Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 208
Filter
1.
Cardiovasc Toxicol ; 24(9): 904-917, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39008239

ABSTRACT

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.


Subject(s)
AMP-Activated Protein Kinases , Disease Models, Animal , Enzyme Activation , Hypertension, Renovascular , Mitogen-Activated Protein Kinase 3 , Paraventricular Hypothalamic Nucleus , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1 , Animals , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/enzymology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/physiopathology , Hypertension, Renovascular/physiopathology , Hypertension, Renovascular/enzymology , Hypertension, Renovascular/metabolism , Hypertension, Renovascular/drug therapy , Male , AMP-Activated Protein Kinases/metabolism , Phosphorylation , Receptor, Angiotensin, Type 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Transcription Factor RelA/metabolism , Ribonucleotides/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Sympathetic Nervous System/physiopathology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , NF-kappa B/metabolism , Signal Transduction , Antihypertensive Agents/pharmacology , Rats
2.
Am J Hypertens ; 37(9): 682-691, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-38782571

ABSTRACT

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.


Subject(s)
Blood Pressure , Hypertension , Janus Kinase 2 , Paraventricular Hypothalamic Nucleus , Rats, Inbred SHR , Rats, Inbred WKY , STAT3 Transcription Factor , Signal Transduction , Animals , Janus Kinase 2/metabolism , STAT3 Transcription Factor/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/enzymology , Paraventricular Hypothalamic Nucleus/drug effects , Hypertension/metabolism , Hypertension/physiopathology , Male , Disease Models, Animal , Oxidative Stress/drug effects , Norepinephrine/metabolism , Rats , Tyrphostins/pharmacology , Phosphorylation
3.
Thyroid ; 32(1): 105-114, 2022 01.
Article in English | MEDLINE | ID: mdl-34726513

ABSTRACT

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.


Subject(s)
Paraventricular Hypothalamic Nucleus/enzymology , Thyroid Hormones/metabolism , Thyrotropin-Releasing Hormone/metabolism , Animals , Disease Models, Animal , Mice , Paraventricular Hypothalamic Nucleus/physiopathology , Statistics, Nonparametric
4.
Cardiovasc Toxicol ; 21(12): 1045-1057, 2021 12.
Article in English | MEDLINE | ID: mdl-34537923

ABSTRACT

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.


Subject(s)
Antihypertensive Agents/pharmacology , Arterial Pressure/radiation effects , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Oxidative Stress/drug effects , Paraventricular Hypothalamic Nucleus/drug effects , Prehypertension/prevention & control , Reactive Oxygen Species/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Disease Models, Animal , Male , Paraventricular Hypothalamic Nucleus/enzymology , Paraventricular Hypothalamic Nucleus/physiopathology , Phosphorylation , Prehypertension/enzymology , Prehypertension/etiology , Prehypertension/physiopathology , Rats, Inbred Dahl , Signal Transduction , Sodium Chloride, Dietary , Xanthophylls/pharmacology
5.
Cardiovasc Toxicol ; 21(9): 721-736, 2021 09.
Article in English | MEDLINE | ID: mdl-34076830

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antihypertensive Agents/pharmacology , Antioxidants/pharmacology , Apigenin/pharmacology , Cardiomegaly/drug therapy , Cytokines/metabolism , Hypertension/drug therapy , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , Paraventricular Hypothalamic Nucleus/drug effects , Reactive Oxygen Species/metabolism , Animals , Arterial Pressure/drug effects , Cardiomegaly/enzymology , Cardiomegaly/physiopathology , Disease Models, Animal , Fibrosis , Hypertension/enzymology , Hypertension/physiopathology , Male , Myocardium/metabolism , Myocardium/pathology , NADPH Oxidases/genetics , Paraventricular Hypothalamic Nucleus/enzymology , Paraventricular Hypothalamic Nucleus/physiopathology , Rats, Inbred SHR , Rats, Inbred WKY , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
6.
Am J Physiol Heart Circ Physiol ; 318(1): H34-H48, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31675258

ABSTRACT

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.


Subject(s)
Apoptosis , Arterial Pressure , Caspase 3/metabolism , Hypertension/prevention & control , Hypoxia/complications , Paraventricular Hypothalamic Nucleus/enzymology , Preoptic Area/enzymology , Animals , Caspase 3/genetics , Circadian Rhythm , Disease Models, Animal , Heart Rate , Hypertension/enzymology , Hypertension/pathology , Hypertension/physiopathology , Hypoxia/enzymology , Hypoxia/pathology , Hypoxia/physiopathology , Male , Oxidative Stress , Paraventricular Hypothalamic Nucleus/pathology , Paraventricular Hypothalamic Nucleus/physiopathology , Preoptic Area/pathology , Preoptic Area/physiopathology , Proto-Oncogene Proteins c-fos/metabolism , Rats, Sprague-Dawley , Signal Transduction
7.
Acta Histochem ; 121(6): 690-694, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31227177

ABSTRACT

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.


Subject(s)
Ependymoglial Cells/enzymology , Leptin/pharmacology , NADPH Dehydrogenase/metabolism , Paraventricular Hypothalamic Nucleus/enzymology , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/enzymology , Ependymoglial Cells/cytology , Male , Neurons/cytology , Neurons/enzymology , Nitric Oxide/metabolism , Paraventricular Hypothalamic Nucleus/cytology , Rats , Rats, Wistar , Receptors, Leptin/metabolism
8.
Am J Physiol Heart Circ Physiol ; 316(6): H1389-H1405, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30925093

ABSTRACT

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.


Subject(s)
Blood Pressure , Calcium Signaling , Hypertension/prevention & control , Neurons/enzymology , Paraventricular Hypothalamic Nucleus/enzymology , Proton-Translocating ATPases/deficiency , Receptor, Angiotensin, Type 1/metabolism , Receptors, Cell Surface/deficiency , Animals , Autonomic Nervous System/metabolism , Autonomic Nervous System/physiopathology , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Desoxycorticosterone Acetate , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Knockdown Techniques , Hypertension/enzymology , Hypertension/genetics , Hypertension/physiopathology , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Paraventricular Hypothalamic Nucleus/physiopathology , Phosphorylation , Proton-Translocating ATPases/genetics , Receptor, Angiotensin, Type 1/genetics , Receptors, Cell Surface/genetics , Prorenin Receptor
9.
J Cardiol ; 73(1): 81-88, 2019 01.
Article in English | MEDLINE | ID: mdl-30487059

ABSTRACT

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.


Subject(s)
Arrhythmias, Cardiac/enzymology , Myocardial Infarction/enzymology , Paraventricular Hypothalamic Nucleus/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Sympathetic Nervous System/enzymology , Animals , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/prevention & control , Chromones/therapeutic use , Enzyme Inhibitors/therapeutic use , Male , Morpholines/therapeutic use , Myocardial Infarction/drug therapy , Myocardial Infarction/physiopathology , Myocardium/enzymology , Norepinephrine/blood , Rats , Rats, Inbred SHR , Signal Transduction/drug effects
10.
Psychopharmacology (Berl) ; 236(4): 1293-1301, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30539267

ABSTRACT

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.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Nitric Oxide Synthase Type I/metabolism , Penile Erection/drug effects , Preoptic Area/drug effects , Sexual Behavior, Animal/drug effects , Animals , Enzyme Activation/drug effects , Enzyme Activation/physiology , Female , Male , Neurons/drug effects , Neurons/enzymology , Nitric Oxide , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/enzymology , Penile Erection/physiology , Preoptic Area/enzymology , Rats , Rats, Long-Evans , Sexual Behavior, Animal/physiology , Testis/drug effects , Testis/enzymology
11.
Hypertension ; 72(3): 667-675, 2018 09.
Article in English | MEDLINE | ID: mdl-30012866

ABSTRACT

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.


Subject(s)
Denervation/methods , Heart Failure/physiopathology , Kidney/innervation , Nitric Oxide Synthase Type I/metabolism , Paraventricular Hypothalamic Nucleus/physiopathology , Sympathetic Nervous System/physiopathology , Afferent Pathways/drug effects , Afferent Pathways/physiology , Animals , Capsaicin/pharmacology , Chronic Disease , Enzyme Inhibitors/pharmacology , Male , Nitric Oxide Synthase Type I/antagonists & inhibitors , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/enzymology , Rats, Sprague-Dawley , Sensory System Agents/pharmacology , Sympathetic Nervous System/drug effects , omega-N-Methylarginine/pharmacology
12.
Exp Physiol ; 103(3): 419-428, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29215757

ABSTRACT

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.


Subject(s)
Blood Pressure/physiology , Kidney/metabolism , Nitric Oxide Synthase Type I/metabolism , Paraventricular Hypothalamic Nucleus/enzymology , Sympathetic Nervous System/metabolism , Animals , Male , Neurons/metabolism , Nitric Oxide Synthase Type I/genetics , RNA, Small Interfering , Rats , Rats, Wistar
13.
J Huazhong Univ Sci Technolog Med Sci ; 37(1): 63-69, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28224417

ABSTRACT

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.


Subject(s)
Brain/virology , Herpesvirus 1, Suid/physiology , Kidney/innervation , Neural Pathways , Animals , Brain/enzymology , Male , Mesencephalon/enzymology , Mesencephalon/virology , Mice , Mice, Inbred C57BL , Neural Pathways/anatomy & histology , Neural Pathways/virology , Paraventricular Hypothalamic Nucleus/enzymology , Paraventricular Hypothalamic Nucleus/virology , Telencephalon/enzymology , Telencephalon/virology , Tyrosine 3-Monooxygenase/metabolism
14.
Sci Rep ; 7: 43038, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28225041

ABSTRACT

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.


Subject(s)
Hypertension/chemically induced , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Neurotransmitter Agents/metabolism , Oxidative Stress , Paraventricular Hypothalamic Nucleus/enzymology , Sodium Chloride/metabolism , Animals , Disease Models, Animal , Paraventricular Hypothalamic Nucleus/physiology , Rats, Inbred Dahl , Signal Transduction
16.
Science ; 351(6279): 1293-6, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26989246

ABSTRACT

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.


Subject(s)
Energy Metabolism/physiology , Feeding Behavior/physiology , Hyperphagia/genetics , N-Acetylglucosaminyltransferases/physiology , Paraventricular Hypothalamic Nucleus/physiology , Acetylglucosamine/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Energy Metabolism/genetics , Gene Deletion , Homeostasis/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , N-Acetylglucosaminyltransferases/genetics , Neurons/enzymology , Obesity/genetics , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/enzymology , Protein Processing, Post-Translational , Satiety Response/physiology
17.
Cardiovasc Toxicol ; 16(4): 345-54, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26438340

ABSTRACT

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.


Subject(s)
Antihypertensive Agents/administration & dosage , Arterial Pressure/drug effects , Caspase 1/metabolism , Hypertension/prevention & control , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Pyrrolidines/administration & dosage , Sodium Chloride, Dietary , Thiocarbamates/administration & dosage , Transcription Factor RelA/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/administration & dosage , Antioxidants/administration & dosage , Biomarkers/blood , Disease Models, Animal , Epinephrine/blood , Hypertension/enzymology , Hypertension/physiopathology , I-kappa B Kinase/metabolism , Infusions, Parenteral , Interleukin-1beta/metabolism , Male , Norepinephrine/blood , Oxidative Stress/drug effects , Paraventricular Hypothalamic Nucleus/enzymology , Paraventricular Hypothalamic Nucleus/physiopathology , Phosphorylation , Rats, Inbred Dahl , Signal Transduction , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/physiopathology , Transcription Factor RelA/metabolism
18.
J Comp Neurol ; 524(11): 2251-65, 2016 08 01.
Article in English | MEDLINE | ID: mdl-26659944

ABSTRACT

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.


Subject(s)
Hypertension/metabolism , NADPH Oxidases/metabolism , Paraventricular Hypothalamic Nucleus/enzymology , Postmenopause/metabolism , Sex Characteristics , Angiotensin II/toxicity , Animals , Disease Models, Animal , Female , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Microscopy, Immunoelectron , Neurons/enzymology
19.
J Neuroendocrinol ; 27(11): 835-49, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26403275

ABSTRACT

An accumulating body of evidence suggests that the activity of the mineralocorticoid, aldosterone, in the brain via the mineralocorticoid receptor (MR) plays an important role in the regulation of blood pressure. MR was recently found in vasopressin and oxytocin synthesising magnocellular neurosecretory cells (MNCs) in both the paraventricular (PVN) and supraoptic (SON) nuclei in the hypothalamus. Considering the physiological effects of these hormones, MR in these neurones may be an important site mediating the action of aldosterone in blood pressure regulation within the brain. However, aldosterone activation of MR in the hypothalamus remains controversial as a result of the high binding affinity of glucocorticoids to MR at substantially higher concentrations compared to aldosterone. In aldosterone-sensitive epithelia, the enzyme 11ß-hydroxysteroid dehydrogenase type 2 (11ß-HSD2) prevents glucocorticoids from binding to MR by converting glucocorticoids into inactive metabolites. The present study aimed to determine whether 11ß-HSD2, which increases aldosterone selectivity, is expressed in MNCs. Specific 11ß-HSD2 immunoreactivity was found in the cytoplasm of the MNCs in both the SON and PVN. In addition, double-fluorescence confocal microscopy demonstrated that MR-immunoreactivity and 11ß-HSD2-in situ hybridised products are colocalised in MNCs. Lastly, single-cell reverse transcriptase-polymerase chain reaction detected MR and 11ß-HSD2 mRNAs from cDNA libraries derived from single identified MNCs. These findings strongly suggest that MNCs in the SON and PVN are aldosterone-sensitive neurones.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Neurosecretory Systems/cytology , Neurosecretory Systems/metabolism , Paraventricular Hypothalamic Nucleus/cytology , Receptors, Mineralocorticoid/metabolism , Supraoptic Nucleus/cytology , Animals , Male , Neurosecretory Systems/enzymology , Paraventricular Hypothalamic Nucleus/enzymology , Paraventricular Hypothalamic Nucleus/metabolism , Rats , Supraoptic Nucleus/enzymology , Supraoptic Nucleus/metabolism
20.
Nature ; 525(7570): 519-22, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26375004

ABSTRACT

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.


Subject(s)
Hypothalamus/cytology , Hypothalamus/physiology , Maternal Behavior/physiology , Oxytocin/metabolism , Sex Characteristics , Aggression , Animals , Anterior Hypothalamic Nucleus/cytology , Anterior Hypothalamic Nucleus/enzymology , Anterior Hypothalamic Nucleus/physiology , Dopaminergic Neurons/enzymology , Dopaminergic Neurons/metabolism , Female , Hypothalamus/enzymology , Male , Mice , Oxytocin/blood , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/enzymology , Paraventricular Hypothalamic Nucleus/physiology , Postpartum Period , Synapses/metabolism , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL