Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.458
Filtrar
1.
J Hypertens ; 42(6): 1027-1038, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38690904

RESUMEN

OBJECTIVE: Reno-renal reflexes are disturbed in cardiovascular and hypertensive conditions when elevated levels of pro-inflammatory mediators/cytokines are present within the kidney. We hypothesised that exogenously administered inflammatory cytokines tumour necrosis factor alpha (TNF-α) and interleukin (IL)-1ß modulate the renal sympatho-excitatory response to chemical stimulation of renal pelvic sensory nerves. METHODS: In anaesthetised rats, intrarenal pelvic infusions of vehicle [0.9% sodium chloride (NaCl)], TNF-α (500 and 1000 ng/kg) and IL-1ß (1000 ng/kg) were maintained for 30 min before chemical activation of renal pelvic sensory receptors was performed using randomized intrarenal pelvic infusions of hypertonic NaCl, potassium chloride (KCl), bradykinin, adenosine and capsaicin. RESULTS: The increase in renal sympathetic nerve activity (RSNA) in response to intrarenal pelvic hypertonic NaCl was enhanced during intrapelvic TNF-α (1000 ng/kg) and IL-1ß infusions by almost 800% above vehicle with minimal changes in mean arterial pressure (MAP) and heart rate (HR). Similarly, the RSNA response to intrarenal pelvic adenosine in the presence of TNF-α (500 ng/kg), but not IL-1ß, was almost 200% above vehicle but neither MAP nor HR were changed. There was a blunted sympatho-excitatory response to intrapelvic bradykinin in the presence of TNF-α (1000 ng/kg), but not IL-1ß, by almost 80% below vehicle, again without effect on either MAP or HR. CONCLUSION: The renal sympatho-excitatory response to renal pelvic chemoreceptor stimulation is modulated by exogenous TNF-α and IL-1ß. This suggests that inflammatory mediators within the kidney can play a significant role in modulating the renal afferent nerve-mediated sympatho-excitatory response.


Asunto(s)
Interleucina-1beta , Riñón , Sistema Nervioso Simpático , Factor de Necrosis Tumoral alfa , Animales , Interleucina-1beta/farmacología , Ratas , Riñón/inervación , Riñón/efectos de los fármacos , Masculino , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiología , Ratas Sprague-Dawley , Frecuencia Cardíaca/efectos de los fármacos , Bradiquinina/farmacología , Reflejo/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Adenosina/administración & dosificación , Adenosina/farmacología , Solución Salina Hipertónica/administración & dosificación , Solución Salina Hipertónica/farmacología
2.
Physiol Behav ; 281: 114575, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38692384

RESUMEN

Fibromyalgia (FM) is characterized by chronic widespread musculoskeletal pain accompanied by fatigue and muscle atrophy. Although its etiology is not known, studies have shown that FM patients exhibit altered function of the sympathetic nervous system (SNS), which regulates nociception and muscle plasticity. Nevertheless, the precise SNS-mediated mechanisms governing hyperalgesia and skeletal muscle atrophy in FM remain unclear. Thus, we employed two distinct FM-like pain models, involving intramuscular injections of acidic saline (pH 4.0) or carrageenan in prepubertal female rats, and evaluated the catecholamine content, adrenergic signaling and overall muscle proteolysis. Subsequently, we assessed the contribution of the SNS to the development of hyperalgesia and muscle atrophy in acidic saline-injected rats treated with clenbuterol (a selective ß2-adrenergic receptor agonist) and in animals maintained under baseline conditions and subjected to epinephrine depletion through adrenodemedullation (ADM). Seven days after inducing an FM-like model with acidic saline or carrageenan, we observed widespread mechanical hyperalgesia along with loss of strength and/or muscle mass. These changes were associated with reduced catecholamine content, suggesting a common underlying mechanism. Notably, treatment with a ß2-agonist alleviated hyperalgesia and prevented muscle atrophy in acidic saline-induced FM-like pain, while epinephrine depletion induced mechanical hyperalgesia and increased muscle proteolysis in animals under baseline conditions. Together, the results suggest that reduced sympathetic activity is involved in the development of pain and muscle atrophy in the murine model of FM analyzed.


Asunto(s)
Clenbuterol , Modelos Animales de Enfermedad , Fibromialgia , Hiperalgesia , Atrofia Muscular , Sistema Nervioso Simpático , Animales , Femenino , Fibromialgia/patología , Fibromialgia/fisiopatología , Atrofia Muscular/patología , Atrofia Muscular/fisiopatología , Hiperalgesia/fisiopatología , Hiperalgesia/patología , Sistema Nervioso Simpático/fisiopatología , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/patología , Clenbuterol/farmacología , Ratas , Carragenina/toxicidad , Ratas Sprague-Dawley , Dolor/patología , Dolor/fisiopatología , Epinefrina , Músculo Esquelético/patología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiopatología , Catecolaminas/metabolismo , Agonistas Adrenérgicos beta/farmacología
3.
J Mater Chem B ; 12(17): 4217-4231, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38596904

RESUMEN

Numerous studies have shown that there are multiple neural activities involved in the process of bone resorption and bone regeneration, and promoting osteogenesis by promoting neural network reconstruction is an effective strategy for repairing critical size bone defects. However, traumatic bone defects often cause activation of the sympathetic nervous system (SNS) in the damaged area, releasing excess catecholamines (CAs), resulting in a decrease in the rate of bone formation. Herein, a 3D-printed scaffold loaded with propranolol (PRN) is proposed to reduce CA concentrations in bone defect areas and promote bone regeneration through drug release. For this purpose, PRN-loaded methacrylated gelatin (GelMA) microspheres were mixed with low-concentration GelMA and perfused into a 3D-printed porous hydroxyapatite (HAp) scaffold. By releasing PRN, which can block ß-adrenergic receptors, it hinders the activation of sympathetic nerves and inhibits the release of excess CA by the SNS. At the same time, the composite scaffold recruits bone marrow mesenchymal stem cells (BMSCs) and promotes the differentiation of BMSCs in the direction of osteoblasts, which effectively promotes bone regeneration in the rabbit femoral condyle defect model. The results of the study showed that the release of PRN from the composite scaffold could effectively hinder the activation of sympathetic nerves and promote bone regeneration, providing a new strategy for the treatment of bone defects.


Asunto(s)
Regeneración Ósea , Células Madre Mesenquimatosas , Impresión Tridimensional , Sistema Nervioso Simpático , Andamios del Tejido , Regeneración Ósea/efectos de los fármacos , Animales , Conejos , Sistema Nervioso Simpático/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Andamios del Tejido/química , Propranolol/farmacología , Propranolol/química , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Gelatina/química , Osteogénesis/efectos de los fármacos , Durapatita/química , Durapatita/farmacología
4.
Hypertens Res ; 47(5): 1323-1337, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38491106

RESUMEN

Paroxysmal sympathetic hyperactivity (PSH) is a common clinical feature secondary to ischemic stroke (IS), but its mechanism is poorly understood. We aimed to investigate the role of H2S in the pathogenesis of PSH. IS patients were divided into malignant (MCI) and non-malignant cerebral infarction (NMCI) group. IS in rats was induced by the right middle cerebral artery occlusion (MCAO). H2S donor (NaHS) or inhibitor (aminooxy-acetic acid, AOAA) were microinjected into the hypothalamic paraventricular nucleus (PVN). Compared with the NMCI group, patients in the MCI group showed PSH, including tachycardia, hypertension, and more plasma norepinephrine (NE) that was positively correlated with levels of creatine kinase, glutamate transaminase, and creatinine respectively. The 1-year survival rate of patients with high plasma NE levels was lower. The hypothalamus of rats with MCAO showed increased activity, especially in the PVN region. The levels of H2S in PVN of the rats with MCAO were reduced, while the blood pressure and renal sympathetic discharge were increased, which could be ameliorated by NaHS and exacerbated by AOAA. NaHS completely reduced the disulfide bond of NMDAR1 in PC12 cells. The inhibition of NMDAR by MK-801 microinjected in PVN of rats with MCAO also could lower blood pressure and renal sympathetic discharge. In conclusion, PSH may be associated with disease progression and survival in patients with IS. Decreased levels of H2S in PVN were involved in regulating sympathetic efferent activity after cerebral infarction. Our results might provide a new strategy and target for the prevention and treatment of PSH.


Asunto(s)
Sulfuro de Hidrógeno , Núcleo Hipotalámico Paraventricular , Animales , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Sulfuro de Hidrógeno/metabolismo , Sulfuro de Hidrógeno/sangre , Masculino , Ratas , Humanos , Anciano , Infarto Cerebral , Persona de Mediana Edad , Ratas Sprague-Dawley , Femenino , Norepinefrina/sangre , Enfermedades del Sistema Nervioso Autónomo , Ácido Aminooxiacético/farmacología , Sistema Nervioso Simpático/fisiopatología , Sistema Nervioso Simpático/efectos de los fármacos , Infarto de la Arteria Cerebral Media/complicaciones , Presión Sanguínea/efectos de los fármacos
5.
Am J Physiol Heart Circ Physiol ; 326(6): H1337-H1349, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38551482

RESUMEN

Nicotine is the primary addictive component of tobacco products. Through its actions on the heart and autonomic nervous system, nicotine exposure is associated with electrophysiological changes and increased arrhythmia susceptibility. To assess the underlying mechanisms, we treated rabbits with transdermal nicotine (NIC, 21 mg/day) or control (CT) patches for 28 days before performing dual optical mapping of transmembrane potential (RH237) and intracellular Ca2+ (Rhod-2 AM) in isolated hearts with intact sympathetic innervation. Sympathetic nerve stimulation (SNS) was performed at the first to third thoracic vertebrae, and ß-adrenergic responsiveness was additionally evaluated following norepinephrine (NE) perfusion. Baseline ex vivo heart rate (HR) and SNS stimulation threshold were higher in NIC versus CT (P = 0.004 and P = 0.003, respectively). Action potential duration alternans emerged at longer pacing cycle lengths (PCL) in NIC versus CT at baseline (P = 0.002) and during SNS (P = 0.0003), with similar results obtained for Ca2+ transient alternans. SNS shortened the PCL at which alternans emerged in CT but not in NIC hearts. NIC-exposed hearts tended to have slower and reduced HR responses to NE perfusion, but ventricular responses to NE were comparable between groups. Although fibrosis was unaltered, NIC hearts had lower sympathetic nerve density (P = 0.03) but no difference in NE content versus CT. These results suggest both sympathetic hypoinnervation of the myocardium and regional differences in ß-adrenergic responsiveness with NIC. This autonomic remodeling may contribute to the increased risk of arrhythmias associated with nicotine exposure, which may be further exacerbated with long-term use.NEW & NOTEWORTHY Here, we show that chronic nicotine exposure was associated with increased heart rate, increased susceptibility to alternans, and reduced sympathetic electrophysiological responses in the intact rabbit heart. We suggest that this was due to sympathetic hypoinnervation of the myocardium and diminished ß-adrenergic responsiveness of the sinoatrial node following nicotine treatment. Though these differences did not result in increased arrhythmia propensity in our study, we hypothesize that prolonged nicotine exposure may exacerbate this proarrhythmic remodeling.


Asunto(s)
Potenciales de Acción , Frecuencia Cardíaca , Corazón , Nicotina , Sistema Nervioso Simpático , Animales , Nicotina/toxicidad , Nicotina/efectos adversos , Conejos , Frecuencia Cardíaca/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Corazón/inervación , Corazón/efectos de los fármacos , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiopatología , Masculino , Agonistas Nicotínicos/toxicidad , Agonistas Nicotínicos/administración & dosificación , Señalización del Calcio/efectos de los fármacos , Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/fisiopatología , Arritmias Cardíacas/metabolismo , Parche Transdérmico , Preparación de Corazón Aislado , Administración Cutánea , Norepinefrina/metabolismo
6.
Eur J Pharmacol ; 974: 176373, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38341079

RESUMEN

BACKGROUND: Oxidative stress and inflammatory cytokines in the hypothalamus paraventricular nucleus (PVN) have been implicated in sympathetic nerve activity and the development of hypertension, but the specific mechanisms underlying their production in the PVN remains to be elucidated. Previous studies have demonstrated that activation of nuclear transcription related factor-2 (Nrf2) in the PVN reduced the production of reactive oxygen species (ROS) and inflammatory mediators. Moreover, AMP-activated protein kinase (AMPK), has been observed to decrease ROS and inflammatory cytokine production when activated in the periphery. 5-amino-1-ß-D-ribofuranosyl-imidazole-4-carboxamide (AICAR) is an AMPK agonist. However, little research has been conducted on the role of AMPK in the PVN during hypertension. Therefore, we hypothesized that AICAR in the PVN is involved in regulating AMPK/Nrf2 pathway, affecting ROS and inflammatory cytokine expression, influencing sympathetic nerve activity. METHODS: Adult male Sprague-Dawley rats were utilized to induce two-kidney, one-clip (2K1C) hypertension via constriction of the right renal artery. Bilateral PVN was microinjected with either artificial cerebrospinal fluid or AICAR once a day for 4 weeks. RESULTS: Compared to the SHAM group, the PVN of 2K1C hypertensive rats decreased p-AMPK and p-Nrf2 expression, increased Fra-Like, NAD(P)H oxidase (NOX)2, NOX4, tumor necrosis factor-α and interleukin (IL)-1ß expression, elevated ROS levels, decreased superoxide dismutase 1 and IL-10 expression, and elevated plasma norepinephrine levels. Bilateral PVN microinjection of AICAR significantly ameliorated these changes. CONCLUSION: These findings suggest that repeated injection of AICAR in the PVN suppresses ROS and inflammatory cytokine production through the AMPK/Nrf2 pathway, reducing sympathetic nerve activity and improving hypertension.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Aminoimidazol Carboxamida , Hipertensión , Factor 2 Relacionado con NF-E2 , Núcleo Hipotalámico Paraventricular , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno , Ribonucleótidos , Transducción de Señal , Animales , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Masculino , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Aminoimidazol Carboxamida/administración & dosificación , Ribonucleótidos/farmacología , Ribonucleótidos/administración & dosificación , Proteínas Quinasas Activadas por AMP/metabolismo , Hipertensión/tratamiento farmacológico , Hipertensión/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Presión Sanguínea/efectos de los fármacos , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/metabolismo , Estrés Oxidativo/efectos de los fármacos , Citocinas/metabolismo
7.
Steroids ; 188: 109132, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36273542

RESUMEN

Several studies have demonstrated that the underlying mechanism of insulin resistance (IR) is linked with developing diseases like diabetes mellitus, hypertension, metabolic syndrome, and polycystic ovary syndrome. In turn, the dysfunction of female gonadal hormones (especially 17ß-estradiol) may be related to the development of IR complications since different studies have shown that 17ß-estradiol has a cardioprotector and vasorelaxant effect. This study aimed was to determine the effect of the 17ß-estradiol administration in insulin-resistant rats and its effects on cardiovascular responses in pithed rats. Thus, the vasopressor responses are induced by sympathetic stimulation or i.v. bolus injections of noradrenaline (α1/2), methoxamine (α1), and UK 14,304 (α2) adrenergic agonist were determined in female pithed rats with fructose-induced insulin resistance or control rats treated with: 1) 17ß-estradiol or 2) its vehicle (oil) for 5 weeks. Thus, 17ß-estradiol decreased heart rate, prevented the increase of blood pressure induced by ovariectomy, but with the opposite effect on sham-operated rats; and decreased vasopressor responses induced by i.v. bolus injections of noradrenaline on sham-operated (control and fructose group) and ovariectomized (control) rats, and those induced by i.v. bolus injections of methoxamine (α1 adrenergic agonist). Overall, these results suggest 17ß-estradiol has a cardioprotective effect, and its effect on vasopressor responses could be mediated mainly by the α1 adrenergic receptor. In contrast, IR with ovariectomy 17ß-estradiol decreases or loses its cardioprotector effect, this could suggest a possible link between the adrenergic receptors and the insulin pathway.


Asunto(s)
Estradiol , Resistencia a la Insulina , Sistema Nervioso Simpático , Animales , Femenino , Humanos , Ratas , Agonistas Adrenérgicos/farmacología , Estradiol/farmacología , Fructosa/farmacología , Insulina , Resistencia a la Insulina/fisiología , Metoxamina/farmacología , Norepinefrina/farmacología , Ovariectomía , Ratas Wistar , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiología , Vasoconstrictores/farmacología
8.
Med Sci Sports Exerc ; 54(3): 408-416, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34711708

RESUMEN

INTRODUCTION: Postmenopausal women (PMW) display exaggerated increases in blood pressure (BP) during exercise, yet the mechanism(s) involved remain unclear. Moreover, research on the impact of menopausal changes in estradiol on cardiovascular control during exercise are limited. Herein, we tested the hypothesis that sympathetic responses during exercise are augmented in PMWcompared with young women (YW), and estradiol administration attenuates these responses. METHODS: Muscle sympathetic nerve activity (MSNA) and mean arterial pressure (MAP) were measured in 13 PMW (58 ± 1 yr) and 17 YW (22 ± 1 yr) during 2 min of isometric handgrip. Separately, MSNA and BP responses were measured during isometric handgrip in six PMW (53 ± 1 yr) before and after 1 month of transdermal estradiol (100 µg·d-1). A period of postexercise ischemia (PEI) to isolate muscle metaboreflex activation followed all handgrip bouts. RESULTS: Resting MAP was similar between PMW and YW, whereas MSNA was greater in PMW (23 ± 3 vs 8 ± 1 bursts per minute; P < 0.05). During handgrip, the increases in MSNA (PMW Δ16 ± 2 vs YW Δ6 ± 1 bursts per minute; P < 0.05) and MAP (PMW Δ18 ± 2 vs YW Δ12 ± 2 mm Hg; P < 0.05) were greater in PMW and remained augmented during PEI. Estradiol administration decreased resting MAP but not MSNA in PMW. Moreover, MSNA (PMW (-E2) Δ27 ± 8 bursts per minute versus PMW (+E2) Δ12 ± 5 bursts per minute; P < 0.05) and MAP (Δ31 ± 8 mm Hg vs Δ20 ± 6 mm Hg; P < 0.05) responses during handgrip were attenuated in PMW after estradiol administration. Likewise, MAP responses during PEI were lower after estradiol. CONCLUSIONS: These data suggest that PMW exhibit an exaggerated MSNA and BP response to isometric exercise, due in part to heightened metaboreflex activation. Furthermore, estradiol administration attenuated BP and MSNA responses to exercise in PMW.


Asunto(s)
Barorreflejo/fisiología , Presión Sanguínea/fisiología , Estradiol/administración & dosificación , Ejercicio Físico/fisiología , Posmenopausia/fisiología , Sistema Nervioso Simpático/fisiología , Factores de Edad , Barorreflejo/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Estrógenos/administración & dosificación , Femenino , Humanos , Persona de Mediana Edad , Posmenopausia/efectos de los fármacos , Sistema Nervioso Simpático/efectos de los fármacos , Adulto Joven
9.
Nutrients ; 13(12)2021 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-34959764

RESUMEN

We previously found increases in uncoupling protein (Ucp)-1 transcription in brown adipose tissue (BAT) of mice following a single oral dose of flavan 3-ol (FL)s, a fraction of catechins and procyanidins. It was confirmed that these changes were totally reduced by co-treatment of adrenaline blockers. According to these previous results, FLs possibly activate sympathetic nervous system (SNS). In this study, we confirmed the marked increase in urinary catecholamine (CA) s projecting SNS activity following a single dose of 50 mg/kg FLs. In addition, we examined the impact of the repeated administration of 50 mg/kg FLs for 14 days on adipose tissues in mice. In BAT, FLs tended to increase the level of Ucp-1 along with significant increase of thermogenic transcriptome factors expressions, such as peroxisome proliferator-activated receptor γ coactivator (PGC)-1α and PR domain-containing (PRDM)1. Expression of browning markers, CD137 and transmembrane protein (TMEM) 26, in addition to PGC-1α were increased in epididymal adipose (eWAT) by FLs. A multilocular morphology with cell size reduction was shown in the inguinal adipose (iWAT), together with increasing the level of Ucp-1 by FLs. These results exert that FLs induce browning in adipose, and this change is possibly produced by the activation of the SNS.


Asunto(s)
Tejido Adiposo/metabolismo , Flavonoides/administración & dosificación , Sistema Nervioso Simpático/efectos de los fármacos , Administración Oral , Animales , Catecolaminas/orina , Proteínas de la Membrana/metabolismo , Ratones , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Termogénesis/efectos de los fármacos , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Proteína Desacopladora 1/metabolismo
10.
Circ Heart Fail ; 14(12): e008365, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34789005

RESUMEN

BACKGROUND: Recent clinical studies demonstrate that SGLT2 (sodium-glucose cotransporter 2) inhibitors ameliorate heart failure (HF). The present study was conducted to assess the expression and function of renal SGLT2 and the influence of enhanced renal sympathetic tone in HF. METHODS: Four weeks after coronary artery ligation surgery to induce HF, surgical bilateral renal denervation (RDN) was performed in rats. Four groups of rats (Sham-operated control [Sham], Sham+RDN, HF and HF+RDN; n=6/group) were used. Immunohistochemistry and Western blot analysis were performed to evaluate the renal SGLT2 expression. One week after RDN (5 weeks after induction of HF), intravenous injection of SGLT2 inhibitor dapagliflozin were performed to assess renal excretory responses. In vitro, human embryonic kidney cells were used to investigate the fractionation of SGLT2 after norepinephrine treatment. RESULTS: In rats with HF, (1) SGLT2 expression in the proximal tubule of the kidney was increased; (2) the response of increases in urine flow, sodium excretion, and glucose excretion to dapagliflozin were greater; and (3) RDN attenuated renal SGLT2 expression and normalized renal functional responses to dapagliflozin. In vitro, norepinephrine promoted translocation of SGLT2 to the cell membrane. CONCLUSIONS: These results indicate that the enhanced tonic renal sympathetic nerve activation in HF increases the expression and functional activity of renal SGLT2. Potentiated trafficking of SGLT2 to cell surface in renal proximal tubules mediated by norepinephrine may contribute to this functional activation of SGLT2 in HF. These findings provide critical insight into the underlying mechanisms for the beneficial effects of SGLT2 inhibitors on HF reported in the clinical studies.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Riñón/inervación , Transportador 2 de Sodio-Glucosa/metabolismo , Animales , Glucosa/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Masculino , Ratas Sprague-Dawley , Sodio/metabolismo , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiopatología
11.
Am J Physiol Regul Integr Comp Physiol ; 321(3): R513-R521, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34346721

RESUMEN

Experiments aimed to evaluate the tissue distribution of Mas-related G protein-coupled receptor D (MrgD) revealed the presence of immunoreactivity for the MrgD protein in the rostral insular cortex (rIC), an important area for autonomic and cardiovascular control. To investigate the relevance of this finding, we evaluated the cardiovascular effects produced by the endogenous ligand of MrgD, alamandine, in this brain region. Mean arterial pressure (MAP), heart rate (HR), and renal sympathetic nerve activity (RSNA) were recorded in urethane anesthetized rats. Unilateral microinjection of equimolar doses of alamandine (40 pmol/100 nL), angiotensin-(1-7), angiotensin II, angiotensin A, and Mas/MrgD antagonist d-Pro7-Ang-1-7 (50 pmol/100 nL), Mas antagonist A779 (100 pmol/100 nL), or vehicle (0.9% NaCl) were made in different rats (n = 4-6/group) into rIC. To verify the specificity of the region, a microinjection of alamandine was also performed into intermediate insular cortex (iIC). Microinjection of alamandine in rIC produced an increase in MAP (Δ = 15 ± 2 mmHg), HR (Δ = 36 ± 4 beats/min), and RSNA (Δ = 31 ± 4%), but was without effects at iIC. Strikingly, an equimolar dose of angiotensin-(1-7) at rIC did not produce any change in MAP, HR, and RSNA. Angiotensin II and angiotensin A produced only minor effects. Alamandine effects were not altered by A-779, a Mas antagonist, but were completely blocked by the Mas/MrgD antagonist d-Pro7-Ang-(1-7). Therefore, we have identified a brain region in which alamandine/MrgD receptor but not angiotensin-(1-7)/Mas could be involved in the modulation of cardiovascular-related neuronal activity. This observation also suggests that alamandine might possess unique effects unrelated to angiotensin-(1-7) in the brain.


Asunto(s)
Angiotensina I/farmacología , Presión Arterial/efectos de los fármacos , Sistema Cardiovascular/inervación , Corteza Cerebral/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Riñón/inervación , Proteínas del Tejido Nervioso/agonistas , Oligopéptidos/farmacología , Fragmentos de Péptidos/farmacología , Receptores Acoplados a Proteínas G/agonistas , Sistema Nervioso Simpático/efectos de los fármacos , Animales , Corteza Cerebral/fisiología , Ligandos , Masculino , Microinyecciones , Proteínas del Tejido Nervioso/metabolismo , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/agonistas , Proteínas Proto-Oncogénicas/metabolismo , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Sistema Nervioso Simpático/fisiología
12.
J Neuroendocrinol ; 33(8): e13006, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34235800

RESUMEN

In hungry animals, neuropeptide Y (NPY) neurones in the arcuate nucleus (ArcN) are activated to suppress energy expenditure, in part by decreasing brown adipose tissue sympathetic nerve activity (BAT SNA); however, the NPY receptor subtype and brain neurocircuitry are unclear. In the present study, we investigated the inhibition of BAT SNA by exogenous and endogenous NPY via binding to Y1 receptors (NPY1R) in the hypothalamic paraventricular nucleus (PVN) and dorsomedial hypothalamus (DMH), in anaesthetised male rats. Downstream projections of PVN/DMH NPY1R-expressing neurones were identified using male Npy1r-cre mice and localised unilateral DMH or PVN injections of an adeno-associated virus, which allows for the cre-dependent expression of a fluorescent protein (mCherry) in the cell bodies, axon fibres and nerve terminals of NPY1R-containing neurones. Nanoinjections of NPY into the DMH of cooled rats decreased BAT SNA, as well as mean arterial pressure (MAP) and heart rate (HR), and these responses were reversed by subsequent injection of the selective NPY1R antagonist, BIBO3304. In warmed rats, with little to no BAT SNA, bilateral nanoinjections of BIBO3304 into the DMH or PVN increased BAT SNA, MAP and HR. DMH NPY1R-expressing neurones projected heavily to the raphe pallidus (RPa), which houses BAT presympathetic neurones, as well as the PVN. In anaesthetised mice, DMH BIBO3304 increased splanchnic SNA, MAP and HR, all of which were reversed by nonselective blockade of the PVN with muscimol, suggesting that DMH-to-PVN connections are involved in this DMH BIBO3304 disinhibition. PVN Y1R expressing neurones also projected to the RPa, as well as to the nucleus tractus solitarius. We conclude that NPY tonically released in the DMH and PVN suppresses BAT SNA, MAP and HR via Y1R. Downstream neuropathways for BAT SNA may utilise direct projections to the RPa. Release of tonic NPY inhibition of BAT SNA may contribute to feeding- and diet-induced thermogenesis.


Asunto(s)
Sistema Cardiovascular/efectos de los fármacos , Núcleo Hipotalámico Dorsomedial/efectos de los fármacos , Neuropéptido Y/farmacología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Termogénesis/efectos de los fármacos , Animales , Presión Sanguínea/efectos de los fármacos , Sistema Cardiovascular/inervación , Núcleo Hipotalámico Dorsomedial/metabolismo , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Núcleo Hipotalámico Paraventricular/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Neuropéptido Y/metabolismo , Receptores de Neuropéptido Y/fisiología , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/metabolismo
13.
J Cardiovasc Pharmacol ; 77(6): 745-755, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-34057159

RESUMEN

ABSTRACT: The NLRP3 inflammasome is activated by myocardial infarction and then induces the activation of inflammatory caspase-1 activation and maturation of IL-1ß, a regulator of synthesis of the nerve growth factor (NGF). Here, we studied whether taurine, 2-aminoethanesulphonic acid, can attenuate cardiac sympathetic reinnervation by modulating NLRP3 inflammasome-mediated NGF in a rat model of myocardial infarction. Male Wistar rats were subjected to coronary ligation and then randomized to either saline or taurine for 3 days or 4 weeks. Postinfarction was associated with activation of NF-κB (p65) and NLRP3 inflammasome component and increased the protein and expression of IL-1ß. Macrophages at the border zone were shown to be positive for IL-1ß 3 days postinfarction. Compared with vehicle, infarcted rats treated with taurine significantly attenuated myocardial messenger RNA and protein levels of NF-κB, NLRP3 inflammasome, mature caspase-1, and IL-1ß. Immunofluorescent analysis, real-time quantitative reverse transcription polymerase chain reaction, and Western blotting of NGF showed that sympathetic hyperinnervation was blunted after administering taurine. Arrhythmia vulnerability in the taurine-treated infarcted rats was significantly improved than those in vehicle. Ex vivo studies showed that taurine infusion reduced myocardial IL-1ß level at the extent similar to either pyrrolidine dithiocarbamate or CP-456,773, inhibitors of NF-κB and NLRP3 inflammasome, implying the key axis of NF-κB/NLRP3 inflammasome in mediating taurine-related anti-inflammation. Furthermore, administration of anti-IL-1ß antibody reduced NGF levels. Taurine attenuated sympathetic innervation mainly by NLRP3 inflammasome/IL-1ß-dependent pathway, which downregulated expression of NGF in infarcted rats. These findings may provide a new insight into the anti-inflammation effect of taurine.


Asunto(s)
Antiinflamatorios/farmacología , Infarto del Miocardio/tratamiento farmacológico , Sistema Nervioso Simpático/efectos de los fármacos , Taurina/farmacología , Animales , Modelos Animales de Enfermedad , Inflamasomas/efectos de los fármacos , Inflamasomas/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/patología , Interleucina-1beta/inmunología , Macrófagos/metabolismo , Masculino , Infarto del Miocardio/fisiopatología , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Distribución Aleatoria , Ratas , Ratas Wistar , Sistema Nervioso Simpático/fisiopatología , Factor de Transcripción ReIA/metabolismo
14.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-33972441

RESUMEN

Neuromodulation of immune function by stimulating the autonomic connections to the spleen has been demonstrated in rodent models. Consequently, neuroimmune modulation has been proposed as a new therapeutic strategy for the treatment of inflammatory conditions. However, demonstration of the translation of these immunomodulatory mechanisms in anatomically and physiologically relevant models is still lacking. Additionally, translational models are required to identify stimulation parameters that can be transferred to clinical applications of bioelectronic medicines. Here, we performed neuroanatomical and functional comparison of the mouse, rat, pig, and human splenic nerve using in vivo and ex vivo preparations. The pig was identified as a more suitable model of the human splenic innervation. Using functional electrophysiology, we developed a clinically relevant marker of splenic nerve engagement through stimulation-dependent reversible reduction in local blood flow. Translation of immunomodulatory mechanisms were then assessed using pig splenocytes and two models of acute inflammation in anesthetized pigs. The pig splenic nerve was shown to locally release noradrenaline upon stimulation, which was able to modulate cytokine production by pig splenocytes. Splenic nerve stimulation was found to promote cardiovascular protection as well as cytokine modulation in a high- and a low-dose lipopolysaccharide model, respectively. Importantly, splenic nerve-induced cytokine modulation was reproduced by stimulating the efferent trunk of the cervical vagus nerve. This work demonstrates that immune responses can be modulated by stimulation of spleen-targeted autonomic nerves in translational species and identifies splenic nerve stimulation parameters and biomarkers that are directly applicable to humans due to anatomical and electrophysiological similarities.


Asunto(s)
Sistema Inmunológico/inervación , Inmunomodulación/efectos de los fármacos , Bazo/inmunología , Sistema Nervioso Simpático/inmunología , Nervio Vago/inmunología , Animales , Femenino , Expresión Génica , Humanos , Sistema Inmunológico/efectos de los fármacos , Inflamación , Interleucina-6/genética , Interleucina-6/inmunología , Lipopolisacáridos/farmacología , Ratones , Microcirculación/efectos de los fármacos , Microcirculación/genética , Microcirculación/inmunología , Norepinefrina/farmacología , Ratas , Especificidad de la Especie , Bazo/efectos de los fármacos , Bazo/inervación , Bazo/patología , Porcinos , Sistema Nervioso Simpático/efectos de los fármacos , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Nervio Vago/efectos de los fármacos , Estimulación del Nervio Vago/métodos
15.
Sci Rep ; 11(1): 9167, 2021 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-33911125

RESUMEN

There is a close relationship between the gut microbiota and metabolic disorders. In this study, acute administration of the glucagon-like peptide-1 receptor agonist (GLP-1RA) liraglutide to mice increased the cecal levels of caseinolytic protease B, a component of Escherichia coli, and of norepinephrine. Chemical sympathectomy blocked these events. Norepinephrine was found to pass into the intestinal lumen in vitro. c-Fos staining of the intermediolateral nucleus was identified as indirect evidence of sympathetic nervous system activation of the intestinal tract by GLP-1RA. Under normal conditions, the increase in E. coli did not affect the host. However, in mice with colitis, bacterial translocation was observed with attenuation of tight junction gene expression. This is the first study to investigate the unique underlying mechanisms related the effects of GLP-1RA on changes in the gut bacterium.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Receptor del Péptido 1 Similar al Glucagón/agonistas , Liraglutida/farmacología , Norepinefrina/metabolismo , Adrenalectomía , Animales , Apetito , Ciego/efectos de los fármacos , Ciego/metabolismo , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/genética , Colitis/microbiología , Endopeptidasa Clp/metabolismo , Escherichia coli/efectos de los fármacos , Proteínas de Escherichia coli/metabolismo , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal/efectos de los fármacos , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Proteínas de Choque Térmico/metabolismo , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-fos/metabolismo , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/metabolismo
16.
Toxicol Lett ; 344: 34-45, 2021 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-33667609

RESUMEN

ASP7962 is a small molecule inhibitor for the nerve growth factor (NGF) receptor, tropomyosin-related kinase A (TrkA). NGF contributes to the survival of sensory and sympathetic neurons through TrkA receptor activation. Gross, microscopic, and quantitative effects to the nervous system were evaluated following oral ASP7962 administration to Sprague Dawley rats for 4 weeks and 13 weeks and after a recovery period. Histopathological findings included reversible neuronal atrophy but no neuronal death in the sympathetic ganglia (cervicothoracic ganglion, cranial mesenteric ganglion or superior [cranial] cervical ganglion). Stereological analysis showed reversible decreased ganglion volume and/or decreased neuron size in the superior (cranial) cervical ganglion in both the 4-week and the 13-week repeated dose studies. There were no test article related changes in the brain, dorsal root ganglia with spinal nerve roots or trigeminal ganglia and no functional deficits. ASP7962 did not cause any detectable dysfunction of the sympathetic and sensory nervous system in either study.


Asunto(s)
Neuronas Aferentes/efectos de los fármacos , Receptor trkA/antagonistas & inhibidores , Sistema Nervioso Simpático/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Masculino , Neuronas/efectos de los fármacos , Síndromes de Neurotoxicidad/metabolismo , Ratas , Ratas Sprague-Dawley , Ganglio Estrellado/citología , Ganglio Estrellado/efectos de los fármacos , Ganglio Cervical Superior/citología , Ganglio Cervical Superior/efectos de los fármacos , Ganglio del Trigémino/efectos de los fármacos
17.
Physiol Res ; 70(2): 265-272, 2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-33676384

RESUMEN

It has been suggested that sympathetic activity, measured as changes in electrical skin impedance (SI), can be used to assess the adequacy of general anesthesia. Our prospective study investigated if measurements of skin impedance can determine levels of sedation induced by midazolam. Twenty-seven patients scheduled for arthroscopy requiring general anesthesia were served as their own control. These were blinded to the order of injections by telling them that they will be randomly administered a placebo (saline) orsedative agent. A DM 3900 multimeter was used for SI measurements. The degree of sedation was measured using the modified Observer's Assessment of Alertness and Sedation (mOAAS) scale. Resting SI values were noted, and all participants were then administered the placebo followed 5 min later by midazolam 2 mg i.v. Five min after that, patients were administered standard general anesthesia with propofol, oxygen, nitrous oxide 60 %, and isoflurane 1 MAC via a laryngeal mask, and sufentanil 5 - 10 µg. SI significantly increased after administration of midazolam and induction of anesthesia. There were no significant differences between pre-administration (baseline) and placebo and end of surgery and end of anesthesia with closed eyes. There were highly significant differences (p<0.001) between pre-administration vs. midazolam, placebo vs. midazolam, pre-administration vs. induction of anesthesia. We found slight correlation between mOAAS and SI. There were no significant changes between the end of surgery and the end of anesthesia with closed eyes, but SI significantly decreased (p<0.01) after eyes opened.


Asunto(s)
Periodo de Recuperación de la Anestesia , Estado de Conciencia/efectos de los fármacos , Respuesta Galvánica de la Piel/efectos de los fármacos , Hipnóticos y Sedantes/uso terapéutico , Midazolam/uso terapéutico , Piel/inervación , Sistema Nervioso Simpático/efectos de los fármacos , Adulto , Anestesia General , República Checa , Método Doble Ciego , Impedancia Eléctrica , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Sistema Nervioso Simpático/fisiología , Factores de Tiempo
18.
Biochem Biophys Res Commun ; 548: 84-90, 2021 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-33636639

RESUMEN

Brain nicotinic acetylcholine receptors (nAChRs) reportedly suppress the micturition, but the mechanisms responsible for this suppression remain unclear. We previously reported that intracerebroventricularly administered (±)-epibatidine (non-selective nAChR agonist) activated the sympatho-adrenomedullary system, which can affect the micturition. Therefore, we investigated (1) whether intracerebroventricularly administered (±)-epibatidine-induced effects on the micturition were dependent on the sympatho-adrenomedullary system, and (2) brain nAChR subtypes involved in the (±)-epibatidine-induced effects in urethane-anesthetized male Wistar rats. Plasma noradrenaline and adrenaline (catecholamines) were measured just before and 5 min after (±)-epibatidine administration. Evaluation of urodynamic parameters, intercontraction intervals (ICI) and maximal voiding pressure (MVP) by cystometry was started 1 h before (±)-epibatidine administration or intracerebroventricular pretreatment with other drugs and continued 1 h after (±)-epibatidine administration. Intracerebroventricularly administered (±)-epibatidine elevated plasma catecholamines and prolonged ICI without affecting MVP, and these changes were suppressed by intracerebroventricularly pretreated mecamylamine (non-selective nAChR antagonist). Acute bilateral adrenalectomy abolished the (±)-epibatidine-induced elevation of plasma catecholamines, but had no effect on the (±)-epibatidine-induced ICI prolongation. The latter was suppressed by intracerebroventricularly pretreated methyllycaconitine (selective α7-nAChR antagonist), SR95531 (GABAA antagonist), and SCH50911 (GABAB antagonist), but not by dihydro-ß-erythroidine (selective α4ß2-nAChR antagonist). Intracerebroventricularly administered PHA568487 (selective α7-nAChR agonist) prolonged ICI without affecting MVP, similar to (±)-epibatidine. These results suggest that stimulation of brain α7-nAChRs suppresses the rat micturition through brain GABAA/GABAB receptors, independently of the sympatho-adrenomedullary outflow modulation.


Asunto(s)
Encéfalo/metabolismo , Receptores de GABA/metabolismo , Micción , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Médula Suprarrenal/efectos de los fármacos , Médula Suprarrenal/metabolismo , Adrenalectomía , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Epinefrina/sangre , Masculino , Contracción Muscular/efectos de los fármacos , Norepinefrina/sangre , Piridinas/farmacología , Ratas Wistar , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/metabolismo
19.
Pflugers Arch ; 473(1): 107-120, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33074398

RESUMEN

Oxaliplatin is a platinum-based alkylating chemotherapeutic agent used for cancer treatment. Neurotoxicity is one of its major adverse effects that often demands dose limitation. However, the effects of chronic oxaliplatin on the toxicity of the autonomic nervous system regulating cardiorespiratory function and adaptive reflexes are unknown. Male Sprague Dawley rats were treated with intraperitoneal oxaliplatin (3 mg kg-1 per dose) 3 times a week for 14 days. The effects of chronic oxaliplatin treatment on baseline mean arterial pressure (MAP); heart rate (HR); splanchnic sympathetic nerve activity (sSNA); phrenic nerve activity (PNA) and its amplitude (PNamp) and frequency (PNf); and sympathetic reflexes were investigated in anaesthetised, vagotomised and artificially ventilated rats. The same parameters were evaluated after acute oxaliplatin injection, and in the chronic treatment group following a single dose of oxaliplatin. The amount of platinum in the brain was determined with atomic absorption spectrophotometry. Chronic oxaliplatin treatment significantly increased MAP, sSNA and PNf and decreased HR and PNamp, while acute oxaliplatin had no effects. Platinum was accumulated in the brain after chronic oxaliplatin treatment. In the chronic oxaliplatin treatment group, further administration of a single dose of oxaliplatin increased MAP and sSNA. The baroreceptor sensitivity and somatosympathetic reflex were attenuated at rest while the sympathoexcitatory response to hypercapnia was increased in the chronic treatment group. This is the first study to reveal oxaliplatin-induced alterations in the central regulation of cardiovascular and respiratory functions as well as reflexes that may lead to hypertension and breathing disorders which may be mediated via accumulated platinum in the brain.


Asunto(s)
Fenómenos Fisiológicos Cardiovasculares/efectos de los fármacos , Oxaliplatino/efectos adversos , Oxaliplatino/farmacocinética , Platino (Metal)/metabolismo , Fenómenos Fisiológicos Respiratorios/efectos de los fármacos , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Barorreflejo/efectos de los fármacos , Sangre/efectos de los fármacos , Células Quimiorreceptoras/efectos de los fármacos , Esquema de Medicación , Frecuencia Cardíaca , Masculino , Oxaliplatino/administración & dosificación , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Nervios Esplácnicos/efectos de los fármacos , Sistema Nervioso Simpático/efectos de los fármacos , Distribución Tisular
20.
Am J Physiol Heart Circ Physiol ; 320(2): H772-H786, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33337962

RESUMEN

Peripherally or centrally administered TNF-α elicits a prolonged sympathetically mediated pressor response, but the underlying molecular mechanisms are unknown. Activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2) in cardiovascular regions of the brain has recently been recognized as a key mediator of sympathetic excitation, and ERK1/2 signaling is induced by activation of epidermal growth factor receptor (EGFR) tyrosine kinase activity. The present study examined the role of EGFR and ERK1/2 signaling in the sympathetic response to TNF-α. In urethane-anesthetized rats, intracarotid artery injection of TNF-α increased phosphorylation of EGFR and ERK1/2 in the subfornical organ (SFO) and the hypothalamic paraventricular nucleus (PVN); upregulated the gene expression of excitatory mediators in SFO and PVN; and increased blood pressure (BP), heart rate (HR), and renal sympathetic nerve activity (RSNA). A continuous intracerebroventricular infusion of the selective EGFR tyrosine kinase inhibitor AG1478 or the ERK1/2 inhibitor PD98059 significantly attenuated these responses. Bilateral PVN microinjections of TNF-α also increased phosphorylated ERK1/2 and the gene expression of excitatory mediators in PVN, along with increases in BP, HR, and RSNA, and these responses were substantially reduced by prior bilateral PVN microinjections of AG1478. These results identify activation of EGFR in cardiovascular regulatory regions of the forebrain as an important molecular mediator of TNF-α-driven sympatho-excitatory responses and suggest that EGFR activation of the ERK1/2 signaling pathway plays an essential role. These mechanisms likely contribute to sympathetic excitation in pathophysiological states like heart failure and hypertension, in which circulating and brain TNF-α levels are increased.NEW & NOTEWORTHY Proinflammatory cytokines contribute to the augmented sympathetic nerve activity in hypertension and heart failure, but the central mechanisms involved are largely unknown. The present study reveals that TNF-α transactivates EGFR in the subfornical organ and the hypothalamic paraventricular nucleus to initiate ERK1/2 signaling, upregulate the gene expression of excitatory mediators, and increase sympathetic nerve activity. These findings identify EGFR as a gateway to sympathetic excitation and a potential target for intervention in cardiovascular disease states.


Asunto(s)
Sistema Cardiovascular/inervación , Receptores ErbB/metabolismo , Hemodinámica/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Prosencéfalo/efectos de los fármacos , Sistema Nervioso Simpático/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Fosforilación , Prosencéfalo/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Ratas Sprague-Dawley , Transducción de Señal , Tirfostinos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA