Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Curr Hypertens Rep ; 25(7): 107-116, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37058193

RESUMEN

PURPOSE OF REVIEW: The response to natural stressors involves both cardiac stimulation and vascular changes, primarily triggered by increases in sympathetic activity. These effects lead to immediate flow redistribution that provides metabolic support to priority target organs combined with other key physiological responses and cognitive strategies, against stressor challenges. This extremely well-orchestrated response that was developed over millions of years of evolution is presently being challenged, over a short period of time. In this short review, we discuss the neurogenic background for the origin of emotional stress-induced hypertension, focusing on sympathetic pathways from related findings in humans and animals. RECENT FINDINGS: The urban environment offers a variety of psychological stressors. Real or anticipatory, emotional stressors may increase baseline sympathetic activity. From routine day-to-day traffic stress to job-related anxiety, chronic or abnormal increases in sympathetic activity caused by emotional stressors can lead to cardiovascular events, including cardiac arrhythmias, increases in blood pressure and even sudden death. Among the various alterations proposed, chronic stress could modify neuroglial circuits or compromise antioxidant systems that may alter the responsiveness of neurons to stressful stimuli. These phenomena lead to increases in sympathetic activity, hypertension and consequent cardiovascular diseases. The link between anxiety, emotional stress, and hypertension may result from an altered neuronal firing rate in central pathways controlling sympathetic activity. The participation of neuroglial and oxidative mechanisms in altered neuronal function is primarily involved in enhanced sympathetic outflow. The significance of the insular cortex-dorsomedial hypothalamic pathway in the evolution of enhanced overall sympathetic outflow is discussed.


Asunto(s)
Hipertensión , Distrés Psicológico , Animales , Humanos , Hipertensión/etiología , Corazón , Presión Sanguínea/fisiología , Hipotálamo , Sistema Nervioso Simpático
3.
Neural Plast ; 2017: 2361675, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28845307

RESUMEN

Accumulated evidence indicates that obesity-induced type 2 diabetes (T2D) is associated with enhanced sympathetic activation. The present study was conducted to investigate the role for leptin-glutamate signaling within the hypothalamus in regulating sympathetic nerve activity. In anesthetized rats, microinjections of leptin (5 ng ~ 100 ng) into the arcuate nucleus (ARCN) and paraventricular nucleus (PVN) induced increases in renal sympathetic nerve activity (RSNA), blood pressure (BP), and heart rate (HR). Prior microinjections of NMDA receptor antagonist AP5 (16 pmol) into the ARCN or PVN reduced leptin-induced increases in RSNA, BP, and HR in both ARCN and PVN. Knockdown of a leptin receptor with siRNA inhibited NMDA-induced increases in RSNA, BP, and HR in the ARCN but not in the PVN. Confocal calcium imaging in the neuronal NG108 and astrocytic C6 cells demonstrated that preincubation with leptin induced an increase in intracellular calcium green fluorescence when the cells were challenged with glutamate. In high-fat diet and low-dose streptozotocin-induced T2D rats, we found that leptin receptor and NMDA NR1 receptor expressions in the ARCN and PVN were significantly increased. In conclusion, these studies provide evidence that within the hypothalamic nuclei, leptin-glutamate signaling regulates the sympathetic activation. This may contribute to the sympathoexcitation commonly observed in obesity-related T2D.


Asunto(s)
Ácido Glutámico/metabolismo , Hipotálamo/efectos de los fármacos , Riñón/inervación , Leptina/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Sistema Nervioso Simpático/efectos de los fármacos , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Diabetes Mellitus Experimental/metabolismo , Antagonistas de Aminoácidos Excitadores/farmacología , Técnicas de Silenciamiento del Gen , Frecuencia Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/fisiología , Hipotálamo/metabolismo , Leptina/metabolismo , Masculino , Microinyecciones , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Sistema Nervioso Simpático/metabolismo , Valina/análogos & derivados , Valina/farmacología
4.
Am J Physiol Regul Integr Comp Physiol ; 310(7): R612-8, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26843582

RESUMEN

Chronic heart failure (CHF) is characterized by autonomic dysfunction combined with baroreflex attenuation. The hypotensive and bradycardic responses produced by electrical stimulation of the aortic depressor nerve (ADN) were examined in conscious CHF and control male Wistar rats (12-13 wk old). Furthermore, the role of parasympathetic and sympathetic nervous system in mediating the cardiovascular responses to baroreflex activation was evaluated by selective ß1-adrenergic and muscarinic receptor antagonists. CHF was induced by myocardial infarction. After 6 wk, the subjects were implanted with electrodes for ADN stimulation. Twenty-four hours later, electrical stimulation of the ADN was applied for 20 s using five different frequencies (5, 15, 30, 60, and 90 Hz), while the arterial pressure was recorded by a catheter implanted into the femoral artery. Electrical stimulation of the ADN elicited progressive and similar hypotensive and bradycardic responses in control (n = 12) and CHF (n = 11) rats, while the hypotensive response was not affected by methylatropine. Nevertheless, the reflex bradycardia was attenuated by methylatropine in control, but not in CHF rats. Atenolol did not affect the hypotensive or bradycardic response in either group. The ADN function was examined under anesthesia through electroneurographic recordings. The arterial pressure-ADN activity relationship was attenuated in CHF rats. In conclusion, despite the attenuation of baroreceptor function in CHF rats, the electrical stimulation of the ADN elicited a stimulus-dependent hypotension and bradycardia of similar magnitude as observed in control rats. Therefore, electrical activation of the aortic baroreflex overcomes both the attenuation of parasympathetic function and the sympathetic overdrive.


Asunto(s)
Aorta/inervación , Barorreflejo , Presión Sanguínea , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/terapia , Estimulación Eléctrica Transcutánea del Nervio/métodos , Animales , Terapia por Estimulación Eléctrica/métodos , Insuficiencia Cardíaca/diagnóstico , Frecuencia Cardíaca , Masculino , Ratas , Ratas Wistar , Resultado del Tratamiento
5.
Neuron ; 78(6): 1036-49, 2013 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-23791197

RESUMEN

Although communication between neurons is considered a function of the synapse, neurons also release neurotransmitter from their dendrites. We found that dendritic transmitter release coordinates activity across distinct neuronal populations to generate integrative homeostatic responses. We show that activity-dependent vasopressin release from hypothalamic neuroendocrine neurons in the paraventricular nucleus stimulates neighboring (~100 µm soma-to-soma) presympathetic neurons, resulting in a sympathoexcitatory population response. This interpopulation crosstalk was engaged by an NMDA-mediated increase in dendritic Ca(2+), influenced by vasopressin's ability to diffuse in the extracellular space, and involved activation of CAN channels at the target neurons. Furthermore, we demonstrate that this interpopulation crosstalk plays a pivotal role in the generation of a systemic, polymodal neurohumoral response to a hyperosmotic challenge. Because dendritic release is emerging as a widespread process, our results suggest that a similar mechanism could mediate interpopulation crosstalk in other brain systems, particularly those involved in generating complex behaviors.


Asunto(s)
Dendritas/metabolismo , Hipotálamo/metabolismo , Red Nerviosa/metabolismo , Neuropéptidos/metabolismo , Neurosecreción/fisiología , Animales , Dendritas/química , Hipotálamo/química , Masculino , Red Nerviosa/química , Técnicas de Cultivo de Órganos , Ratas , Ratas Transgénicas , Ratas Wistar
6.
Am J Physiol Regul Integr Comp Physiol ; 305(1): R50-9, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23616108

RESUMEN

The cardiac sympathetic afferent reflex (CSAR) is induced by stimulating the cardiac sympathetic afferents, which evokes increases in sympathetic outflow and arterial pressure. In the present study, we attempted to identify the contribution of thalamic and hypothalamic nuclei involved in the CSAR. First, we observed that there was an increase in the number of c-Fos-labeled cells in the paraventricular nucleus (PVN) (190 ± 18 vs. 101 ± 15; P < 0.05), the paraventricular nucleus of the thalamus (PVT) (239 ± 23 vs. 151 ± 15; P < 0.05), and the mediodorsal thalamic nucleus (MD) (92 ± 9 vs. 63 ± 6; P < 0.05) following epicardial application of bradykinin (BK) compared with the control group (P < 0.05). Second, using extracellular single-unit recording, we found 25% of spontaneously active neurons in the thalamus were stimulated by epicardial application of BK or capsaicin in intact rats. However, 24% of spontaneously active neurons in the thalamus were still stimulated by epicardial application of BK or capsaicin despite vagotomy and sinoaortic denervation. None of the neurons in the thalamus responded to baroreflex changes in arterial pressure, induced by intravenous injection of phenylephrine or sodium nitroprusside. The CSAR was inhibited by microinjection of muscimol or lidocaine into the PVN. However, it was not inhibited or blocked by microinjection of muscimol or lidocaine into the thalamus. Taken together, these data suggest that the thalamus, while activated, is not critical for autonomic adjustments in response to activation of the CSAR. On the other hand, the PVN is critically involved in the central pathway of the CSAR.


Asunto(s)
Barorreflejo/fisiología , Presión Sanguínea/fisiología , Sistema Cardiovascular/inervación , Neuronas Aferentes/fisiología , Núcleo Hipotalámico Paraventricular/fisiología , Sistema Nervioso Simpático/fisiología , Tálamo/fisiología , Animales , Barorreflejo/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Bradiquinina/farmacología , Capsaicina/farmacología , Sistema Cardiovascular/efectos de los fármacos , Masculino , Modelos Animales , Neuronas Aferentes/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley , Fármacos del Sistema Sensorial/farmacología , Sistema Nervioso Simpático/efectos de los fármacos , Vasodilatadores/farmacología
7.
Am J Physiol Cell Physiol ; 302(8): C1162-73, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22218225

RESUMEN

We have previously observed an increased of angiotensin II (ANG II) type 1 receptor (AT(1)R) with enhanced AT(1)R-mediated sympathetic outflow and concomitant downregulation of neuronal nitric oxide (NO) synthase (nNOS) with reduced NO-mediated inhibition from the paraventricular nucleus (PVN) in rats with heart failure. To test the hypothesis that NO exerts an inhibitory effect on AT(1)R expression in the PVN, we used primary cultured hypothalamic cells of neonatal rats and neuronal cell line NG108-15 as in vitro models. In hypothalamic primary culture, NO donor sodium nitroprusside (SNP) induced dose-dependent decreases in mRNA and protein of AT(1)R (10(-5) M SNP, AT(1)R protein was 10 ± 2% of control level) while NOS inhibitor N(G)-monomethyl-l-arginine (l-NMMA) induced dose-dependent increases in mRNA and protein levels of AT(1)R (10(-5) M l-NMMA, AT(1)R protein was 148 ± 8% of control level). Similar effects of SNP and l-NMMA on AT(1)R expression were also observed in NG108-15 cell line (10(-6) M SNP, AT(1)R protein was 30 ± 4% of control level while at the dose of 10(-6) M l-NMMA, AT(1)R protein was 171 ± 15% of the control level). Specific inhibition of nNOS, using antisense, caused an increase in AT(1)R expression while overexpression of nNOS, using adenoviral gene transfer (Ad.nNOS), caused an inhibition of AT(1)R expression in NG108 cells. Antisense nNOS transfection augmented the increase while Ad.nNOS infection blunted the increase in intracellular calcium concentration in response to ANG II treatment in NG108 cells. In addition, downregulation of AT(1)R mRNA as well as protein level in neuronal cell line in response to S-nitroso-N-acetyl pencillamine (SNAP) treatment was blocked by protein kinase G (PKG) inhibitor, while the peroxynitrite scavenger deforxamine had no effect. These results suggest that NO acts as an inhibitory regulator of AT(1)R expression and the activation of PKG is the required step in the regulation of AT(1)R gene expression via cGMP-dependent signaling pathway.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Óxido Nítrico/farmacología , Receptor de Angiotensina Tipo 1/biosíntesis , Adenoviridae/genética , Angiotensina II/farmacología , Animales , Calcio/metabolismo , Línea Celular Tumoral , Células Cultivadas , GMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , ADN sin Sentido/genética , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Óxido Nítrico/farmacocinética , Donantes de Óxido Nítrico/farmacocinética , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo I/metabolismo , Nitroprusiato/farmacología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , S-Nitroso-N-Acetilpenicilamina/farmacología , Transfección/métodos , omega-N-Metilarginina/farmacología
8.
Brain Res ; 964(1): 128-35, 2003 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-12573521

RESUMEN

Diabetes is characterized by hyperphagia, polydypsia and activation of the HPA axis. However, the mechanisms by which diabetes produces these effects are not clear. This study was conducted to examine the effects of diabetes on the neuroendocrine system and to see if treatment with insulin and/or leptin is capable of reversing these effects. Streptozotocin-induced diabetic adult male rats were subjected to the following treatments: vehicle, insulin (2 U/day, s.c.), leptin (100 microg/kg BW) or leptin+insulin every day for 2 weeks. Food intake, water intake, and body weight were monitored daily. We measured changes in monoamine concentrations in discrete nuclei of the hypothalamus at the end of treatment. Diabetes produced a marked increase in food intake and water intake and this effect was completely reversed by insulin treatment and partially reversed by leptin treatment (P<0.05). Diabetes caused an increase in norepinephrine (NE) concentrations in the paraventricular nucleus with a concurrent increase in serum corticosterone. Treatment with insulin and leptin completely reversed these effects. Induction of diabetes also increased the concentrations of NE, dopamine and serotonin in the arcuate nucleus and NE concentrations in the lateral hypothalamus, ventromedial hypothalamus (VMH) and suprachiasmatic nucleus (P<0.05). Although insulin treatment was capable of reversing all these changes, leptin treatment was unable to decrease diabetes-induced increase in NE concentrations in the VMH. These data provide evidence that hypothalamic monoamines could mediate the neuroendocrine effects of diabetes and that insulin and leptin act as important signals in this process.


Asunto(s)
Monoaminas Biogénicas/metabolismo , Diabetes Mellitus Experimental/complicaciones , Hiperfagia/tratamiento farmacológico , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Insulina/farmacología , Leptina/farmacología , Animales , Glucemia/efectos de los fármacos , Glucemia/fisiología , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Corticosterona/sangre , Ingestión de Líquidos/efectos de los fármacos , Ingestión de Líquidos/fisiología , Interacciones Farmacológicas/fisiología , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Hiperfagia/etiología , Hiperfagia/fisiopatología , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipotálamo-Hipofisario/fisiopatología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Insulina/metabolismo , Leptina/sangre , Masculino , Norepinefrina/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
9.
Brain Res ; 956(2): 268-75, 2002 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-12445695

RESUMEN

We sought to identify the areas that have altered neuronal activity within the hypothalamus of diabetic rats by mapping neuronal expression of c-fos protein (Fos) and Fos-related antigens. After a standard PAP immunocytochemical protocol, Fos-like immunoreactivity was observed in the paraventricular nucleus (PVN), supraoptic nucleus (SON), median preoptic area (MnPO), anterior hypothalamus (AH) and posterior hypothalamus (PH) of control (vehicle; n=6) and diabetic rats (Sprague-Dawley rats injected with STZ 65 mg/kg/ip 4 weeks prior to the experiment; n=6). Blood glucose levels were significantly elevated in the diabetic group (370+/-8 mg/dl) compared to control group (104+/-3 mg/dl). Diabetic rats had a significantly higher number of Fos-positive cells in PVN (2.5x), SON (7x) and MnPO (2x) compared to the control rats. However, diabetic rats had significantly fewer Fos-positive cells in the AH (0.3x) and no difference was observed in the PH between the diabetic and control rats. Despite the elevated number of Fos-positive cells in the diabetic rats, dehydration (water withdrawal for 24 h) or hypertonic challenge (1.5 ml of 0.1 M NaCl i.p. injection) produced a further increase in the number of Fos-positive cells in the PVN, SON and MnPO. Dehydration did not alter the number of Fos-positive cells in the AH or PH, but hypertonic challenge produced a significant increase in the Fos-positive cells in both the AH and PH of diabetic rats. This study demonstrates that: (1) there is increased basal neuronal activity in the PVN, SON and MnPO, a decrease in neuronal activity in the AH and no change in neuronal activity in the PH as indicated by Fos staining in diabetic rats; and (2) dehydration or hypertonic challenge produces a further increase in the number of Fos-positive cells in the PVN, SON, and MnPO which is comparable to control rats. These data support the conclusion that vasopressin producing neurons in the PVN and SON and autonomic areas within the lamina terminalis and hypothalamus are activated during diabetes and may contribute to the elevated levels of vasopressin and autonomic dysfunction during diabetes.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Hipotálamo/metabolismo , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Animales , Glucemia/metabolismo , Deshidratación/metabolismo , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/inducido químicamente , Hipotálamo/citología , Inmunohistoquímica , Masculino , Prosencéfalo/metabolismo , Ratas , Ratas Sprague-Dawley , Solución Salina Hipertónica
10.
Brain Res ; 955(1-2): 153-60, 2002 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12419531

RESUMEN

It is well known that nitric oxide (NO), within the paraventricular nucleus (PVN) of the hypothalamus, mediates sympatho-inhibition via an inhibitory GABA-ergic mechanism. Furthermore, the inhibitory GABA-ergic mechanism is impaired in the spontaneously hypertensive rat (SHR). These data suggest that the NO system, within the PVN, may also be impaired in the SHR. In addition, previous studies have documented that daily exercise attenuates the development of tachycardia, hypertension and blood pressure related cardiovascular disease risk factors in SHR. These data suggest that daily exercise enhances the inhibitory GABA-ergic and/or NO systems. Therefore, this study was designed to test the hypothesis that hypertension, in the SHR, is associated with a lower number of NADPH-diaphorase (a commonly used marker for neuronal NOS activity) positive neurons within the PVN and that daily exercise increases the number of NOS positive neurons. Using a standard histochemical protocol, NOS positive neurons were measured in the PVN, supraoptic nucleus, median preoptic area, lateral hypothalamus, nucleus of the tractus solitarius and rostral ventrolateral medulla. Results document that SHR have significantly fewer NOS-positive neurons in the PVN than their genetic control, the Wistar-Kyoto (WKY) rats (110+/-11 versus 139+/-17). Furthermore, daily exercise increased the number of NOS positive neurons in the SHR to levels seen in the WKY rats. These data demonstrate that hypertension, in the SHR, is associated with a lower number of NOS positive neurons within the PVN and that daily exercise increases the number of NOS positive neurons within the PVN.


Asunto(s)
Dihidrolipoamida Deshidrogenasa/metabolismo , Hipertensión/enzimología , Hipotálamo/enzimología , Neuronas/enzimología , Óxido Nítrico Sintasa/metabolismo , Carrera/fisiología , Animales , Recuento de Células/métodos , Dihidrolipoamida Deshidrogenasa/análisis , Hipertensión/fisiopatología , Hipotálamo/química , Masculino , Neuronas/química , Óxido Nítrico Sintasa/análisis , Fotoperiodo , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA