular/enzimologia , ATPases Translocadoras de Prótons/deficiência , Receptor Tipo 1 de Angiotensina/metabolismo , Receptores de Superfície Celular/deficiência , Animais , Sistema Nervoso Autônomo/metabolismo , Sistema Nervoso Autônomo/fisiopatologia , Receptor 1 de Quimiocina CX3C/genética , Receptor 1 de Quimiocina CX3C/metabolismo , Acetato de Desoxicorticosterona , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Hipertensão/enzimologia , Hipertensão/genética , Hipertensão/fisiopatologia , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Fosforilação , ATPases Translocadoras de Prótons/genética , Receptor Tipo 1 de Angiotensina/genética , Receptores de Superfície Celular/genética , Receptor de Pró-Renina
3.
J Cardiol ; 73(1): 81-88, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30487059

RESUMO

BACKGROUND: The hypothalamic paraventricular nucleus (PVN) is the center of the regulation of autonomic nervous system functions and cardiovascular activity. Phosphoinositide-3 kinase (PI3K)-AKT pathway in PVN contributes to mediate sympathetic nerve activity and is activated in spontaneously hypertensive rats. Overactivation of the sympathetic output was considered as an important mechanism of the arrhythmias. In the present study, we aimed to explore whether targeted regulation of sympathetic activity in PVN could reduce the peripheral sympathoexcitatory and attenuate the ventricular arrhythmias (VAs) in myocardial infarction (MI) rats via PI3K-AKT pathway. METHODS: A stainless steel gauge guide cannula was stereotaxically implanted into the PVN, and 7 days later, rats were randomly divided into the following 4 groups: group A, control+dimethyl sulfoxide (DMSO); group B, control+LY294002; group C, MI surgery+DMSO; and group D, MI surgery+LY294002. Studies were conducted seven days post-MI. Myocardial function, infarct size, inducible VAs by programmed electrical stimulation, renal sympathetic nerve activity (RSNA), and protein level of PI3K and AKT were measured. RESULTS: MI increased the protein ratios of p-PI3K-to-total-PI3K and p-AKT-to-total-AKT in PVN but can be reduced by LY294002 treatment. Inhibition of sympathetic nerve activity in PVN led to a reversion in plasma norepinephrine, RSNA and inducible VAs in MI rats. CONCLUSIONS: PI3K-AKT pathway in the PVN was a main mechanism in regulating sympathetic activity and arrhythmias in MI rats. Targeted inhibition of sympathetic activity in PVN may be a potential treatment for the VAs via PI3K-AKT pathway.


Assuntos
Arritmias Cardíacas/enzimologia , Infarto do Miocárdio/enzimologia , Núcleo Hipotalâmico Paraventricular/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Sistema Nervoso Simpático/enzimologia , Animais , Arritmias Cardíacas/fisiopatologia , Arritmias Cardíacas/prevenção & controle , Cromonas/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Masculino , Morfolinas/uso terapêutico , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/fisiopatologia , Miocárdio/enzimologia , Norepinefrina/sangue , Ratos , Ratos Endogâmicos SHR , Transdução de Sinais/efeitos dos fármacos
4.
Hypertension ; 72(3): 667-675, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30012866

RESUMO

Renal denervation (RDN) has been shown to restore endogenous neuronal nitric oxide synthase (nNOS) in the paraventricular nucleus (PVN) and reduce sympathetic drive during chronic heart failure (CHF). The purpose of the present study was to assess the contribution of afferent renal nerves to the nNOS-mediated sympathetic outflow within the PVN in rats with CHF. CHF was induced in rats by ligation of the left coronary artery. Four weeks after surgery, selective afferent RDN (A-RDN) was performed by bilateral perivascular application of capsaicin on the renal arteries. Seven days after intervention, nNOS protein expression, nNOS immunostaining signaling, and diaphorase-positive stained cells were significantly decreased in the PVN of CHF rats, changes that were reversed by A-RDN. A-RDN reduced basal lumbar sympathetic nerve activity in rats with CHF (8.5%±0.5% versus 17.0%±1.2% of max). Microinjection of nNOS inhibitor L-NMMA (L-NG-monomethyl arginine citrate) into the PVN produced a blunted increase in lumbar sympathetic nerve activity in rats with CHF. This response was significantly improved after A-RDN (Δ lumbar sympathetic nerve activity: 25.7%±2.4% versus 11.2%±0.9%). Resting afferent renal nerves activity was substantially increased in CHF compared with sham rats (56.3%±2.4% versus 33.0%±4.7%). These results suggest that intact afferent renal nerves contribute to the reduction of nNOS in the PVN. A-RDN restores nNOS and thus attenuates the sympathoexcitation. Also, resting afferent renal nerves activity is elevated in CHF rats, which may highlight a crucial neural mechanism arising from the kidney in the maintenance of enhanced sympathetic drive in CHF.


Assuntos
Denervação/métodos , Insuficiência Cardíaca/fisiopatologia , Rim/inervação , Óxido Nítrico Sintase Tipo I/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Sistema Nervoso Simpático/fisiopatologia , Vias Aferentes/efeitos dos fármacos , Vias Aferentes/fisiologia , Animais , Capsaicina/farmacologia , Doença Crônica , Inibidores Enzimáticos/farmacologia , Masculino , Óxido Nítrico Sintase Tipo I/antagonistas & inibidores , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/enzimologia , Ratos Sprague-Dawley , Fármacos do Sistema Sensorial/farmacologia , Sistema Nervoso Simpático/efeitos dos fármacos , ômega-N-Metilarginina/farmacologia
5.
Science ; 351(6279): 1293-6, 2016 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-26989246

RESUMO

Maintaining energy homeostasis is crucial for the survival and health of organisms. The brain regulates feeding by responding to dietary factors and metabolic signals from peripheral organs. It is unclear how the brain interprets these signals. O-GlcNAc transferase (OGT) catalyzes the posttranslational modification of proteins by O-GlcNAc and is regulated by nutrient access. Here, we show that acute deletion of OGT from αCaMKII-positive neurons in adult mice caused obesity from overeating. The hyperphagia derived from the paraventricular nucleus (PVN) of the hypothalamus, where loss of OGT was associated with impaired satiety. These results identify O-GlcNAcylation in αCaMKII neurons of the PVN as an important molecular mechanism that regulates feeding behavior.


Assuntos
Metabolismo Energético/fisiologia , Comportamento Alimentar/fisiologia , Hiperfagia/genética , N-Acetilglucosaminiltransferases/fisiologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Acetilglucosamina/metabolismo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Metabolismo Energético/genética , Deleção de Genes , Homeostase/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , N-Acetilglucosaminiltransferases/genética , Neurônios/enzimologia , Obesidade/genética , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/enzimologia , Processamento de Proteína Pós-Traducional , Resposta de Saciedade/fisiologia
6.
Cardiovasc Toxicol ; 16(4): 345-54, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26438340

RESUMO

High-salt-induced inflammation and oxidative stress in the hypothalamic paraventricular nucleus (PVN) contribute to the pathogenesis of salt-sensitive hypertension. In this study, we hypothesized that chronic inhibition of nuclear factor-κB (NF-κB) activity in the PVN delays the progression of hypertension by upregulating anti-inflammatory cytokines, reducing NLRP3 (NOD-like receptor family pyrin domain containing 3) and IL-1ß and attenuating p-IKKß, NF-κB p65 activity and NAD(P)H oxidase in the PVN of salt-sensitive hypertensive rats. Dahl salt-sensitive rats received a high-salt diet (HS, 8 % NaCl) or a normal-salt diet (NS, 0.3 % NaCl) for 6 weeks and were treated with bilateral PVN infusion with either vehicle or pyrrolidine dithiocarbamate (PDTC, 5 µg/h), a NF-κB inhibitor via osmotic minipump. The mean arterial pressure and plasma levels of norepinephrine (NE) and epinephrine (EPI) were significantly increased in high-salt-fed rats. In addition, rats with high-salt diet had higher levels of p-IKKß, NF-κB p65 activity, Fra-like (Fra-LI) activity (an indicator of chronic neuronal activation), NOX-4 (subunits of NAD(P)H oxidase), NLRP3 and IL-1ß, and lower levels of IL-10 in the PVN than normal diet rats. Bilateral PVN infusions of PDTC attenuated these high-salt-induced changes. These findings suggest that high-salt-induced NF-κB activation in the PVN caused hypertension via sympathoexcitation, which are associated with the increases of NLRP3, IL-1ß and oxidative stress in the PVN; PVN inhibition of NF-κB activity attenuates NLRP3, IL-1ß and oxidative stress in the PVN and thereby attenuates hypertension.


Assuntos
Anti-Hipertensivos/administração & dosagem , Pressão Arterial/efeitos dos fármacos , Caspase 1/metabolismo , Hipertensão/prevenção & controle , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Pirrolidinas/administração & dosagem , Cloreto de Sódio na Dieta , Tiocarbamatos/administração & dosagem , Fator de Transcrição RelA/antagonistas & inibidores , Animais , Anti-Inflamatórios/administração & dosagem , Antioxidantes/administração & dosagem , Biomarcadores/sangue , Modelos Animais de Doenças , Epinefrina/sangue , Hipertensão/enzimologia , Hipertensão/fisiopatologia , Quinase I-kappa B/metabolismo , Infusões Parenterais , Interleucina-1beta/metabolismo , Masculino , Norepinefrina/sangue , Estresse Oxidativo/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/enzimologia , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Fosforilação , Ratos Endogâmicos Dahl , Transdução de Sinais , Sistema Nervoso Simpático/efeitos dos fármacos , Sistema Nervoso Simpático/metabolismo , Sistema Nervoso Simpático/fisiopatologia , Fator de Transcrição RelA/metabolismo
7.
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
8.
Physiol Behav ; 133: 107-14, 2014 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-24874775

RESUMO

Genistein is a phytoestrogen, particularly abundant in soybeans, that is able to bind estrogen receptors exerting both estrogenic and antiestrogenic activities. Genistein is largely present in the human diet even during pregnancy. Embryos and fetuses are therefore, commonly exposed to genistein during the development and after birth. In the present study, we used a murine model as a test end-point to investigate the effects of early exposure to genistein on adult male behavior and related neural circuits. Daily exposure of dams to genistein (100 µg/g of body weight) during late pregnancy and early lactation, produced in male offspring, when adults, significant changes in anxiety and aggressive behaviors. Moreover, we found statistically significant variations in the number of neuronal nitric-oxide synthase positive cells in the amygdala. In conclusions, these data indicate that early exposure to phytoestrogens may induce life-long effects on the differentiation of brain structures and behaviors.


Assuntos
Agressão/efeitos dos fármacos , Ansiedade/induzido quimicamente , Genisteína/administração & dosagem , Óxido Nítrico Sintase/metabolismo , Fitoestrógenos/administração & dosagem , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Fatores Etários , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/enzimologia , Animais , Peso Corporal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Comportamento Exploratório/efeitos dos fármacos , Feminino , Lactação/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/enzimologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Comportamento Sexual Animal/efeitos dos fármacos
9.
Acta Histochem ; 116(1): 182-90, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23953641

RESUMO

Heroin is one of the most dangerous drugs of abuse, which may exert various neurotoxic actions on the brain (such as gray matter loss, neuronal apoptosis, mitochondrial dysfunction, synaptic defects, depression of adult neurogenensis, as well as development of spongiform leucoencephalopathy). Some of these toxic effects are probably mediated by the gas nitric oxide (NO). We studied by morphometric analysis the numerical density of neurons expressing neuronal nitric oxide synthase (nNOS) in cortical and hypothalamic areas of eight heroin overdose victims and nine matched controls. Heroin addicts showed significantly increased numerical densities of nNOS immunoreactive cells in the right temporal cortex and the left paraventricular nucleus. Remarkably, in heroin abusers, but not in controls, we observed not only immunostained interneurons, but also cortical pyramidal cells. Given that increased cellular expression of nNOS was accompanied by elevated NO generation in brains of heroin addicts, these elevated levels of NO might have contributed to some of the known toxic effects of heroin (for example, reduced adult neurogenesis, mitochondrial pathology or disturbances in synaptic functioning).


Assuntos
Overdose de Drogas/enzimologia , Dependência de Heroína/enzimologia , Heroína/intoxicação , Entorpecentes/intoxicação , Óxido Nítrico Sintase/metabolismo , Núcleo Hipotalâmico Paraventricular/enzimologia , Lobo Temporal/enzimologia , Adolescente , Adulto , Estudos de Casos e Controles , Overdose de Drogas/mortalidade , Feminino , Glutamato Descarboxilase/metabolismo , Glutamato-Amônia Ligase/metabolismo , Dependência de Heroína/mortalidade , Humanos , Masculino , Pessoa de Meia-Idade , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/patologia , Lobo Temporal/efeitos dos fármacos , Lobo Temporal/patologia
10.
Neuroscience ; 248: 127-35, 2013 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-23707802

RESUMO

Estrogen receptors (ERs) α and ß are involved in the regulation of the nitrergic system in the supraoptic (SON) and paraventricular (PVN) nuclei under basal conditions. In this study we have assessed whether ERs are also involved in the modulation of the nitrergic system in the SON and PVN under acute systemic hypertonic conditions. Adult ovariectomized rats received a single injection of either estradiol, a selective ERα agonist, a selective ERß agonist, a selective ERα antagonist, a selective ERß antagonist or vehicle. Twenty-four hours later, animals received one i.p. injection of 1.5M NaCl to induce osmotic stress and were sacrificed after two additional hours. The number of NADPH-diaphorase-positive cells in the SON and PVN was determined. Their number in the SON was not affected by NaCl administration, whereas in the four PVN subdivisions it was decreased after NaCl administration. Estradiol and the ERα agonist prevented the action of NaCl in the four subdivisions of the PVN. In contrast, the inhibition of ERα enhanced the effect of NaCl, inducing a further decrease in the number of NADPH-diaphorase-positive cells. Moreover, the ERß agonist enhanced and the ERß antagonist blocked the effect of NaCl on the number of NADPH-diaphorase-positive neurons in the SON and in the medial magnocellular subdivision of the PVN. These findings indicate that estradiol regulates the nitrergic system in the SON and PVN under acute osmotic stress conditions, but the effects specifically depend on the anatomical subregions and different ERs.


Assuntos
Estradiol/fisiologia , NADPH Desidrogenase/metabolismo , Neurônios/enzimologia , Pressão Osmótica , Núcleo Hipotalâmico Paraventricular/enzimologia , Estresse Fisiológico , Núcleo Supraóptico/enzimologia , Animais , Estradiol/farmacologia , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/antagonistas & inibidores , Feminino , Ovariectomia , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Ratos , Ratos Wistar , Núcleo Supraóptico/efeitos dos fármacos
11.
Dev Neurobiol ; 73(7): 502-17, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23420586

RESUMO

In the sexually dimorphic anteroventral periventricular nucleus (AVPV) of the hypothalamus, females have a greater number of tyrosine hydroxylase-immunoreactive (TH-ir) and kisspeptin-immunoreactive (kisspeptin-ir) neurons than males. In this study, we used proteomics analysis and gene-deficient mice to identify proteins that regulate the number of TH-ir and kisspeptin-ir neurons in the AVPV. Analysis of protein expressions in the rat AVPV on postnatal day 1 (PD1; the early phase of sex differentiation) using two-dimensional fluorescence difference gel electrophoresis followed by MALDI-TOF-MS identified collapsin response mediator protein 4 (CRMP4) as a protein exhibiting sexually dimorphic expression. Interestingly, this sexually differential expressions of CRMP4 protein and mRNA in the AVPV was not detected on PD6. Prenatal testosterone exposure canceled the sexual difference in the expression of Crmp4 mRNA in the rat AVPV. Next, we used CRMP4-knockout (CRMP4-KO) mice to determine the in vivo function of CRMP4 in the AVPV. Crmp4 knockout did not change the number of kisspeptin-ir neurons in the adult AVPV in either sex. However, the number of TH-ir neurons was increased in the AVPV of adult female CRMP4-KO mice as compared with the adult female wild-type mice. During development, no significant difference in the number of TH-ir neurons was detected between sexes or genotypes on embryonic day 15, but a female-specific increase in TH-ir neurons was observed in CRMP4-KO mice on PD1, when the sex difference was not yet apparent in wild-type mice. These results indicate that CRMP4 regulates the number of TH-ir cell number in the female AVPV.


Assuntos
Proteínas do Tecido Nervoso/fisiologia , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiologia , Caracteres Sexuais , Tirosina 3-Mono-Oxigenase/metabolismo , Animais , Animais Recém-Nascidos , Contagem de Células , Feminino , Kisspeptinas/metabolismo , Kisspeptinas/fisiologia , Masculino , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Neurônios/citologia , Neurônios/enzimologia , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/enzimologia , Gravidez , Ratos , Ratos Sprague-Dawley
12.
J Neuroendocrinol ; 25(3): 244-50, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22967140

RESUMO

Modulation of the nitric oxide producing system (demonstrated via the NADPH-diaphorase histochemical reaction) by oestradiol has been established in several structures of the rat brain. The present study aimed to explore the possible regulation of NADPH-diaphorase activity by oestradiol in neurones of the supraoptic (SON) and paraventricular (PVN) nuclei and the role of oestrogen receptors (ERα and ERß) in this regulation. Adult ovariectomised rats were divided into six groups and injected either with vehicle or a single dose of oestradiol, a selective ERα agonist-PPT [4,4',4″-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol], a selective ERß agonist-DPN [2,3-bis(4-hydroxyphenyl)-propionitrile], a selective ERα antagonist-MPP [1,3-bis(4-hydroxyphenyl)-4-methyl-5-[4-(2-piperidinylethoxy)phenol]-1H-pyrazole dihydrochloride] or a selective ERß antagonist-PHTPP (4-[2-phenyl-5,7-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-3-yl]phenol). The number of NADPH-diaphorase positive elements in the SON and the PVN was modulated by both ERs but, depending on the nucleus, ERα and ERß ligands induced different effects. These results suggest that the regulation of nitrergic system by ERs may play a role in the control of oestrogen-dependent physiological mechanisms regulated by the SON and the PVN.


Assuntos
NADPH Desidrogenase/metabolismo , Ovariectomia , Núcleo Hipotalâmico Paraventricular/enzimologia , Receptores de Estrogênio/fisiologia , Núcleo Supraóptico/enzimologia , Animais , Feminino , Núcleo Hipotalâmico Paraventricular/citologia , Ratos , Ratos Wistar , Núcleo Supraóptico/citologia
13.
Am J Physiol Heart Circ Physiol ; 302(3): H733-41, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22140041

RESUMO

Mineralocorticoid excess increases superoxide production by activating NADPH oxidase (NOX), and intracerebroventricular infusions of NADPH oxidase inhibitors attenuate aldosterone (Aldo)/salt-induced hypertension. It has been hypothesized that increased reactive oxygen species (ROS) in the brain may be a key mechanism in the development of hypertension. The present study investigated the brain regional specificity of NADPH oxidase and the role of NOX2 and NOX4 NADPH oxidase subunits in the hypothalamic paraventricular nucleus (PVN) in Aldo/salt-induced hypertension. PVN injections of adenoviral vectors expressing small interfering (si)RNA targeting NOX2 (AdsiRNA-NOX2) or NOX4 (AdsiRNA-NOX4) mRNAs were used to knock down NOX2 and NOX4 proteins. Three days later, delivery of Aldo (0.2 mg·kg(-1)·day(-1) sc) via osmotic pump commenced and 1% NaCl was provided in place of water. PVN injections of either AdsiRNA-NOX2 or AdsiRNA-NOX4 significantly attenuated the development of Aldo/NaCl-induced hypertension. In an additional study, Aldo/salt-induced hypertension was also significantly attenuated in NOX2 (genomic) knockout mice compared with wild-type controls. When animals from both functional studies underwent ganglionic blockade, there was a reduced fall in blood pressure in the NOX2 and NOX4 knockdown/knockout mice. Western blot analyses of the PVN of siRNA-NOX2- or siRNA-NOX4-injected mice confirmed a marked reduction in the expression of NOX2 or NOX4 protein. In cultured PVN neurons, silencing either NOX2 or NOX4 protein production by culturing PVN cells with siRNA-NOX2 or siRNA-NOX4 attenuated Aldo-induced ROS. These data indicate that both NOX2 and NOX4 in the PVN contribute to elevated sympathetic activity and the hypertensivogenic actions induced by mineralocorticoid excess.


Assuntos
Terapia Genética/métodos , Hipertensão/terapia , Glicoproteínas de Membrana/genética , NADPH Oxidases/genética , Núcleo Hipotalâmico Paraventricular/enzimologia , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Adenoviridae/genética , Aldosterona/farmacologia , Animais , Sistema Nervoso Autônomo/enzimologia , Sistema Nervoso Autônomo/fisiopatologia , Pressão Sanguínea/fisiologia , Feminino , Terapia Genética/instrumentação , Hipertensão/induzido quimicamente , Hipertensão/fisiopatologia , Bombas de Infusão , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/metabolismo , RNA Interferente Pequeno/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Cloreto de Sódio/farmacologia
14.
Hypertension ; 58(5): 966-73, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21968757

RESUMO

Our previous studies have shown that the decreased NO and increased glutamatergic mechanisms on sympathetic regulation within the paraventricular nucleus (PVN) may contribute to the elevated sympathoexcitation during chronic heart failure (CHF). In the present study, we investigated the effects of neuronal NO synthase (nNOS) gene transfer on N-methyl-D-aspartic acid receptor subunit NR(1) in the rats with a coronary ligation model of CHF. Adenovirus vectors encoding nNOS (AdnNOS) or adenovirus vectors encoding ß-galactosidase were transfected into the PVN in vivo. Five days after application of AdnNOS, the increased expression of nNOS within the PVN was confirmed by NADPH-diaphorase staining, real-time PCR, and Western blot. In anesthetized rats, AdnNOS treatment significantly enhanced the blunted renal sympathetic nerve activity, blood pressure, and heart rate responses to NO synthase inhibitor N(G)-monomethyl-L-arginine in the rats with CHF compared with CHF-adenovirus vectors encoding ß-galactosidase group. AdnNOS significantly decreased the enhanced renal sympathetic nerve activity, blood pressure, and heart rate responses to N-methyl-D-aspartic acid in the rats with CHF (renal sympathetic nerve activity: 44±2% versus 79±6%; P<0.05) compared with CHF-adenovirus vectors encoding the ß-galactosidase group. AdnNOS transfection significantly reduced the increased NR(1) receptor mRNA expression (Δ35±5%) and protein levels (Δ24±4%) within the PVN in CHF rats. Furthermore, in neuronal NG-108 cells, NR(1) receptor protein expression decreased in a dose-dependent manner after AdnNOS transfection. According to our results, nNOS downregulation enhances glutamate transmission in the PVN by increasing NR(1) subunit expression. This mechanism may enhance renal sympathetic nerve activity in CHF rats.


Assuntos
Insuficiência Cardíaca/fisiopatologia , N-Metilaspartato/metabolismo , Óxido Nítrico Sintase/genética , Núcleo Hipotalâmico Paraventricular/enzimologia , Transfecção , Adenoviridae , Análise de Variância , Animais , Western Blotting , Modelos Animais de Doenças , Regulação para Baixo , Fármacos Atuantes sobre Aminoácidos Excitatórios/metabolismo , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Imunofluorescência , Masculino , Óxido Nítrico Sintase/antagonistas & inibidores , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real
15.
Am J Physiol Heart Circ Physiol ; 301(6): H2402-12, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21963832

RESUMO

Angiotensin (ANG)-converting enzyme (ACE)2 in brain regions such as the paraventricular nucleus (PVN) controlling cardiovascular function may be involved in the regulation of sympathetic outflow in chronic heart failure (CHF). The purpose of this study was to determine if ACE2 plays a role in the central regulation of sympathetic outflow by regulating neuronal nitric oxide (NO) synthase (nNOS) in the PVN. We investigated ACE2 and nNOS expression within the PVN of rats with CHF. We then determined the effects of ACE2 gene transfer in the PVN on the contribution of NO-mediated sympathoinhibition in rats with CHF. The results showed that there were decreased expressions for ACE2, the ANG-(1-7) receptor, and nNOS within the PVN of rats with CHF. After the application of adenovirus vectors encoding ACE2 (AdACE2) into the PVN, the increased expression of ACE2 in the PVN was confirmed by Western blot analysis. AdACE2 transfection significantly increased nNOS protein levels (change of 50 ± 5%) in the PVN of CHF rats. In anesthetized rats, AdACE2 treatment attenuated the responses of renal sympathetic nerve activity (RSNA), mean arterial pressure, and heart rate to the NOS inhibitor N-monomethyl-L-arginine in rats with CHF (RSNA: 28 ± 3% vs. 16 ± 3%, P < 0.05) compared with CHF + AdEGFP group. Furthermore, neuronal NG-108 cells incubated with increasing doses of AdACE2 showed a dose-dependent increase in nNOS protein expression (60% at the highest dose). Taken together, our data highlight the importance of increased expression and subsequent interaction of ACE2 and nNOS within the PVN, leading to a reduction in sympathetic outflow in the CHF condition.


Assuntos
Sistema Cardiovascular/inervação , Terapia Genética , Insuficiência Cardíaca/terapia , Rim/inervação , Inibição Neural , Óxido Nítrico/metabolismo , Núcleo Hipotalâmico Paraventricular/enzimologia , Peptidil Dipeptidase A/biossíntese , Sistema Nervoso Simpático/enzimologia , Enzima de Conversão de Angiotensina 2 , Animais , Pressão Sanguínea , Western Blotting , Linhagem Celular Tumoral , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Insuficiência Cardíaca/enzimologia , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/fisiopatologia , Frequência Cardíaca , Humanos , Masculino , Óxido Nítrico Sintase Tipo I/antagonistas & inibidores , Óxido Nítrico Sintase Tipo I/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Peptidil Dipeptidase A/genética , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/metabolismo , Ratos , Ratos Wistar , Receptores Acoplados a Proteínas G/metabolismo , Sistema Nervoso Simpático/fisiopatologia , Fatores de Tempo , Transfecção , Regulação para Cima
16.
Cardiovasc Res ; 92(3): 401-8, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21952934

RESUMO

AIMS: Angiotensin II (Ang II) has been shown to have both central and peripheral effects in mediating hypertension, for which the hypothalamic paraventricular nucleus (PVN) is an important brain cardio-regulatory centre. Angiotensin-converting enzyme 2 (ACE2) has been identified as a negative regulator of the pro-hypertensive actions of Ang II. Recent findings from our laboratory suggest that Ang II infusion decreases ACE2 expression in the PVN. In the present study, we hypothesized that ACE2 overexpression in the PVN will have beneficial effects in counteracting Ang II-induced hypertension. METHODS AND RESULTS: Male Sprague-Dawley rats were used in this study. Bilateral microinjection of an adenovirus encoding hACE2 (Ad-ACE2) into the PVN was used to overexpress ACE2 within this region. Mean arterial pressure measured by radiotelemetry was significantly increased after 14 days in Ang II-infused (200 ng/kg/min) rats vs. saline-infused controls (162.9 ± 3.6 vs. 102.3 ± 1.5 mmHg). Bilateral PVN microinjection of Ad-ACE2 attenuated this Ang II-induced hypertension (130.2 ± 5.7 vs. 162.9 ± 3.6 mmHg). ACE2 overexpression also significantly decreased AT(1)R and ACE expression and increased AT(2)R and Mas expression in the PVN. Additionally, ACE2 overexpression in the PVN attenuated the Ang II-induced increase in the expression of the pro-inflammatory cytokines tumour necrosis factor-α, interleukin (IL)-1ß and IL-6 in the PVN. CONCLUSION: Our findings suggest that attenuation of pro-inflammatory cytokines in the PVN in combination with the shift of the renin-angiotensin system towards the anti-hypertensive axis (ACE2/Ang-(1-7)/Mas) may be responsible for the overall beneficial effects of ACE2 overexpression in the PVN on the Ang II-induced hypertensive response.


Assuntos
Angiotensina II , Terapia Genética , Hipertensão/prevenção & controle , Núcleo Hipotalâmico Paraventricular/enzimologia , Peptidil Dipeptidase A/biossíntese , Adenoviridae/genética , Enzima de Conversão de Angiotensina 2 , Animais , Pressão Sanguínea , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Hipertensão/induzido quimicamente , Hipertensão/enzimologia , Hipertensão/genética , Mediadores da Inflamação/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Microinjeções , Peptidil Dipeptidase A/genética , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 2 de Angiotensina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sistema Renina-Angiotensina , Telemetria , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo
17.
Cardiovasc Res ; 92(2): 348-57, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21831995

RESUMO

AIMS: Previously, we showed an enhanced excitatory (N-methyl d-aspartate receptor-NR(1)) and decreased inhibitory neuronal nitric oxide (NO) synthase (nNOS) influence within the paraventricular nucleus (PVN) of rats with chronic heart failure (CHF). Although NR(1) and nNOS are normally linked, they can be disconnected by nNOS sequestering with nNOS-associated protein (CAPON). The aim of this study was to elucidate the underlying mechanism for the disconnection between increased expression of NR(1) and decreased nNOS in the PVN of rats with CHF which leads to enhanced sympathoexcitation. METHODS AND RESULTS: CAPON expression was augmented while nNOS expression was decreased in the PVN of rats with CHF (6-8 weeks after left coronary artery ligation). Angiotensin II (Ang II) type I receptor (AT(1)) antagonist losartan (Los) treatment in rats with CHF reduced renal sympathetic nerve activity with concomitant normalization of protein expression of CAPON and nNOS in the PVN. Los treatment also reversed the blunting of endogenous NO-mediated sympatho-inhibition in rats with CHF. Moreover, Ang II-induced increase in CAPON expression in NG108 neuronal cells was also ameliorated by Los. CONCLUSION: Blocking AT(1) receptors prevents the overexpression of CAPON and concomitant decrease in nNOS in the PVN, resulting in attenuation of sympathoexcitation commonly observed in CHF. Taken together, our data highlight the importance of altered expression and subsequent interaction of nNOS and CAPON within the PVN, leading to increased sympathoexcitation in CHF. Identifying this crucial nNOS/CAPON interaction regulated by AT(1) receptors may provide an important potential therapeutic target in CHF.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Angiotensina II/metabolismo , Insuficiência Cardíaca/enzimologia , Óxido Nítrico Sintase Tipo I/metabolismo , Núcleo Hipotalâmico Paraventricular/enzimologia , Sistema Nervoso Simpático/fisiopatologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Linhagem Celular Tumoral , Doença Crônica , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Regulação para Baixo , Inibidores Enzimáticos/administração & dosagem , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/fisiopatologia , Hemodinâmica/efeitos dos fármacos , Imuno-Histoquímica , Rim/inervação , Losartan/farmacologia , Masculino , Microinjeções , Óxido Nítrico Sintase Tipo I/antagonistas & inibidores , Óxido Nítrico Sintase Tipo I/genética , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Interferência de RNA , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Receptores de N-Metil-D-Aspartato/metabolismo , Sistema Nervoso Simpático/efeitos dos fármacos , Fatores de Tempo , ômega-N-Metilarginina/administração & dosagem
18.
Tohoku J Exp Med ; 222(4): 251-63, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21135513

RESUMO

Proinflammatory cytokines, including tumor necrosis factor (TNF)-α, augment the progression of heart failure (HF) that is characterized by sympathoexcitation. In this study, we explored the role of TNF-α in hypothalamic paraventricular nucleus (PVN) in the exaggerated sympathetic activity observed in HF. Heart failure rats were made by ligating the left anterior descending coronary artery. The expression levels of angiotensin type 1 receptor (AT1-R) and neurotransmitters were analyzed in the PVN of HF rats that received direct PVN infusion of a TNF-α blocker (pentoxifylline or etanercept) or vehicle. Sham-operated control (SHAM) or HF rats were treated for 4 weeks through PVN infusion with each TNF-α blocker or vehicle. Rats with HF had higher levels of glutamate, norepinephrine, AT1-R and tyrosine hydroxylase (TH), and lower levels of gamma-aminobutyric acid (GABA), neuronal nitric oxide synthase (nNOS) and the 67-kDa isoform of glutamate decarboxylase (GAD67) in the PVN when compared to SHAM rats. Plasma levels of cytokines, norepinephrine and angiotensin II and renal sympathetic nerve activity (RSNA) were increased in HF rats. PVN infusion of pentoxifylline or etanercept attenuated the decreases in PVN GABA, nNOS and GAD67, and the increases in RSNA and PVN glutamate, norepinephrine, TH and AT1-R observed in HF rats. We have developed a novel method for chronic and continuous infusion of drugs directly into the PVN and provided evidence that TNF-α in the PVN modulates neurotransmitters and the expression of AT1 receptor, which could account for exaggerated sympathetic activity in HF.


Assuntos
Insuficiência Cardíaca/fisiopatologia , Neurotransmissores/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Receptor Tipo 1 de Angiotensina/metabolismo , Sistema Nervoso Simpático/fisiopatologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Regulação da Expressão Gênica , Glutamato Descarboxilase/metabolismo , Insuficiência Cardíaca/sangue , Insuficiência Cardíaca/diagnóstico por imagem , Testes de Função Cardíaca , Hemodinâmica , Mediadores da Inflamação/metabolismo , Rim/inervação , Rim/metabolismo , Rim/fisiopatologia , Masculino , Modelos Biológicos , Óxido Nítrico Sintase Tipo I/genética , Óxido Nítrico Sintase Tipo I/metabolismo , Núcleo Hipotalâmico Paraventricular/enzimologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/genética , Sistema Nervoso Simpático/enzimologia , Tirosina 3-Mono-Oxigenase/metabolismo , Ultrassonografia
19.
Circ Res ; 106(11): 1763-74, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20413786

RESUMO

RATIONALE: Myocardial infarction (MI)-induced heart failure is characterized by central nervous system-driven sympathoexcitation and deteriorating cardiac function. The paraventricular nucleus (PVN) of the hypothalamus is a key regulator of sympathetic nerve activity and is implicated in heart failure. Redox signaling in the PVN and other central nervous system sites is a primary mechanism of neuro-cardiovascular regulation, and excessive oxidant production by activation of NADPH oxidases (Noxs) is implicated in some neuro-cardiovascular diseases. OBJECTIVE: We tested the hypothesis that Nox-mediated redox signaling in the PVN contributes to MI-induced sympathoexcitation and cardiac dysfunction in mice. METHODS AND RESULTS: Real-time PCR revealed that Nox4 was the most abundantly expressed Nox in PVN under basal conditions. Coronary arterial ligation (MI) caused a selective upregulation of this homolog compared to Nox1 and Nox2. Adenoviral gene transfer of Nox4 (AdsiNox4) to PVN (bilateral) attenuated MI-induced superoxide formation in this brain region (day 14) to the same level as that produced by PVN-targeted gene transfer of cytoplasmic superoxide dismutase (AdCu/ZnSOD). MI mice treated with AdsiNox4 or AdCu/ZnSOD in the PVN showed marked improvement in cardiac function as assessed by echocardiography and left ventricular hemodynamic analysis. This was accompanied by significantly diminished sympathetic outflow and apoptosis in the periinfarct region of the heart. CONCLUSIONS: These results suggest that MI causes dysregulation of Nox4-mediated redox signaling in the PVN, which leads to sympathetic overactivation and a decline in cardiac function. Targeted inhibition of oxidant signaling in the PVN could provide a novel treatment for MI-induced heart failure.


Assuntos
Apoptose , Inativação Gênica , Insuficiência Cardíaca/enzimologia , Coração/inervação , Infarto do Miocárdio/enzimologia , Miocárdio/patologia , NADPH Oxidases/metabolismo , Núcleo Hipotalâmico Paraventricular/enzimologia , Sistema Nervoso Simpático/fisiopatologia , Adenoviridae/genética , Animais , Catalase/genética , Catalase/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Bloqueadores Ganglionares/farmacologia , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/prevenção & controle , Hemodinâmica , Peróxido de Hidrogênio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/terapia , NADPH Oxidase 4 , NADPH Oxidases/genética , Norepinefrina/urina , Oxirredução , Interferência de RNA , Transdução de Sinais , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxidos/metabolismo , Sistema Nervoso Simpático/efeitos dos fármacos , Sistema Nervoso Simpático/metabolismo , Fatores de Tempo , Função Ventricular Esquerda
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA