Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 207
Filtrar
Más filtros

Base de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cardiovasc Pharmacol ; 84(4): 468-478, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39115898

RESUMEN

ABSTRACT: The hypothalamic paraventricular nucleus (PVN) plays a central role in regulating cardiovascular activity and blood pressure. We administered hydroxylamine hydrochloride (HA), a cystathionine-ß-synthase inhibitor, into the PVN to suppress endogenous hydrogen sulfide and investigate its effects on the mitogen-activated protein kinase (MAPK) pathway in high salt (HS)-induced hypertension. We randomly divided 40 male Dahl salt-sensitive rats into 4 groups: the normal salt (NS) + PVN vehicle group, the NS + PVN HA group, the HS + PVN vehicle group, and the HS + PVN HA group, with 10 rats in each group. The rats in the NS groups were fed a NS diet containing 0.3% NaCl, while the HS groups were fed a HS diet containing 8% NaCl. The mean arterial pressure was calculated after noninvasive measurement using an automatic sphygmomanometer to occlude the tail cuff once a week. HA or vehicle was infused into the bilateral PVN using Alzet osmotic mini pumps for 6 weeks after the hypertension model was successfully established. We measured the levels of H 2 S in the PVN and plasma norepinephrine using enzyme linked immunosorbent assay. In addition, we assessed the parameters of the MAPK pathway, inflammation, and oxidative stress through western blotting, immunohistochemical analysis, or real-time polymerase chain reaction. In this study, we discovered that decreased levels of endogenous hydrogen sulfide in the PVN contributed to the onset of HS-induced hypertension. This was linked to the activation of the MAPK signaling pathway, proinflammatory cytokines, and oxidative stress in the PVN, as well as the activation of the sympathetic nervous system.


Asunto(s)
Modelos Animales de Enfermedad , Sulfuro de Hidrógeno , Hipertensión , Núcleo Hipotalámico Paraventricular , Ratas Endogámicas Dahl , Cloruro de Sodio Dietético , Animales , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/enzimología , Núcleo Hipotalámico Paraventricular/fisiopatología , Masculino , Sulfuro de Hidrógeno/metabolismo , Hipertensión/inducido químicamente , Hipertensión/fisiopatología , Hipertensión/metabolismo , Norepinefrina/metabolismo , Hidroxilamina/farmacología , Presión Arterial/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratas , Estrés Oxidativo/efectos de los fármacos , Inhibidores Enzimáticos/farmacología
2.
Cardiovasc Toxicol ; 24(9): 904-917, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39008239

RESUMEN

Hypertension is a globally prevalent disease, but the pathogenesis remains largely unclear. AMP-activated protein kinase (AMPK) is a nutrition-sensitive signal of cellular energy metabolism, which has a certain influence on the development of hypertension. Previously, we found a down-regulation of the phosphorylated (p-) form of AMPK, and the up-regulation of the angiotensin II type 1 receptor (AT1-R) and that of p-ERK1/2 in the hypothalamic paraventricular nucleus (PVN) of hypertensive rats. However, the exact mechanism underlying the relationship between AMPK and AT1-R in the PVN during hypertension remains unclear. Thus, we hypothesized that AMPK modulates AT1-R through the ERK1/2-NF-κB pathway in the PVN, thereby inhibiting sympathetic nerve activity and improving hypertension. To examine this hypothesis, we employed a renovascular hypertensive animal model developed via two-kidney, one-clip (2K1C) and sham-operated (SHAM). Artificial cerebrospinal fluid (aCSF), used as vehicle, or 5-amino-1-ß-D-ribofuranosyl-imidazole-4-carboxamide (AICAR, an AMPK activator, 60 µg/day) was microinjected bilaterally in the PVN of these rats for 4 weeks. In 2K1C rats, there an increase in systolic blood pressure (SBP) and circulating norepinephrine (NE). Also, the hypertensive rats had lowered expression of p-AMPK and p-AMPK/AMPK, elevated expression of p-ERK1/2, p-ERK1/2/ERK1/2 and AT1-R, increased NF-κB p65 activity in the PVN compared with the levels of these biomarkers in SHAM rats. Four weeks of bilateral PVN injection of AMPK activator AICAR, attenuated the NE level and SBP, increased the expression of p-AMPK and p-AMPK/AMPK, lessened the NF-κB p65 activity, decreased the expression of p-ERK1/2, p-ERK1/2/ERK1/2 and AT1-R in the PVN of 2K1C rats. Data from this study imply that the activation of AMPK within the PVN suppressed AT1-R expression through inhibiting the ERK1/2-NF-κB pathway, decreased the activity of the sympathetic nervous system, improved hypertension.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Modelos Animales de Enfermedad , Activación Enzimática , Hipertensión Renovascular , Proteína Quinasa 3 Activada por Mitógenos , Núcleo Hipotalámico Paraventricular , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1 , Animales , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/enzimología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/fisiopatología , Hipertensión Renovascular/fisiopatología , Hipertensión Renovascular/enzimología , Hipertensión Renovascular/metabolismo , Hipertensión Renovascular/tratamiento farmacológico , Masculino , Proteínas Quinasas Activadas por AMP/metabolismo , Fosforilación , Receptor de Angiotensina Tipo 1/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Factor de Transcripción ReIA/metabolismo , Ribonucleótidos/farmacología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Sistema Nervioso Simpático/fisiopatología , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , FN-kappa B/metabolismo , Transducción de Señal , Antihipertensivos/farmacología , Ratas
3.
Am J Hypertens ; 37(9): 682-691, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-38782571

RESUMEN

BACKGROUND: In the hypothalamic paraventricular nucleus (PVN) of spontaneously hypertensive rats (SHRs), the expression of the testis-specific protein, Y-encoded-like 2 (TSPYL2) and the phosphorylation level of Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) are higher comparing with the normotensive Wistar Kyoto rats (WKY). But how they are involved in hypertension remains unclear. TSPYL2 may interact with JAK2/STAT3 in PVN to sustain high blood pressure during hypertension. METHODS: Knockdown of TSPYL2 via adeno-associated virus (AAV) carrying shRNA was conducted through bilateral microinjection into the PVN of SHR and WKY rats. JAK2/STAT3 inhibition was achieved by intraperitoneally or PVN injection of AG490 into the SHRs. Blood pressure (BP), plasma norepinephrine (NE), PVN inflammatory response, and PVN oxidative stress were measured. RESULTS: TSPYL2 knock-down in the PVN of SHRs but not WKYs led to reduced BP and plasma NE, deactivation of JAK2/STAT3, decreased expression of pro-inflammatory cytokine IL-1ß, and increased expression of anti-inflammatory cytokine IL-10 in the PVN. Meanwhile, AG490 administrated in both ways reduced the BP in the SHRs and deactivated JAK2/STAT3 but failed to change the expression of TSPYL2 in PVN. AG490 also downregulated expression of IL-1ß and upregulated expression of IL-10. Both knockdown of TSPYL2 and inhibition of JAK2/STAT3 can reduce the oxidative stress in the PVN of SHRs. CONCLUSION: JAK2/STAT3 is regulated by TSPYL2 in the PVN of SHRs, and PVN TSPYL2/JAK2/STAT3 is essential for maintaining high BP in hypertensive rats, making it a potential therapeutic target for hypertension.


Asunto(s)
Presión Sanguínea , Hipertensión , Janus Quinasa 2 , Núcleo Hipotalámico Paraventricular , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Factor de Transcripción STAT3 , Transducción de Señal , Animales , Masculino , Ratas , Modelos Animales de Enfermedad , Hipertensión/metabolismo , Hipertensión/fisiopatología , Janus Quinasa 2/metabolismo , Norepinefrina/metabolismo , Estrés Oxidativo/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/enzimología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Fosforilación , Factor de Transcripción STAT3/metabolismo , Tirfostinos/farmacología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo
4.
Thyroid ; 32(1): 105-114, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34726513

RESUMEN

Background: Thyrotropin-releasing hormone (TRH) was the first hypothalamic hormone isolated that stimulates pituitary thyrotropin (TSH) secretion. TRH was also later found to be a stimulator of pituitary prolactin and distributed throughout the brain, gastrointestinal tract, and pancreatic ß cells. We previously reported the development of TRH null mice (conventional TRHKO), which exhibit characteristic tertiary hypothyroidism and impaired glucose tolerance due to insufficient insulin secretion. Although in the past five decades many investigators, us included, have attempted to determine the hypothalamic nucleus responsible for the hypothalamic-pituitary-thyroid (HPT) axis, it remained obscure because of the broad expression of TRH. Methods: To determine the hypothalamic region functionally responsible for the HPT axis, we established paraventricular nucleus (PVN)-specific TRH knockout (PVN-TRHKO) mice by mating Trh floxed mice and single-minded homolog 1 (Sim1)-Cre transgenic mice. We originally confirmed that most Sim1 was expressed in the PVN using Sim1-Cre/tdTomato mice. Results: These PVN-TRHKO mice exhibited tertiary hypothyroidism similar to conventional TRHKO mice; however, they did not show the impaired glucose tolerance observed in the latter, suggesting that TRH from non-PVN sources is essential for glucose regulation. In addition, a severe reduction in prolactin expression was observed in the pituitary of PVN-TRHKO mice compared with that in TRHKO mice. Conclusions: These findings are conclusive evidence that the PVN is the center of the HPT axis for regulation of serum levels of thyroid hormones and that the serum TSH levels are not decreased in tertiary hypothyroidism. We also noted that TRH from the PVN regulated prolactin, whereas TRH from non-PVN sources regulated glucose metabolism.


Asunto(s)
Núcleo Hipotalámico Paraventricular/enzimología , Hormonas Tiroideas/metabolismo , Hormona Liberadora de Tirotropina/metabolismo , Animales , Modelos Animales de Enfermedad , Ratones , Núcleo Hipotalámico Paraventricular/fisiopatología , Estadísticas no Paramétricas
5.
Cardiovasc Toxicol ; 21(12): 1045-1057, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34537923

RESUMEN

Astaxanthin (AST) has a variety of biochemical effects, including anti-inflammatory, antioxidative, and antihypertensive functions. The aim of the present study was to determine whether AST ameliorates blood pressure in salt-induced prehypertensive rats by ROS/MAPK/NF-κB pathways in hypothalamic paraventricular nucleus.To explore the central effects of AST on the development of blood pressure, prehypertensive rats were induced by a high-salt diet (HS, 8% NaCl) and its control groups were treated with normal-salt diet (NS, 0.3% NaCl). The Dahl salt-sensitive (S) rats with HS diet for 6 weeks received AST or vehicle by gastric perfusion for 6 weeks. Compared to those with NS diet, rats with HS diet exhibited increased mean arterial pressure (MAP) and heart rate (HR). These increases were associated with higher plasma level of norepinephrine (NE), interleukin 1ß (IL-1ß), and interleukin 6 (IL-6); elevated PVN level of reactive oxygen species (ROS), NOX2, and NOX4, that of IL-1ß, IL-6, monocyte chemotactic protein 1 (MCP-1), tyrosine hydroxylase (TH), phosphorylation extracellular-signal-regulated kinase (p-ERK1/2), phosphorylation Jun N-terminal kinases (p-JNK), nuclear factor-kappa B (NF-κB) activity; and lower levels of IL-10, superoxide dismutase (SOD), and catalase (CAT) in the PVN. In addition, our data demonstrated that chronic AST treatment ameliorated these changes in the HS but not NS diet rats. These data suggested that AST could alleviate prehypertensive response in HS-induced prehypertension through ROS/MAPK/NF-κB pathways in the PVN.


Asunto(s)
Antihipertensivos/farmacología , Presión Arterial/efectos de la radiación , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Prehipertensión/prevención & control , Especies Reactivas de Oxígeno/metabolismo , Animales , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Modelos Animales de Enfermedad , Masculino , Núcleo Hipotalámico Paraventricular/enzimología , Núcleo Hipotalámico Paraventricular/fisiopatología , Fosforilación , Prehipertensión/enzimología , Prehipertensión/etiología , Prehipertensión/fisiopatología , Ratas Endogámicas Dahl , Transducción de Señal , Cloruro de Sodio Dietético , Xantófilas/farmacología
6.
Cardiovasc Toxicol ; 21(9): 721-736, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34076830

RESUMEN

Apigenin, identified as 4', 5, 7-trihydroxyflavone, is a natural flavonoid compound that has many interesting pharmacological activities and nutraceutical potential including anti-inflammatory and antioxidant functions. Chronic, low-grade inflammation and oxidative stress are involved in both the initiation and progression of hypertension and hypertension-induced cardiac hypertrophy. However, whether or not apigenin improves hypertension and cardiac hypertrophy through modulating NADPH oxidase-dependent reactive oxygen species (ROS) generation and inflammation in hypothalamic paraventricular nucleus (PVN) has not been reported. This study aimed to investigate the effects of apigenin on hypertension in spontaneously hypertensive rats (SHRs) and its possible central mechanism of action. SHRs and Wistar-Kyoto (WKY) rats were randomly assigned and treated with bilateral PVN infusion of apigenin or vehicle (artificial cerebrospinal fluid) via osmotic minipumps (20 µg/h) for 4 weeks. The results showed that after PVN infusion of apigenin, the mean arterial pressure (MAP), heart rate, plasma norepinephrine (NE), Beta 1 receptor in kidneys, level of phosphorylation of PKA in the ventricular tissue and cardiac hypertrophy, perivascular fibrosis, heart level of oxidative stress, PVN levels of oxidative stress, interleukin 1ß (IL-1ß), interleukin 6 (IL-6), iNOS, monocyte chemotactic protein 1 (MCP-1), tyrosine hydroxylase (TH), NOX2 and NOX4 were attenuated and PVN levels of interleukin 10 (IL-10), superoxide dismutase 1 (Cu/Zn-SOD) and the 67-kDa isoform of glutamate decarboxylase (GAD67) were increased. These results revealed that apigenin improves hypertension and cardiac hypertrophy in SHRs which are associated with the down-regulation of NADPH oxidase-dependent ROS generation and inflammation in the PVN.


Asunto(s)
Antiinflamatorios/farmacología , Antihipertensivos/farmacología , Antioxidantes/farmacología , Apigenina/farmacología , Cardiomegalia/tratamiento farmacológico , Citocinas/metabolismo , Hipertensión/tratamiento farmacológico , NADPH Oxidasas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Animales , Presión Arterial/efectos de los fármacos , Cardiomegalia/enzimología , Cardiomegalia/fisiopatología , Modelos Animales de Enfermedad , Fibrosis , Hipertensión/enzimología , Hipertensión/fisiopatología , Masculino , Miocardio/metabolismo , Miocardio/patología , NADPH Oxidasas/genética , Núcleo Hipotalámico Paraventricular/enzimología , Núcleo Hipotalámico Paraventricular/fisiopatología , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos
7.
Am J Physiol Heart Circ Physiol ; 318(1): H34-H48, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31675258

RESUMEN

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.


Asunto(s)
Apoptosis , Presión Arterial , Caspasa 3/metabolismo , Hipertensión/prevención & control , Hipoxia/complicaciones , Núcleo Hipotalámico Paraventricular/enzimología , Área Preóptica/enzimología , Animales , Caspasa 3/genética , Ritmo Circadiano , Modelos Animales de Enfermedad , Frecuencia Cardíaca , Hipertensión/enzimología , Hipertensión/patología , Hipertensión/fisiopatología , Hipoxia/enzimología , Hipoxia/patología , Hipoxia/fisiopatología , Masculino , Estrés Oxidativo , Núcleo Hipotalámico Paraventricular/patología , Núcleo Hipotalámico Paraventricular/fisiopatología , Área Preóptica/patología , Área Preóptica/fisiopatología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas Sprague-Dawley , Transducción de Señal
8.
Acta Histochem ; 121(6): 690-694, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31227177

RESUMEN

Leptin, a hormone mainly produced by adipocytes in proportion to fat mass, is a key component in the regulation of energy homeostasis and reproductive, neuroendocrine, immune, and metabolic functions. Leptin binds to the leptin receptor, which is expressed throughout the central nervous system but particularly in neurons of several nuclei of the hypothalamus, such as the arcuate nucleus (ARC) and paraventricular nucleus (PVN). It has been found that nitric oxide (NO) plays an important role in mediating effects of leptin. Since PVN and ARC neurons are known to express leptin receptors, we investigated the effects of leptin on nicotinamide adenine dinucleotide phosphate-diaphorase (NADPH-d) reactivity in the PVN and ARC of male Wistar rats. Our results have shown that systemic administration of leptin resulted in increased NADPH-d positive cell number in the PVN and ARC, suggesting that both the PVN and ARC may be important centers in the hypothalamus for the leptin action, mediated by increased NO production. In addition, we have also observed that hypothalamic tanycytes in the ventral portion of the third ventricle were NADPH-d positive. We speculate that leptin may affect the release of neurohormones and hypothalamic neurogenesis by activating nitric oxide synthase in hypothalamic tanycytes.


Asunto(s)
Células Ependimogliales/enzimología , Leptina/farmacología , NADPH Deshidrogenasa/metabolismo , Núcleo Hipotalámico Paraventricular/enzimología , Animales , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/enzimología , Células Ependimogliales/citología , Masculino , Neuronas/citología , Neuronas/enzimología , Óxido Nítrico/metabolismo , Núcleo Hipotalámico Paraventricular/citología , Ratas , Ratas Wistar , Receptores de Leptina/metabolismo
9.
Am J Physiol Heart Circ Physiol ; 316(6): H1389-H1405, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30925093

RESUMEN

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.


Asunto(s)
Presión Sanguínea , Señalización del Calcio , Hipertensión/prevención & control , Neuronas/enzimología , Núcleo Hipotalámico Paraventricular/enzimología , ATPasas de Translocación de Protón/deficiencia , Receptor de Angiotensina Tipo 1/metabolismo , Receptores de Superficie Celular/deficiencia , Animales , Sistema Nervioso Autónomo/metabolismo , Sistema Nervioso Autónomo/fisiopatología , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Acetato de Desoxicorticosterona , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Hipertensión/enzimología , Hipertensión/genética , Hipertensión/fisiopatología , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Núcleo Hipotalámico Paraventricular/fisiopatología , Fosforilación , ATPasas de Translocación de Protón/genética , Receptor de Angiotensina Tipo 1/genética , Receptores de Superficie Celular/genética , Receptor de Prorenina
10.
Psychopharmacology (Berl) ; 236(4): 1293-1301, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30539267

RESUMEN

RATIONALE: Lycium barbarum polysaccharide (LBP) is known to promote reproductive functions. However, its role in noncontact erection (NCE) of penis initiated by brain regions including medial preoptic area (MPOA) and paraventricular nucleus (PVN) regions responsible for sexual behavior has not been investigated. OBJECTIVES: Therefore, this study initially investigated the effects of LBP on male sexual function, and subsequently, the mechanistic insight was investigated through assessing the expression of neuronal nitric oxide synthase (nNOS) in the MPOA and PVN. METHODS: The adult male rats were treated with 100 mg/kg of LBP or vehicle by oral gavage. Before and after 14 days of treatment, copulatory behavior and noncontact erection (NCE) were recorded. After the last behavioral test, the brain was isolated to measure nNOS expression in the MPOA and PVN. RESULTS: Data showed that LBP treatment significantly increased both the frequencies of intromission as well as ejaculation, compared to the control group. Whereas, a reduced post-ejaculatory interval was observed compared to same group on day 0. Furthermore, the treatment led to an increased intromission ratio, inter-intromission interval, and the number of MPOA nNOS-immunoreactive cells (nNOS-ir). Additionally, a significantly positive correlation between ejaculation frequency and MPOA nNOS-ir cells was recorded. Of note, LBP treatment had no effects on NCE and PVN nNOS-ir expression. CONCLUSION: These findings suggest that LBP enhances sexual behavior through increased nNOS expression in the MPOA in male rats.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Erección Peniana/efectos de los fármacos , Área Preóptica/efectos de los fármacos , Conducta Sexual Animal/efectos de los fármacos , Animales , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Femenino , Masculino , Neuronas/efectos de los fármacos , Neuronas/enzimología , Óxido Nítrico , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/enzimología , Erección Peniana/fisiología , Área Preóptica/enzimología , Ratas , Ratas Long-Evans , Conducta Sexual Animal/fisiología , Testículo/efectos de los fármacos , Testículo/enzimología
11.
J Cardiol ; 73(1): 81-88, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30487059

RESUMEN

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.


Asunto(s)
Arritmias Cardíacas/enzimología , Infarto del Miocardio/enzimología , Núcleo Hipotalámico Paraventricular/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Sistema Nervioso Simpático/enzimología , Animales , Arritmias Cardíacas/fisiopatología , Arritmias Cardíacas/prevención & control , Cromonas/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Masculino , Morfolinas/uso terapéutico , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/fisiopatología , Miocardio/enzimología , Norepinefrina/sangre , Ratas , Ratas Endogámicas SHR , Transducción de Señal/efectos de los fármacos
12.
Hypertension ; 72(3): 667-675, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30012866

RESUMEN

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.


Asunto(s)
Desnervación/métodos , Insuficiencia Cardíaca/fisiopatología , Riñón/inervación , Óxido Nítrico Sintasa de Tipo I/metabolismo , Núcleo Hipotalámico Paraventricular/fisiopatología , Sistema Nervioso Simpático/fisiopatología , Vías Aferentes/efectos de los fármacos , Vías Aferentes/fisiología , Animales , Capsaicina/farmacología , Enfermedad Crónica , Inhibidores Enzimáticos/farmacología , Masculino , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/enzimología , Ratas Sprague-Dawley , Fármacos del Sistema Sensorial/farmacología , Sistema Nervioso Simpático/efectos de los fármacos , omega-N-Metilarginina/farmacología
13.
Exp Physiol ; 103(3): 419-428, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29215757

RESUMEN

NEW FINDINGS: What is the central question of this study? Does chronic reduction of neuronally generated nitric oxide in the hypothalamic paraventricular nucleus affect the set-point regulation of blood pressure and sympathetic activity destined to the kidneys? What is the main finding and its importance? Within the hypothalamic paraventricular nucleus, nitric oxide generated by neuronal nitric oxide synthase plays a major constitutive role in suppressing long term the levels of both ongoing renal sympathetic activity and arterial pressure in conscious Wistar rats. This finding unequivocally demonstrates a mechanism by which the diencephalon exerts a tonic influence on sympathetic discharge to the kidney and may provide the basis for both blood volume and osmolality homeostasis. ABSTRACT: The paraventricular nucleus (PVN) of the hypothalamus plays a crucial role in cardiovascular and neuroendocrine regulation. Application of nitric oxide donors to the PVN stimulates GABAergic transmission, and may suppress sympathetic nerve activity (SNA) to lower arterial pressure. However, the role of endogenous nitric oxide within the PVN in regulating renal SNA chronically remains to be established in conscious animals. To address this, we used our previously established lentiviral vectors to knock down neuronal nitric oxide synthase (nNOS) selectively in the PVN of conscious Wistar rats. Blood pressure and renal SNA were monitored simultaneously and continuously for 21 days (n = 14) using radio-telemetry. Renal SNA was normalized to maximal evoked discharge and expressed as a percentage change from baseline. The PVN was microinjected bilaterally with a neurone-specific tetracycline-controllable lentiviral vector, expressing a short hairpin miRNA30 interference system targeting nNOS (n = 7) or expressing a mis-sense as control (n = 7). Recordings continued for a further 18 days. The vectors also expressed green fluorescent protein, and successful expression in the PVN and nNOS knockdown were confirmed histologically post hoc. Knockdown of nNOS expression in the PVN resulted in a sustained increase in blood pressure (from 95 ± 2 to 104 ± 3 mmHg, P < 0.05), with robust concurrent sustained activation of renal SNA (>70%, P < 0.05). The study reveals a major role for nNOS-derived nitric oxide within the PVN in chronic set-point regulation of cardiovascular autonomic activity in the conscious, normotensive rat.


Asunto(s)
Presión Sanguínea/fisiología , Riñón/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Núcleo Hipotalámico Paraventricular/enzimología , Sistema Nervioso Simpático/metabolismo , Animales , Masculino , Neuronas/metabolismo , Óxido Nítrico Sintasa de Tipo I/genética , ARN Interferente Pequeño , Ratas , Ratas Wistar
14.
J Huazhong Univ Sci Technolog Med Sci ; 37(1): 63-69, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28224417

RESUMEN

The identity of higher-order neurons and circuits playing an associative role to control renal function is not well understood. We identified specific neural populations of rostral elements of brain regions that project multisynaptically to the kidneys in 3-6 days after injecting a retrograde tracer pseudorabies virus (PRV)-614 into kidney of 13 adult male C57BL/6J strain mice. PRV-614 infected neurons were detected in a number of mesencephalic (e.g. central amygdala nucleus), telencephalic regions and motor cortex. These divisions included the preoptic area (POA), dorsomedial hypothalamus (DMH), lateral hypothalamus, arcuate nucleus (Arc), suprachiasmatic nucleus (SCN), periventricular hypothalamus (PeH), and rostral and caudal subdivision of the paraventricular nucleus of the hypothalamus (PVN). PRV-614/Tyrosine hydroxylase (TH) double-labeled cells were found within DMH, Arc, SCN, PeH, PVN, the anterodorsal and medial POA. A subset of neurons in PVN that participated in regulating sympathetic outflow to kidney was catecholaminergic or serotonergic. PRV-614 infected neurons within the PVN also contained arginine vasopressin or oxytocin. These data demonstrate the rostral elements of brain innervate the kidney by the neuroanatomical circuitry.


Asunto(s)
Encéfalo/virología , Herpesvirus Suido 1/fisiología , Riñón/inervación , Vías Nerviosas , Animales , Encéfalo/enzimología , Masculino , Mesencéfalo/enzimología , Mesencéfalo/virología , Ratones , Ratones Endogámicos C57BL , Vías Nerviosas/anatomía & histología , Vías Nerviosas/virología , Núcleo Hipotalámico Paraventricular/enzimología , Núcleo Hipotalámico Paraventricular/virología , Telencéfalo/enzimología , Telencéfalo/virología , Tirosina 3-Monooxigenasa/metabolismo
15.
Sci Rep ; 7: 43038, 2017 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-28225041

RESUMEN

The imbalance of neurotransmitters and excessive oxidative stress responses contribute to the pathogenesis of hypertension. In this study, we determined whether blockade of p44/42 MAPK pathway in the hypothalamic paraventricular nucleus (PVN) ameliorates the development of hypertension through modulating neurotransmitters and attenuating oxidative stress. Dahl salt-sensitive (S) 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 of PD-98059 (0.025 µg/h), a p44/42 MAPK inhibitor, or vehicle via osmotic minipump. HS resulted in higher mean arterial pressure (MAP) and Fra-like (Fra-LI) activity, and plasma and PVN levels of norepinephrine (NE), tyrosine hydroxylase (TH), NOX2 and NOX4, lower PVN levels of gamma-aminobutyric acid (GABA), copper/zinc superoxide dismutase (Cu/Zn-SOD) and the 67-kDa isoform of glutamate decarboxylase (GAD67), as compared with NS group. PD-98059 infusion reduced NE, TH, NOX2 and NOX4 in the PVN, and induced Cu/Zn-SOD and GAD67 in the PVN. It suggests that PVN blockade of p44/42 MAPK attenuates hypertension through modulating neurotransmitters and attenuating oxidative stress.


Asunto(s)
Hipertensión/inducido químicamente , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Neurotransmisores/metabolismo , Estrés Oxidativo , Núcleo Hipotalámico Paraventricular/enzimología , Cloruro de Sodio/metabolismo , Animales , Modelos Animales de Enfermedad , Núcleo Hipotalámico Paraventricular/fisiología , Ratas Endogámicas Dahl , Transducción de Señal
17.
Science ; 351(6279): 1293-6, 2016 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-26989246

RESUMEN

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.


Asunto(s)
Metabolismo Energético/fisiología , Conducta Alimentaria/fisiología , Hiperfagia/genética , N-Acetilglucosaminiltransferasas/fisiología , Núcleo Hipotalámico Paraventricular/fisiología , Acetilglucosamina/metabolismo , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Metabolismo Energético/genética , Eliminación de Gen , Homeostasis/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , N-Acetilglucosaminiltransferasas/genética , Neuronas/enzimología , Obesidad/genética , Núcleo Hipotalámico Paraventricular/citología , Núcleo Hipotalámico Paraventricular/enzimología , Procesamiento Proteico-Postraduccional , Respuesta de Saciedad/fisiología
18.
Cardiovasc Toxicol ; 16(4): 345-54, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26438340

RESUMEN

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.


Asunto(s)
Antihipertensivos/administración & dosificación , Presión Arterial/efectos de los fármacos , Caspasa 1/metabolismo , Hipertensión/prevención & control , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Pirrolidinas/administración & dosificación , Cloruro de Sodio Dietético , Tiocarbamatos/administración & dosificación , Factor de Transcripción ReIA/antagonistas & inhibidores , Animales , Antiinflamatorios/administración & dosificación , Antioxidantes/administración & dosificación , Biomarcadores/sangre , Modelos Animales de Enfermedad , Epinefrina/sangre , Hipertensión/enzimología , Hipertensión/fisiopatología , Quinasa I-kappa B/metabolismo , Infusiones Parenterales , Interleucina-1beta/metabolismo , Masculino , Norepinefrina/sangre , Estrés Oxidativo/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/enzimología , Núcleo Hipotalámico Paraventricular/fisiopatología , Fosforilación , Ratas Endogámicas Dahl , Transducción de Señal , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/fisiopatología , Factor de Transcripción ReIA/metabolismo
19.
J Comp Neurol ; 524(11): 2251-65, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-26659944

RESUMEN

At younger ages, women have a lower risk for hypertension than men, but this sexual dimorphism declines with the onset of menopause. These differences are paralleled in rodents following "slow-pressor" angiotensin II (AngII) administration: young male and aged female mice, but not young females, develop hypertension. There is also an established sexual dimorphism both in the cardiovascular response to the neurohypophyseal hormone arginine vasopressin (AVP) and in the expression of oxidative stress. We examined the relationship between AngII-mediated hypertension and the cellular distribution of the superoxide generating NADPH oxidase (NOX) in AVP-expressing hypothalamic paraventricular nucleus (PVN) neurons in "menopausal" female mice. Dual-labeling immunoelectron microscopy was used to determine whether the subcellular distribution of the organizer/adapter NOX p47(phox) subunit is altered in PVN dendrites following AngII administered (14 days) during the "postmenopausal" stage of accelerated ovarian failure (AOF) in young female mice treated with 4-vinylcyclohexene diepoxide. Slow-pressor AngII elevated blood pressure in AOF females and induced a significant increase in near plasmalemmal p47(phox) and a decrease in cytoplasmic p47(phox) in PVN AVP dendrites. These changes are the opposite of those observed in AngII-induced hypertensive male mice (Coleman et al. [2013] J. Neurosci. 33:4308-4316) and may be ascribed in part to baseline differences between young females and males in the near plasmalemmal p47(phox) on AVP dendrites seen in the present study. These findings highlight fundamental differences in the neural substrates of oxidative stress in the PVN associated with AngII hypertension in postmenopausal females compared with males. J. Comp. Neurol. 524:2251-2265, 2016. © 2015 Wiley Periodicals, Inc.


Asunto(s)
Hipertensión/metabolismo , NADPH Oxidasas/metabolismo , Núcleo Hipotalámico Paraventricular/enzimología , Posmenopausia/metabolismo , Caracteres Sexuales , Angiotensina II/toxicidad , Animales , Modelos Animales de Enfermedad , Femenino , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Inmunoelectrónica , Neuronas/enzimología
20.
Nature ; 525(7570): 519-22, 2015 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-26375004

RESUMEN

It is commonly assumed, but has rarely been demonstrated, that sex differences in behaviour arise from sexual dimorphism in the underlying neural circuits. Parental care is a complex stereotypic behaviour towards offspring that is shared by numerous species. Mice display profound sex differences in offspring-directed behaviours. At their first encounter, virgin females behave maternally towards alien pups while males will usually ignore the pups or attack them. Here we show that tyrosine hydroxylase (TH)-expressing neurons in the anteroventral periventricular nucleus (AVPV) of the mouse hypothalamus are more numerous in mothers than in virgin females and males, and govern parental behaviours in a sex-specific manner. In females, ablating the AVPV TH(+) neurons impairs maternal behaviour whereas optogenetic stimulation or increased TH expression in these cells enhance maternal care. In males, however, this same neuronal cluster has no effect on parental care but rather suppresses inter-male aggression. Furthermore, optogenetic activation or increased TH expression in the AVPV TH(+) neurons of female mice increases circulating oxytocin, whereas their ablation reduces oxytocin levels. Finally, we show that AVPV TH(+) neurons relay a monosynaptic input to oxytocin-expressing neurons in the paraventricular nucleus. Our findings uncover a previously unknown role for this neuronal population in the control of maternal care and oxytocin secretion, and provide evidence for a causal relationship between sexual dimorphism in the adult brain and sex differences in parental behaviour.


Asunto(s)
Hipotálamo/citología , Hipotálamo/fisiología , Conducta Materna/fisiología , Oxitocina/metabolismo , Caracteres Sexuales , Agresión , Animales , Núcleo Hipotalámico Anterior/citología , Núcleo Hipotalámico Anterior/enzimología , Núcleo Hipotalámico Anterior/fisiología , Neuronas Dopaminérgicas/enzimología , Neuronas Dopaminérgicas/metabolismo , Femenino , Hipotálamo/enzimología , Masculino , Ratones , Oxitocina/sangre , Núcleo Hipotalámico Paraventricular/citología , Núcleo Hipotalámico Paraventricular/enzimología , Núcleo Hipotalámico Paraventricular/fisiología , Periodo Posparto , Sinapsis/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA