Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 153
Filtrar
Más filtros

Medicinas Complementárias
Tipo del documento
Intervalo de año de publicación
1.
High Alt Med Biol ; 25(1): 77-88, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38241485

RESUMEN

Guo, Xinqi, Hongyu Ma, Ziye Cui, Qiyue Zhao, Ying Zhang, Lu Jia, Liping Zhang, Hui Guo, Xiangjian Zhang, Yi Zhang, Yue Guan, and Huijie Ma. Chronic intermittent hypobaric hypoxia reduces hypothalamic N-Methyl-d-Aspartate Receptor activity and sympathetic outflow in spontaneously hypertensive rats. High Alt Med Biol. 25:77-88, 2024. Objective: This study aims to determine the role of hypothalamic renin-angiotensin system (RAS) in the antihypertensive effect of chronic intermittent hypobaric hypoxia (CIHH). Methods: Wistar-Kyoto (WKY) rats and spontaneously hypertensive rats (SHRs) received 35 days of hypobaric hypoxia simulating an altitude of 4,000 m, 5 h/day. The levels of RAS, blood pressure, and N-methyl-d-aspartate receptor (NMDAR) activities of hypothalamic paraventricular nucleus (PVN) presympathetic neurons from each group of rats were determined. Results: The systolic blood pressure, diastolic blood pressure, and mean arterial blood pressure (MAP) of SHRs significantly decreased from the third week of CIHH treatment. This blood pressure reduction effect could be maintained for at least 2 weeks after stopping the CIHH treatment. CIHH treatment also attenuated the decrease in MAP and renal sympathetic nerve activity induced by hexamethonium administration in SHRs, but not in WKY rats. Furthermore, CIHH reversed the increase in serum angiotensin (Ang)II concentration and the expression of PVN angiotensin-converting enzyme (ACE) and AngII type 1 (AT1) receptors, as well as the decrease in serum Ang1-7 concentration and the expression of PVN ACE2 and Mas receptors in SHRs. In addition, the administration of CIHH resulted in a reduction in the frequency of miniature excitatory postsynaptic currents and amplitude of NMDAR current in PVN presympathetic neurons of SHRs, which means that CIHH decreased the pre- and postsynaptic NMDAR activity of PVN presympathetic neurons in SHRs. However, pretreatment with A779 (a Mas receptor blocker) or AngII abrogated the above effects. Meanwhile, Ang1-7 pretreatment mimicked the CIHH effect on pre- and postsynaptic NMDAR activity of presympathetic neurons in SHRs. Conclusions: Our data indicate that CIHH reduces pre- and postsynaptic NMDAR activity of PVN presympathetic neurons, sympathetic outflow, and blood pressure by decreasing the activity of the ACE/AngII/AT1 axis and increasing the activity of ACE2/Ang1-7/Mas axis in the hypothalamus in hypertension.


Asunto(s)
Hipertensión , Receptores de N-Metil-D-Aspartato , Ratas , Animales , Ratas Endogámicas SHR , Receptores de N-Metil-D-Aspartato/metabolismo , Ratas Endogámicas WKY , Enzima Convertidora de Angiotensina 2/metabolismo , Hipotálamo , Hipertensión/etiología , Hipertensión/terapia , Presión Sanguínea/fisiología , Sistema Nervioso Simpático/metabolismo , Angiotensinas/metabolismo , Angiotensinas/farmacología
2.
J Biol Chem ; 300(2): 105597, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38160798

RESUMEN

Increased expression of angiotensin II AT1A receptor (encoded by Agtr1a) and Na+-K+-Cl- cotransporter-1 (NKCC1, encoded by Slc12a2) in the hypothalamic paraventricular nucleus (PVN) contributes to hypertension development. However, little is known about their transcriptional control in the PVN in hypertension. DNA methylation is a critical epigenetic mechanism that regulates gene expression. Here, we determined whether transcriptional activation of Agtr1a and Slc12a2 results from altered DNA methylation in spontaneously hypertensive rats (SHR). Methylated DNA immunoprecipitation and bisulfite sequencing-PCR showed that CpG methylation at Agtr1a and Slc12a2 promoters in the PVN was progressively diminished in SHR compared with normotensive Wistar-Kyoto rats (WKY). Chromatin immunoprecipitation-quantitative PCR revealed that enrichment of DNA methyltransferases (DNMT1 and DNMT3A) and methyl-CpG binding protein 2, a DNA methylation reader protein, at Agtr1a and Slc12a2 promoters in the PVN was profoundly reduced in SHR compared with WKY. By contrast, the abundance of ten-eleven translocation enzymes (TET1-3) at Agtr1a and Slc12a2 promoters in the PVN was much greater in SHR than in WKY. Furthermore, microinjecting of RG108, a selective DNMT inhibitor, into the PVN of WKY increased arterial blood pressure and correspondingly potentiated Agtr1a and Slc12a2 mRNA levels in the PVN. Conversely, microinjection of C35, a specific TET inhibitor, into the PVN of SHR markedly reduced arterial blood pressure, accompanied by a decrease in Agtr1a and Slc12a2 mRNA levels in the PVN. Collectively, our findings suggest that DNA hypomethylation resulting from the DNMT/TET switch at gene promoters in the PVN promotes transcription of Agtr1a and Slc12a2 and hypertension development.


Asunto(s)
Desmetilación del ADN , Hipotálamo , Receptor de Angiotensina Tipo 1 , Miembro 2 de la Familia de Transportadores de Soluto 12 , Animales , Ratas , Presión Sanguínea , ADN/metabolismo , Hipertensión/metabolismo , Hipotálamo/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptor de Angiotensina Tipo 1/metabolismo , ARN Mensajero/genética , Sistema Nervioso Simpático/metabolismo , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo
3.
Neuropharmacology ; 230: 109506, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36924924

RESUMEN

Hyperactivity of presympathetic neurons in the hypothalamic paraventricular nucleus (PVN) plays a key role in generating excess sympathetic output in hypertension. However, the mechanisms driving hyperactivity of PVN presympathetic neurons in hypertension are unclear. In this study, we determined the role of corticotropin-releasing factor (CRF) in the PVN in augmented glutamatergic input, neuronal excitability and sympathetic outflow in hypertension. The number of CRF or c-Fos immunoreactive neurons and CRF/c-Fos double-labeled neurons in the PVN was significantly greater in spontaneously hypertensive rats (SHRs) than in normotensive Wistar-Kyoto (WKY) rats. Blocking glutamatergic input reduced the CRF-potentiated excitability of spinally projecting PVN neurons. Furthermore, CRF knockdown via Crispr/Cas9 in the PVN decreased the frequencies of spontaneous firing and miniature excitatory postsynaptic currents (mEPSCs) in spinally projecting PVN neurons in SHRs. In addition, the mRNA and protein levels of CRFR1, but not CRFR2, in the PVN were significantly higher in SHRs than in WKY rats. Blocking CRFR1 with NBI-35965, but not blocking CRFR2 with Antisauvagine-30, reduced the frequencies of spontaneous firing and mEPSCs of spinally projecting PVN neurons in SHRs. Also, microinjection of NBI-35965 into the PVN significantly reduced arterial blood pressure (ABP) and renal sympathetic nerve activity (RSNA) in anesthetized SHRs, but not in WKY rats. However, microinjection of Antisauvagine-30 into the PVN had no effect on ABP or RSNA in WKY rats and SHRs. Our findings suggest that endogenous CRF in the PVN potentiates glutamatergic input and firing activity of PVN presympathetic neurons via CRFR1, resulting in augmented sympathetic outflow in hypertension.


Asunto(s)
Hormona Liberadora de Corticotropina , Hipertensión , Ratas , Animales , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Hormona Liberadora de Corticotropina/metabolismo , Hipotálamo/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Hipertensión/metabolismo , Neuronas/metabolismo , Sistema Nervioso Simpático/metabolismo
4.
Neurosci Lett ; 799: 137130, 2023 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-36792026

RESUMEN

Brown adipose tissue (BAT) activity is controlled by the sympathetic nervous system. Activation of BAT has shown significant promise in preclinical studies to elicit weight loss. Since the hypothalamic paraventricular nucleus (PVN) contributes to the regulation of BAT thermogenic activity, we sought to determine the effects of electrical stimulation of the PVN as a model of deep brain stimulation (DBS) for increasing BAT sympathetic nerve activity (SNA). The rostral raphe pallidus area (rRPa) was also chosen as a target for DBS since it contains the sympathetic premotor neurons for BAT. Electrical stimulation (100 µA, 100 µs, 100 Hz, for 5 min at a 50 % duty cycle) of the PVN increased BAT SNA and BAT thermogenesis. These effects were prevented by a local nanoinjection of bicuculline, a GABAA receptor antagonist. We suggest that electrical stimulation of the PVN elicited local release of GABA, which inhibited BAT sympathoinhibitory neurons in PVN, thereby releasing a restraint on BAT SNA. Electrical stimulation of the rRPa inhibited BAT thermogenesis and this was prevented by a local nanoinjection of bicuculline, suggesting that local release of GABA suppressed BAT SNA. Electrical stimulation of the PVN activates BAT metabolism via a mechanism that may include activation of local GABAA receptors. These findings contribute to our understanding of the mechanisms underlying the effects of DBS in the regulation of fat metabolism and provide a foundation for further DBS studies targeting hypothalamic circuits regulating BAT thermogenesis as a therapy for obesity.


Asunto(s)
Estimulación Encefálica Profunda , Núcleo Hipotalámico Paraventricular , Ratas , Animales , Ratas Sprague-Dawley , Bicuculina/farmacología , Tejido Adiposo Pardo/inervación , Termogénesis , Hipotálamo , Ácido gamma-Aminobutírico/metabolismo , Sistema Nervioso Simpático/metabolismo
6.
J Healthc Eng ; 2022: 4441603, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35432831

RESUMEN

Sympathetic nerve remodeling after myocardial infarction (MI) has an indispensable role in cardiac remodeling. Numerous works have shown that sympathetic nerve remodeling can be delayed by inhibition of inflammatory response. Earlier studies have shown improvement in ventricular remodeling and inhibited chronic stage neural remodeling by Yiqi Huoxue decoction (YQHX). Therefore, the current study looked at the inhibitory effect of YQHX prescription on proinflammatory mediators and macrophages and the effect on neural remodeling at 3 and 7 days after MI. YQHX inhibited the expression of Toll-like receptor 4 (TLR4) and nuclear factor kappa B (NF-κB) proteins and macrophage infiltration within 7 days after myocardial infarction. YQHX could decrease Th-positive nerve fiber density in the area around infarction and reduce the expression of growth-associated protein 43 (GAP43), nerve growth factor (NGF), and tyrosine hydroxylase (TH) proteins, which was associated with the remodeling of sympathetic nerves. Thus, the nerve remodeling inhibition after MI due to YQHX may be through its anti-inflammatory action. These data provide direct evidence for the potential application of traditional Chinese medicine (TCM) in the remodeling of sympathetic nerves after MI.


Asunto(s)
Corazón , Infarto del Miocardio , Animales , Humanos , Macrófagos/metabolismo , Infarto del Miocardio/tratamiento farmacológico , Ratas , Sistema Nervioso Simpático/metabolismo , Remodelación Ventricular
7.
Cell Death Dis ; 12(4): 362, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33824276

RESUMEN

Adhesion G protein-coupled receptor A1 (ADGRA1, also known as GPR123) belongs to the G protein-coupled receptors (GPCRs) family and is well conserved in the vertebrate lineage. However, the structure of ADGRA1 is unique and its physiological function remains unknown. Previous studies have shown that Adgra1 is predominantly expressed in the central nervous system (CNS), indicating its important role in the transduction of neural signals. The aim of this study is to investigate the central function of Adgra1 in vivo and clarify its physiological significance by establishing an Adgra1-deficient mouse (Adgra1-/-) model. The results show that Adgra1-/- male mice exhibit decreased body weight with normal food intake and locomotion, shrinkage of body mass, increased lipolysis, and hypermetabolic activity. Meanwhile, mutant male mice present elevated core temperature coupled with resistance to hypothermia upon cold stimulus. Further studies show that tyrosine hydroxylase (TH) and ß3-adrenergic receptor (ß3-AR), indicators of sympathetic nerve excitability, are activated as well as their downstream molecules including uncoupling protein 1 (UCP1), coactivator 1 alpha (PGC1-α) in brown adipose tissue (BAT), and hormone-sensitive lipase (HSL) in white adipose tissue (WAT). In addition, mutant male mice have higher levels of serum T3, T4, accompanied by increased mRNAs of hypothalamus-pituitary-thyroid axis. Finally, Adgra1-/- male mice present abnormal activation of PI3K/AKT/GSK3ß and MEK/ERK pathways in hypothalamus. Overexpression of ADGRA1 in Neuro2A cell line appears to suppress these two signaling pathways. In contrast, Adgra1-/- female mice show comparable body weight along with normal metabolic process to their sex-matched controls. Collectively, ADGRA1 is a negative regulator of sympathetic nervous system (SNS) and hypothalamus-pituitary-thyroid axis by regulating PI3K/AKT/GSK3ß and MEK/ERK pathways in hypothalamus of male mice, suggesting an important role of ADGRA1 in maintaining metabolic homeostasis including energy expenditure and thermogenic balance.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Hipotálamo/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Termogénesis/fisiología , Tejido Adiposo Pardo/metabolismo , Animales , Metabolismo Energético/fisiología , Masculino , Ratones , Obesidad/metabolismo , Transducción de Señal/fisiología , Sistema Nervioso Simpático/metabolismo , Glándula Tiroides/metabolismo
8.
Endocrinology ; 162(4)2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33460433

RESUMEN

Body energy homeostasis results from balancing energy intake and energy expenditure. Central nervous system administration of pituitary adenylate cyclase activating polypeptide (PACAP) dramatically alters metabolic function, but the physiologic mechanism of this neuropeptide remains poorly defined. PACAP is expressed in the mediobasal hypothalamus (MBH), a brain area essential for energy balance. Ventromedial hypothalamic nucleus (VMN) neurons contain, by far, the largest and most dense population of PACAP in the medial hypothalamus. This region is involved in coordinating the sympathetic nervous system in response to metabolic cues in order to re-establish energy homeostasis. Additionally, the metabolic cue of leptin signaling in the VMN regulates PACAP expression. We hypothesized that PACAP may play a role in the various effector systems of energy homeostasis, and tested its role by using VMN-directed, but MBH encompassing, adeno-associated virus (AAVCre) injections to ablate Adcyap1 (gene coding for PACAP) in mice (Adcyap1MBHKO mice). Adcyap1MBHKO mice rapidly gained body weight and adiposity, becoming hyperinsulinemic and hyperglycemic. Adcyap1MBHKO mice exhibited decreased oxygen consumption (VO2), without changes in activity. These effects appear to be due at least in part to brown adipose tissue (BAT) dysfunction, and we show that PACAP-expressing cells in the MBH can stimulate BAT thermogenesis. While we observed disruption of glucose clearance during hyperinsulinemic/euglycemic clamp studies in obese Adcyap1MBHKO mice, these parameters were normal prior to the onset of obesity. Thus, MBH PACAP plays important roles in the regulation of metabolic rate and energy balance through multiple effector systems on multiple time scales, which highlight the diverse set of functions for PACAP in overall energy homeostasis.


Asunto(s)
Hipotálamo/metabolismo , Obesidad/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Tejido Adiposo Pardo , Animales , Peso Corporal , Metabolismo Energético , Femenino , Humanos , Leptina/metabolismo , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Obesidad/genética , Obesidad/fisiopatología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Sistema Nervioso Simpático/metabolismo , Termogénesis , Núcleo Hipotalámico Ventromedial/metabolismo
9.
Am J Physiol Endocrinol Metab ; 320(2): E270-E280, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33166186

RESUMEN

The G-protein subunits Gqα and G11α (Gq/11α) couple receptors to phospholipase C, leading to increased intracellular calcium. In this study we investigated the consequences of Gq/11α deficiency in the dorsomedial hypothalamus (DMH), a critical site for the control of energy homeostasis. Mice with DMH-specific deletion of Gq/11α (DMHGq/11KO) were generated by stereotaxic injection of adeno-associated virus (AAV)-Cre-green fluorescent protein (GFP) into the DMH of Gqαflox/flox:G11α-/- mice. Compared with control mice that received DMH injection of AAV-GFP, DMHGq/11KO mice developed obesity associated with reduced energy expenditure without significant changes in food intake or physical activity. DMHGq/11KO mice showed no defects in the ability of the melanocortin agonist melanotan II to acutely stimulate energy expenditure or to inhibit food intake. At room temperature (22°C), DMHGq/11KO mice showed reduced sympathetic nervous system activity in brown adipose tissue (BAT) and heart, accompanied with decreased basal BAT uncoupling protein 1 (Ucp1) gene expression and lower heart rates. These mice were cold intolerant when acutely exposed to cold (6°C for 5 h) and had decreased cold-stimulated BAT Ucp1 gene expression. DMHGq/11KO mice also failed to adapt to gradually declining ambient temperatures and to develop adipocyte browning in inguinal white adipose tissue although their BAT Ucp1 was proportionally stimulated. Consistent with impaired cold-induced thermogenesis, the onset of obesity in DMHGq/11KO mice was significantly delayed when housed under thermoneutral conditions (30°C). Thus our results show that Gqα and G11α in the DMH are required for the control of energy homeostasis by stimulating energy expenditure and thermoregulation.NEW & NOTEWORTHY This paper demonstrates that signaling within the dorsomedial hypothalamus via the G proteins Gqα and G11α, which couple cell surface receptors to the stimulation of phospholipase C, is critical for regulation of energy expenditure, thermoregulation by brown adipose tissue and the induction of white adipose tissue browning.


Asunto(s)
Enfermedades del Sistema Nervioso Autónomo/genética , Metabolismo Energético/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Hipotálamo/metabolismo , Obesidad/genética , Animales , Enfermedades del Sistema Nervioso Autónomo/metabolismo , Enfermedades del Sistema Nervioso Autónomo/fisiopatología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/deficiencia , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Obesidad/fisiopatología , Especificidad de Órganos/genética , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/fisiopatología
10.
J Cardiovasc Pharmacol ; 76(5): 602-609, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32868626

RESUMEN

Heart failure (HF) affects around 100 million people and is a staggering burden for health care system worldwide. Rapid and sustained activation of inflammatory response is an important feature of HF after myocardial infarction. Sympathetic overactivation is also an important factor in the occurrence and progression of HF. The beneficial effect of renal denervation (RDN) has been demonstrated in HF. In the current study, we hypothesized that RDN improves cardiac function in HF canine models due to acute myocardial infarction (AMI) and reduced inflammation might be involved. Twenty-four beagles were randomized into the control (n = 8), HF (n = 8), and HF + RDN group (n = 8). The HF model after AMI was established by embolization the anterior descending distal artery with anhydrous ethanol in the HF and HF + RDN group. Bilateral renal artery ablation was performed in the HF + RDN group. Cardiac function, serum creatine kinase, creatine kinase-MB and NT-Pro BNP level, and expression of inflammation-related proteins in myocardial were examined. Because the paraventricular nucleus of the hypothalamus might be involved in inflammation-induced central neural excitation in HF and plays an important role in regulating extracellular fluid volume and sympathetic activity, expression of inflammation-related proteins in hypothalamus was also examined. AMI and post-AMI HF model was created successfully. Compared with the HF group, dogs in the HF + RDN group showed better cardiac function 4 weeks after AMI: lower left ventricular end-diastolic pressure, left ventricular end-diastolic dimension, and left ventricular end-systolic dimension and higher LEVF and left ventricular systolic pressure (P < 0.05 for all) were observed in the HF + RDN group. In addition, dogs in the HF + RDN group had slightly less ventricular fibrosis. Interestingly, RDN had lower expression of inflammation-related proteins including interleukin-6, tumor necrosis factors-α, nuclear factor κB, and monocyte chemotactic protein 1 (P < 0.05 for all) in both myocardial tissue and hypothalamus. RDN can improve cardiac function in dogs with HF after myocardial infarction. Our results suggested that RDN might affect cytokine-induced central neural excitation in HF and later affect sympathetic activity. Our results suggested a potential beneficial mechanism of RDN independent of mechanism involving renal afferent and efferent sympathetic nerves.


Asunto(s)
Ablación por Catéter , Insuficiencia Cardíaca/cirugía , Hipotálamo/metabolismo , Mediadores de Inflamación/metabolismo , Riñón/irrigación sanguínea , Infarto del Miocardio/complicaciones , Miocardio/metabolismo , Arteria Renal/inervación , Simpatectomía , Sistema Nervioso Simpático/cirugía , Función Ventricular Izquierda , Animales , Modelos Animales de Enfermedad , Perros , Femenino , Fibrosis , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Hipotálamo/fisiopatología , Masculino , Miocardio/patología , Volumen Sistólico , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/fisiopatología , Presión Ventricular , Remodelación Ventricular
11.
JCI Insight ; 5(11)2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32493842

RESUMEN

The sympathetic nervous system plays an important role in the occurrence of ventricular tachycardia (VT). Many patients, however, experience VT despite maximal doses of beta blocker therapy, possibly due to the effects of sympathetic cotransmitters such as neuropeptide Y (NPY). The purpose of this study was to determine, in a porcine model, whether propranolol at doses higher than clinically recommended could block ventricular electrophysiological effects of sympathoexcitation via stellate ganglia stimulation, and if any residual effects are mediated by NPY. Greater release of cardiac NPY was observed at higher sympathetic stimulation frequencies (10 and 20 vs. 4 Hz). Despite treatment with even higher doses of propranolol (1.0 mg/kg), electrophysiological effects of sympathetic stimulation remained, with residual shortening of activation recovery interval (ARI), a surrogate of action potential duration (APD). Adjuvant treatment with the NPY Y1 receptor antagonist BIBO 3304, however, reduced these electrophysiological effects while augmenting inotropy. These data demonstrate that high-dose beta blocker therapy is insufficient to block electrophysiological effects of sympathoexcitation, and a portion of these electrical effects in vivo are mediated by NPY. Y1 receptor blockade may represent a promising adjuvant therapy to beta-adrenergic receptor blockade.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Antagonistas Adrenérgicos beta/farmacología , Arginina/análogos & derivados , Neuropéptido Y/metabolismo , Sistema Nervioso Simpático/metabolismo , Taquicardia Ventricular , Animales , Arginina/farmacología , Modelos Animales de Enfermedad , Receptores de Neuropéptido Y/antagonistas & inhibidores , Receptores de Neuropéptido Y/metabolismo , Sus scrofa , Sistema Nervioso Simpático/patología , Taquicardia Ventricular/tratamiento farmacológico , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/patología
12.
Biol Sex Differ ; 11(1): 10, 2020 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-32160920

RESUMEN

Obesity increases sympathetic nerve activity (SNA) in men, but not women. Here, we review current evidence suggesting that sexually dimorphic sympathoexcitatory responses to leptin and insulin may contribute. More specifically, while insulin increases SNA similarly in lean males and females, this response is markedly amplified in obese males, but is abolished in obese females. In lean female rats, leptin increases a subset of sympathetic nerves only during the high estrogen proestrus reproductive phase; thus, in obese females, because reproductive cycling can become impaired, the sporadic nature of leptin-induced sympathoexcitaton could minimize its action, despite elevated leptin levels. In contrast, in males, obesity preserves or enhances the central sympathoexcitatory response to leptin, and current evidence favors leptin's contribution to the well-established increases in SNA induced by obesity in men. Leptin and insulin increase SNA via receptor binding in the hypothalamic arcuate nucleus and a neuropathway that includes arcuate neuropeptide Y (NPY) and proopiomelanocortin (POMC) projections to the paraventricular nucleus. These metabolic hormones normally suppress sympathoinhibitory NPY neurons and activate sympathoexcitatory POMC neurons. However, obesity appears to alter the ongoing activity and responsiveness of arcuate NPY and POMC neurons in a sexually dimorphic way, such that SNA increases in males but not females. We propose hypotheses to explain these sex differences and suggest areas of future research.


Asunto(s)
Hipotálamo/metabolismo , Insulina/metabolismo , Leptina/metabolismo , Obesidad/metabolismo , Caracteres Sexuales , Sistema Nervioso Simpático/metabolismo , Animales , Femenino , Humanos , Insulina/fisiología , Masculino , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Proopiomelanocortina/metabolismo
13.
Acta Physiol (Oxf) ; 228(3): e13401, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31599481

RESUMEN

AIM: Serotonin (5-hydroxytryptamine, 5-HT), an important neurotransmitter and hormone, modulates many physiological functions including body temperature. We investigated neural mechanisms involved in the inhibition of brown adipose tissue (BAT) sympathetic nerve activity (SNA) and BAT thermogenesis evoked by 5-HT. METHODS: Electrophysiological recordings, intravenous (iv) injections and nanoinjections in the brains of anaesthetized rats. RESULTS: Cooling-evoked increases in BAT SNA were inhibited by the intra-rostral raphé pallidus (rRPa) and the iv administration of the 5-HT1A receptor agonist, 8-OH-DPAT or 5-HT. The intra-rRPa 5-HT, the intra-rRPa and the iv 8-OH-DPAT, but not the iv 5-HT-induced inhibition of BAT SNA were prevented by nanoinjection of a 5-HT1A receptor antagonist in the rRPa. The increase in BAT SNA evoked by nanoinjection of NMDA in the rRPa was not inhibited by iv 5-HT, indicating that iv 5-HT does not inhibit BAT SNA by acting in the rRPa or in the sympathetic pathway distal to the rRPa. In contrast, under a warm condition, blockade of 5HT1A receptors in the rRPa increased BAT SNA and BAT thermogenesis, suggesting that endogenous 5-HT in the rRPa contributes to the suppression of BAT SNA and BAT thermogenesis. The increases in BAT SNA and BAT thermogenesis evoked by nanoinjection of NMDA in the dorsomedial hypothalamus (DMH) were inhibited by iv 5-HT, but those following bicuculline nanoinjection in the DMH were not inhibited. CONCLUSIONS: The systemic 5-HT-induced inhibition of BAT SNA requires a GABAergic inhibition of BAT sympathoexcitatory neurones in the DMH. In addition, during warming, 5-HT released endogenously in rRPa inhibits BAT SNA.


Asunto(s)
Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/inervación , Hipotálamo/efectos de los fármacos , Neuronas/efectos de los fármacos , Receptor de Serotonina 5-HT1A/metabolismo , Serotonina/farmacología , Sistema Nervioso Simpático/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo , Tejido Adiposo Pardo/metabolismo , Animales , Hipotálamo/metabolismo , Masculino , Neuronas/metabolismo , Núcleo Pálido del Rafe/efectos de los fármacos , Núcleo Pálido del Rafe/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Serotonina 5-HT1A/química , Agonistas de Receptores de Serotonina/farmacología , Sistema Nervioso Simpático/metabolismo , Termogénesis
14.
Pak J Pharm Sci ; 33(5(Supplementary)): 2341-2345, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33832909

RESUMEN

Acacia jacquemontii Benth. is used traditionally to treat hypertension but no scientific literature supports this claim. So, this study was aimed at validating this claim. This was done by injecting various doses of crude extract of Acacia jacquemontii, AJC (5, 10, 20, 30mg/kg) and all fractions (hexane, ethyl acetate, n-butanol and aqueous) (3, 5, 10, 20mg/kg) intravenously in anaesthetized rat. Based on the results, butanol fraction (AJB) at 20mg/kg was found to be the most potent, so it was selected for exploring mechanisms of action. For this purpose, different groups were injected with various pharmacological inhibitors (L-NAME, atropine, captopril, propranolol and hexamethonium) prior to AJB administration. Also, AJB at 20mg/kg was evaluated for prolonged hypotensive effect for the period of 40 min. Results showed a significant dose dependent reduction in BP in normotensive and in hypertensive rats. AJC and AJB produced a decline in SBP, DBP and MAP with p<0.05 - p<0.001 and p<0.001 respectively in normotensive animals. Whereas in hypertensive animals, AJC showed significant reduction at 5mg/kg with p<0.01 and at 10, 20 and 30 mg/kg with p<0.001. AJB produced a decline in hypertensive animals at all tested doses with p<0.001. AJB resulted in hypotensive effect mediated by ß receptors, ganglionic block operating central sympathetic neural responses and renin angiotensin aldosterone system (RAAS). This study supports the ethnomedicinal claim of Acacia jacquemontii Benth. in treating hypertension.


Asunto(s)
Acacia , Antihipertensivos/farmacología , Presión Sanguínea/efectos de los fármacos , Hipertensión/tratamiento farmacológico , Extractos Vegetales/farmacología , Acacia/química , Animales , Antihipertensivos/aislamiento & purificación , Modelos Animales de Enfermedad , Etnofarmacología , Fructosa , Ganglios Autónomos/efectos de los fármacos , Ganglios Autónomos/fisiopatología , Hipertensión/inducido químicamente , Hipertensión/metabolismo , Hipertensión/fisiopatología , Extractos Vegetales/aislamiento & purificación , Ratas Sprague-Dawley , Receptores Adrenérgicos beta/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/fisiopatología
15.
J Lipid Res ; 60(7): 1212-1224, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31126973

RESUMEN

The sympathetic nervous system (SNS) regulates the functions of white adipose tissue (WAT) and brown adipose tissue (BAT) tightly. Carotid baroreceptor stimulation (CBS) efficiently inhibits SNS activation. We hypothesized that CBS would protect against obesity. We administered CBS to obese rats and measured sympathetic and AMP-activated protein kinase (AMPK)/ PPAR pathway responses as well as changes in perirenal WAT (PWAT), epididymal WAT (EWAT), and interscapular BAT (IBAT). CBS alleviated obesity-related metabolic changes, improving insulin resistance; reducing adipocyte hypertrophy, body weight, and adipose tissue weights; and decreasing norepinephrine but increasing acetylcholine in plasma, PWAT, EWAT, and IBAT. CBS also downregulated fatty acid translocase (CD36), fatty acid transport protein (FATP), phosphorylated and total hormone sensitive lipase, phosphorylated and total protein kinase A, and PPARγ in obese rats. Simultaneously, CBS upregulated phosphorylated adipose triglyceride lipase, phosphorylated and total AMPK, and PPARα in PWAT, EWAT, and IBAT. However, BAT and WAT responses differed; although many responses were more sensitive in IBAT, responses of CD36, FATP, and PPARγ were more sensitive in PWAT and EWAT. Overall, CBS decreased chronically activated SNS and ameliorated obesity-related metabolic disorders by regulating the AMPK/PPARα/γ pathway.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Obesidad/metabolismo , Presorreceptores/metabolismo , Animales , Seno Carotídeo/inervación , Terapia por Estimulación Eléctrica/métodos , Prueba de Tolerancia a la Glucosa , Masculino , Ratas , Ratas Sprague-Dawley , Sistema Nervioso Simpático/metabolismo
16.
Mol Metab ; 25: 142-153, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31014927

RESUMEN

OBJECTIVE: Gsα couples multiple receptors, including the melanocortin 4 receptor (MC4R), to intracellular cAMP generation. Germline inactivating Gsα mutations lead to obesity in humans and mice. Mice with brain-specific Gsα deficiency also develop obesity with reduced energy expenditure and locomotor activity, and impaired adaptive thermogenesis, but the underlying mechanisms remain unclear. METHODS: We created mice (DMHGsKO) with Gsα deficiency limited to the dorsomedial hypothalamus (DMH) and examined the effects on energy balance and thermogenesis. RESULTS: DMHGsKO mice developed severe, early-onset obesity associated with hyperphagia and reduced energy expenditure and locomotor activity, along with impaired brown adipose tissue thermogenesis. Studies in mice with loss of MC4R in the DMH suggest that defective DMH MC4R/Gsα signaling contributes to abnormal energy balance but not to abnormal locomotor activity or cold-induced thermogenesis. Instead, DMHGsKO mice had impaired leptin signaling along with increased expression of the leptin signaling inhibitor protein tyrosine phosphatase 1B in the DMH, which likely contributes to the observed hyperphagia and reductions in energy expenditure, locomotor activity, and cold-induced thermogenesis. CONCLUSIONS: DMH Gsα signaling is critical for energy balance, thermogenesis, and leptin signaling. This study provides insight into how distinct signaling pathways can interact to regulate energy homeostasis and temperature regulation.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Hiperfagia/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Obesidad/metabolismo , Transducción de Señal/fisiología , Termogénesis/fisiología , Tejido Adiposo Pardo/metabolismo , Animales , Metabolismo Energético/fisiología , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad/genética , Glucosa/metabolismo , Homeostasis/fisiología , Masculino , Ratones , Ratones Noqueados , Obesidad/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , Sistema Nervioso Simpático/metabolismo
17.
Cancer Discov ; 9(6): 702-710, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30944117

RESUMEN

Nerves are emerging regulators of cancer progression. Cancer cells induce the outgrowth of nerves in the tumor microenvironment through the release of neurotrophic factors, and in return nerves liberate neurotransmitters that activate cancer growth and dissemination. Although sympathetic nerves drive tumor angiogenesis via the liberation of noradrenaline, sensory and parasympathetic nerves stimulate cancer stem cells. Interestingly, recent evidence indicates that parasympathetic nerves can eventually inhibit tumor progression, suggesting a yin-yang type of regulation of cancer by nerves. From a broader perspective, the question of a higher level of control of cancer development by the central nervous system should be raised. SIGNIFICANCE: Nerves are emerging regulators of cancer initiation, progression, and metastasis. Here, we review the evidence to date and explore the basic and clinical ramifications of these findings.


Asunto(s)
Transformación Celular Neoplásica , Susceptibilidad a Enfermedades , Neoplasias/etiología , Neoplasias/patología , Sistema Nervioso/metabolismo , Microambiente Tumoral , Animales , Axones , Comunicación Celular , Humanos , Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Neovascularización Patológica/metabolismo , Sistema Nervioso/patología , Neurogénesis , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/patología
18.
J Clin Invest ; 129(6): 2305-2317, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30888335

RESUMEN

Chronic alcohol consumption causes liver injury, inflammation and fibrosis, thereby increasing morbidity and mortality. Paradoxically, modest drinking is believed to confer metabolic improvement, but the underlying mechanism remains elusive. Here, we have identified a novel hepatoprotective brain/brown adipose tissue (BAT)/liver axis. Alcohol consumption or direct alcohol administration into the brain stimulated hypothalamic neural circuits and sympathetic nerves innervating BAT, and dramatically increased BAT uncoupling protein 1 (Ucp1) expression and activity in a BAT sympathetic nerve-dependent manner. BAT and beige fat oxidized fatty acids to fuel Ucp1-mediated thermogenesis, thereby inhibiting lipid trafficking into the liver. BAT also secreted several adipokines, including adiponectin that suppressed hepatocyte injury and death. Genetic deletion of Ucp1 profoundly augmented alcohol-induced liver steatosis, injury, inflammation and fibrosis in male and female mice. Conversely, activation of BAT and beige fat through cold exposure suppressed alcoholic liver disease development. Our results unravel an unrecognized brain alcohol-sensing/sympathetic nerve/BAT/liver axis that counteracts liver steatosis and injury.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Etanol/efectos adversos , Hígado Graso Alcohólico/metabolismo , Hígado/metabolismo , Tejido Adiposo Beige/metabolismo , Tejido Adiposo Beige/patología , Tejido Adiposo Pardo/patología , Animales , Frío , Etanol/farmacología , Hígado Graso Alcohólico/genética , Hígado Graso Alcohólico/patología , Femenino , Hipotálamo/metabolismo , Hipotálamo/patología , Hígado/patología , Masculino , Ratones , Ratones Noqueados , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/patología , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
19.
Brain Res ; 1712: 82-92, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30735639

RESUMEN

Aerobic exercise lowers blood pressure in patients with hypertension, but the underlying mechanisms remain incompletely understood. The hypothalamic paraventricular nucleus (PVN) plays a key role in the control of sympathetic outflow and cardiovascular tone. We examined whether chronic aerobic exercise altered synaptic transmission and reactive oxygen species (ROS) production in the PVN. In the present study, spontaneously hypertensive rats (SHRs) were subjected to exercise training for 8 weeks, five times per week, with Wistar Kyoto (WKY) rats as the cohort control. Miniature excitatory and inhibitory postsynaptic currents (mEPSCs and mIPSCs) were recorded from the PVN in ex vivo hypothalamic slice preparations obtained after the last training, and biomarkers of oxidative stress and physical indexes were observed. The mean frequency and amplitude, as well as the rise time and the decay time constant of mIPSCs, significantly decreased in 20-wk-old SHRs compared to WKY 20-wk-old controls. In contrast to mIPSCs, only the mean mEPSC frequency was higher, and there were no other changes in mEPSCs in comparison to the control group. SHRs exhibited higher ROS, 8-OHdG, and MDA; and lower SOD1, SOD2, CAT, Ogg1, and SOD and CAT activity in the PVN. These SHRs also had a significant increase in heart rate, blood pressure and sympathetic nerve activity, and higher levels of norepinephrine (NE). Exercise training ameliorated all these abnormalities, resulting in an increase in the mean frequency, amplitude and kinetics of mIPSCs, accompanied by a decrease in the mean frequency of mEPSCs in the PVN. This study demonstrates that moderate intensity, high frequency exercise training induces a selective enhancement of inhibitory synaptic transmission in the PVN, which may dampen sympathetic activity and reduce blood pressure in hypertension. These changes may be due to antioxidant-related adaptations in the PVNs of SHRs.


Asunto(s)
Núcleo Hipotalámico Paraventricular/metabolismo , Condicionamiento Físico Animal/fisiología , Transmisión Sináptica/fisiología , Animales , Presión Sanguínea , Potenciales Postsinápticos Excitadores/fisiología , Frecuencia Cardíaca , Hipertensión/fisiopatología , Hipotálamo/metabolismo , Masculino , Neuronas/metabolismo , Condicionamiento Físico Animal/métodos , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Especies Reactivas de Oxígeno/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sistema Nervioso Simpático/metabolismo
20.
Brain Res ; 1708: 69-77, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30529283

RESUMEN

Our previous studies have shown that electroacupuncture (EA) at the Jianshi-Neiguan acupoints (P5-6, overlying the median nerve) attenuates sympathoexcitatory responses through its influence on neuronal activity in the rostral ventrolateral medulla (rVLM). The nucleus tractus solitarii (NTS) receives input from somatic nerve stimulation. Connections between the NTS and the rVLM during EA stimulation have not been investigated and thus were the focus of the present study. Seven to ten days after unilateral microinjection of a rhodamine-conjugated microsphere retrograde tracer (100 nl) into the rVLM, rats were subjected to EA or sham-EA without electrical stimulation. EA was performed for 30 min at the P5-6 acupoints bilaterally. Perikarya containing the microsphere tracer were found in the NTS of both groups. Compared to controls (needle placement without electrical stimulation, n = 7), c-Fos immunoreactivity and neurons double-labeled with c-Fos, an immediate early gene, and the tracer were significantly increased in the NTS of EA-treated rats (all P < 0.05; n = 8), particularly, in the medial and lateral subdivisions of NTS at subpostremal and obex levels. These results suggest that EA at the P5-6 acupoints activates NTS neurons. Furthermore, EA-activated NTS neurons directly project to the rVLM and likely influence the rVLM activity.


Asunto(s)
Electroacupuntura/métodos , Bulbo Raquídeo/fisiología , Núcleo Solitario/fisiología , Puntos de Acupuntura , Terapia por Acupuntura/métodos , Animales , Presión Sanguínea/fisiología , Estimulación Eléctrica , Masculino , Nervio Mediano/metabolismo , Vías Nerviosas/fisiología , Neuronas/metabolismo , Sustancia Gris Periacueductal/fisiología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley , Sistema Nervioso Simpático/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA