Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.874
Filter
1.
J Hypertens ; 42(6): 1027-1038, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38690904

ABSTRACT

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.


Subject(s)
Interleukin-1beta , Kidney , Sympathetic Nervous System , Tumor Necrosis Factor-alpha , Animals , Interleukin-1beta/pharmacology , Rats , Kidney/innervation , Kidney/drug effects , Male , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiology , Rats, Sprague-Dawley , Heart Rate/drug effects , Bradykinin/pharmacology , Reflex/drug effects , Blood Pressure/drug effects , Adenosine/administration & dosage , Adenosine/pharmacology , Saline Solution, Hypertonic/administration & dosage , Saline Solution, Hypertonic/pharmacology
2.
Biomed Pharmacother ; 174: 116549, 2024 May.
Article in English | MEDLINE | ID: mdl-38593701

ABSTRACT

This study aimed to determine whether trimethylamine N-oxide (TMAO) was involved in sympathetic activation in aging and the underlying mechanisms. Our hypothesis is TMAO reduces P2Y12 receptor (P2Y12R) and induces microglia-mediated inflammation in the paraventricular nucleus (PVN), then leading to sympathetic activation in aging. This study involved 18 young adults and 16 old adults. Aging rats were established by injecting D-galactose (D-gal, 200 mg/kg/d) subcutaneously for 12 weeks. TMAO (120 mg/kg/d) or 1% 3, 3-dimethyl-l-butanol (DMB) was administrated via drinking water for 12 weeks to investigate their effects on neuroinflammation and sympathetic activation in aging rats. Plasma TMAO, NE and IL-1ß levels were higher in old adults than in young adults. In addition, standard deviation of all normal to normal intervals (SDNN) and standard deviation of the average of normal to normal intervals (SDANN) were lower in old adults and negatively correlated with TMAO, indicating sympathetic activation in old adults, which is associated with an increase in TMAO levels. Treatment of rats with D-gal showed increased senescence-associated protein levels and microglia-mediated inflammation, as well as decreased P2Y12R protein levels in PVN. Plasma TMAO, NE and IL-1ß levels were increased, accompanied by enhanced renal sympathetic nerve activity (RSNA). While TMAO treatment exacerbated the above phenomenon, DMB mitigated it. These findings suggest that TMAO contributes to sympathetic hyperactivity in aging by downregulating P2Y12R in microglia and increasing inflammation in the PVN. These results may provide promising new target for the prevention and treatment of aging and aging-related diseases.


Subject(s)
Down-Regulation , Galactose , Methylamines , Microglia , Receptors, Purinergic P2Y12 , Animals , Rats , Aging/metabolism , Down-Regulation/drug effects , Galactose/pharmacology , Inflammation/chemically induced , Inflammation/metabolism , Interleukin-1beta/metabolism , Methylamines/pharmacology , Microglia/drug effects , Microglia/metabolism , Norepinephrine/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Rats, Sprague-Dawley , Receptors, Purinergic P2Y12/metabolism , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism
3.
J Physiol ; 602(10): 2199-2226, 2024 May.
Article in English | MEDLINE | ID: mdl-38656747

ABSTRACT

During the urine storage phase, tonically contracting urethral musculature would have a higher energy consumption than bladder muscle that develops phasic contractions. However, ischaemic dysfunction is less prevalent in the urethra than in the bladder, suggesting that urethral vasculature has intrinsic properties ensuring an adequate blood supply. Diameter changes in rat or mouse urethral arterioles were measured using a video-tracking system. Intercellular Ca2+ dynamics in arteriolar smooth muscle (SMCs) and endothelial cells were visualised using NG2- and parvalbumin-GCaMP6 mice, respectively. Fluorescence immunohistochemistry was used to visualise the perivascular innervation. In rat urethral arterioles, sympathetic vasoconstrictions were predominantly suppressed by α,ß-methylene ATP (10 µM) but not prazosin (1 µM). Tadalafil (100 nM), a PDE5 inhibitor, diminished the vasoconstrictions in a manner reversed by N-ω-propyl-l-arginine hydrochloride (l-NPA, 1 µM), a neuronal NO synthesis (nNOS) inhibitor. Vesicular acetylcholine transporter immunoreactive perivascular nerve fibres co-expressing nNOS were intertwined with tyrosine hydroxylase immunoreactive sympathetic nerve fibres. In phenylephrine (1 µM) pre-constricted rat or mouse urethral arterioles, nerve-evoked vasodilatations or transient SMC Ca2+ reductions were largely diminished by l-nitroarginine (l-NA, 10 µM), a broad-spectrum NOS inhibitor, but not by l-NPA. The CGRP receptor antagonist BIBN-4096 (1 µM) shortened the vasodilatory responses, while atropine (1 µM) abolished the l-NA-resistant transient vasodilatory responses. Nerve-evoked endothelial Ca2+ transients were abolished by atropine plus guanethidine (10 µM), indicating its neurotransmitter origin and absence of non-adrenergic non-cholinergic endothelial NO release. In urethral arterioles, NO released from parasympathetic nerves counteracts sympathetic vasoconstrictions pre- and post-synaptically to restrict arteriolar contractility. KEY POINTS: Despite a higher energy consumption of the urethral musculature than the bladder detrusor muscle, ischaemic dysfunction of the urethra is less prevalent than that of the bladder. In the urethral arterioles, sympathetic vasoconstrictions are predominately mediated by ATP, not noradrenaline. NO released from parasympathetic nerves counteracts sympathetic vasoconstrictions by its pre-synaptic inhibition of sympathetic transmission as well as post-synaptic arteriolar smooth muscle relaxation. Acetylcholine released from parasympathetic nerves contributes to endothelium-dependent, transient vasodilatations, while CGRP released from sensory nerves prolongs NO-mediated vasodilatations. PDE5 inhibitors could be beneficial to maintain and/or improve urethral blood supply and in turn the volume and contractility of urethral musculature.


Subject(s)
Urethra , Vasoconstriction , Animals , Female , Urethra/innervation , Urethra/physiology , Urethra/drug effects , Vasoconstriction/drug effects , Mice , Arterioles/drug effects , Arterioles/physiology , Arterioles/metabolism , Rats , Mice, Inbred C57BL , Rats, Sprague-Dawley , Sympathetic Nervous System/physiology , Sympathetic Nervous System/drug effects
4.
J Mater Chem B ; 12(17): 4217-4231, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38596904

ABSTRACT

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.


Subject(s)
Bone Regeneration , Mesenchymal Stem Cells , Printing, Three-Dimensional , Sympathetic Nervous System , Tissue Scaffolds , Bone Regeneration/drug effects , Animals , Rabbits , Sympathetic Nervous System/drug effects , Mesenchymal Stem Cells/drug effects , Tissue Scaffolds/chemistry , Propranolol/pharmacology , Propranolol/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Gelatin/chemistry , Osteogenesis/drug effects , Durapatite/chemistry , Durapatite/pharmacology
5.
Hypertens Res ; 47(5): 1323-1337, 2024 May.
Article in English | MEDLINE | ID: mdl-38491106

ABSTRACT

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.


Subject(s)
Hydrogen Sulfide , Paraventricular Hypothalamic Nucleus , Animals , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/blood , Male , Rats , Humans , Aged , Cerebral Infarction , Middle Aged , Rats, Sprague-Dawley , Female , Norepinephrine/blood , Autonomic Nervous System Diseases , Aminooxyacetic Acid/pharmacology , Sympathetic Nervous System/physiopathology , Sympathetic Nervous System/drug effects , Infarction, Middle Cerebral Artery/complications , Blood Pressure/drug effects
6.
Am J Physiol Heart Circ Physiol ; 326(6): H1337-H1349, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38551482

ABSTRACT

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.


Subject(s)
Action Potentials , Heart Rate , Heart , Nicotine , Sympathetic Nervous System , Animals , Nicotine/toxicity , Nicotine/adverse effects , Rabbits , Heart Rate/drug effects , Action Potentials/drug effects , Heart/innervation , Heart/drug effects , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiopathology , Male , Nicotinic Agonists/toxicity , Nicotinic Agonists/administration & dosage , Calcium Signaling/drug effects , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/metabolism , Transdermal Patch , Isolated Heart Preparation , Administration, Cutaneous , Norepinephrine/metabolism
7.
Steroids ; 188: 109132, 2022 12.
Article in English | MEDLINE | ID: mdl-36273542

ABSTRACT

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.


Subject(s)
Estradiol , Insulin Resistance , Sympathetic Nervous System , Animals , Female , Humans , Rats , Adrenergic Agonists/pharmacology , Estradiol/pharmacology , Fructose/pharmacology , Insulin , Insulin Resistance/physiology , Methoxamine/pharmacology , Norepinephrine/pharmacology , Ovariectomy , Rats, Wistar , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiology , Vasoconstrictor Agents/pharmacology
8.
Life Sci ; 295: 120405, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35181311

ABSTRACT

AIMS: The rostral ventrolateral medulla (RVLM) is the main sympathetic output of the central nervous system to control blood pressure. Reportedly, reactive oxygen species (ROS) can increase arterial pressure, leading to hypertension. As ROS increase the sympathetic tone in RVLM and obese animals present grater oxidative stress, it would be important to note this relationship. MAIN METHODS: Therefore, we evaluated the systemic and central effects (in the RVLM) of vitamin C (vit C, an antioxidant) on the redox balance and cardiovascular and autonomic profiles in hyperadipose male rats. We also evaluated the neurotransmission by L-glutamate (L-glu) and vit C in the RVLM of awake hyperadipose rats. KEY FINDINGS: Our study confirmed that hyperadipose rats were hypertensive and tachycardic, presented increased sympathetic and decreased parasympathetic modulation of the heart, and had increased plasma lipoperoxidation compared with the control rats (CTR). Oral vitamin C treatment reverted cardiovascular, autonomic, and plasma redox dysfunction. Hyperadipose rats presented a higher blood pressure increase after L-glu microinjection and a lower response to vit C in the RVLM compared with the CTR group. Biochemical analysis of redox balance in RVLM punches showed that hyperadipose rats have increased NBT and T-BARS, and after treatment with vit C, the oxidative profile decreased. The antioxidative activity of vit C reduced the amount of ROS in the RVLM area that might have resulted in lowered blood pressure and sympathetic modulation. SIGNIFICANCE: Our data suggest central and peripheral benefits of vit C treatment on cardiovascular, autonomic, and oxidative dysfunctions in hyperadipose animals.


Subject(s)
Ascorbic Acid/pharmacology , Hypertension/drug therapy , Medulla Oblongata/metabolism , Animals , Antioxidants/pharmacology , Autonomic Nervous System/physiopathology , Blood Pressure/drug effects , Cardiovascular System/physiopathology , Heart Rate/drug effects , Hypertension/physiopathology , Male , Medulla Oblongata/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Rats, Wistar , Reactive Oxygen Species/pharmacology , Superoxide Dismutase/metabolism , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism
9.
J Neurophysiol ; 127(2): 548-558, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35044844

ABSTRACT

Previous research suggests that the neuropeptide orexin A contributes to sympathetic blood pressure (BP) control inasmuch as hypothalamic injection of orexin A increases sympathetic vasomotor tone and arterial BP in rodents. In humans with narcolepsy, a disorder associated with loss of orexin-producing neurons, vasoconstrictive muscle sympathetic nerve activity (MSNA) is reduced. Since intranasally administered oligopeptides like orexin are known to modulate brain function, we investigated the effect of intranasal orexin A on vascular sympathetic baroreflex function in healthy humans. In a balanced, double-blind crossover study, orexin A (500 nmol) and placebo, respectively, were intranasally administered to 10 lean healthy males (age 25.8 ± 4.6 yr). MSNA was assessed microneurographically before and 30-45 min after either substance administration. Additionally, baroreflex was challenged via graded infusions of vasoactive drugs before and after substance administration. Baroreflex function was defined as the correlation of BP with MSNA and heart rate. Intranasal orexin A compared with placebo induced a significant increase in resting MSNA from pre-to postadministration [Δburst rate, orexin A vs. placebo: +5.8 ± 0.8 vs. +2.1 ± 0.6 bursts/min, P = 0.007; total activity 169 ± 11.5% vs. 115 ± 5.0%; P = 0.002]. BP, heart rate, and sympathovagal balance to the heart, as represented by heart rate variability (HRV), as well as baroreflex sensitivity during the vasoactive challenge were not altered. Intranasally administered orexin A acutely induced vasoconstrictory sympathoactivation in healthy male humans. This result suggests that orexin A mediates upward resetting of the vascular baroreflex set point at centers superordinate to the mere baroreflex feedback loop.NEW & NOTEWORTHY Our pilot study adds another important part to the complex network of neuroendocrine-sympathetic interaction. Our results demonstrate that intranasal orexin A elicits an excitatory effect on sympathetic vascular tone superordinate to mere baroreflex feedback regulation. This resetting of the baroreflex set point suggests an activation of hypothalamic core centers such as the paraventricular nucleus (PVN). The role of the orexinergic system in the development of neurogenic arterial hypertension warrants further investigations.


Subject(s)
Baroreflex/drug effects , Blood Pressure/drug effects , Heart Rate/drug effects , Orexins/pharmacology , Sympathetic Nervous System/drug effects , Vasoconstriction/drug effects , Administration, Intranasal , Adult , Cross-Over Studies , Double-Blind Method , Humans , Male , Orexins/administration & dosage , Pilot Projects , Young Adult
10.
Med Sci Sports Exerc ; 54(3): 408-416, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34711708

ABSTRACT

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.


Subject(s)
Baroreflex/physiology , Blood Pressure/physiology , Estradiol/administration & dosage , Exercise/physiology , Postmenopause/physiology , Sympathetic Nervous System/physiology , Age Factors , Baroreflex/drug effects , Blood Pressure/drug effects , Estrogens/administration & dosage , Female , Humans , Middle Aged , Postmenopause/drug effects , Sympathetic Nervous System/drug effects , Young Adult
11.
Pharmacology ; 107(1-2): 102-110, 2022.
Article in English | MEDLINE | ID: mdl-34718242

ABSTRACT

INTRODUCTION: Ghrelin is an endogenous peptide with potential protective effects on ischemic heart. METHODS: Synthetic ghrelin was administered (100 µg·kg-1 subcutaneous injection, twice daily) for 4 weeks in a rat model of myocardial infarction (MI) with coronary artery occlusion. At the 5th week, electrocardiogram, monophasic action potentials and autonomic nerve function were evaluated. Cardiac tyrosine hydroxylase (TH) was determined by immunofluorescence staining. RESULTS: MI significantly increased sympathetic nerve activity (SNA) and ventricular arrhythmias, and prolonged APD dispersion and APD alternans (p < 0.01). Ghrelin treatment significantly increased ventricular fibrillation threshold (VFT), shortened APD dispersion and APD alternans, inhibited SNA and promoted vagus nerve activities (p < 0.01). Ghrelin also markedly reversed abnormal expression of TH in the peri-infarcted area of the heart (p < 0.01). DISCUSSION/CONCLUSION: Ghrelin provides a sustained electrophysiological protection by the increase of VFT and improvement of APD dispersion and APD alternans. The mechanism may be related to the regulation of autonomic nerve and sympathetic nerve remodeling. Thus, ghrelin represents a novel drug to prevent ventricular arrhythmia in ischemic heart disease.


Subject(s)
Arrhythmias, Cardiac/drug therapy , Cardiotonic Agents/pharmacology , Ghrelin/pharmacology , Myocardial Infarction/drug therapy , Action Potentials/drug effects , Animals , Arrhythmias, Cardiac/etiology , Autonomic Pathways/drug effects , Cardiotonic Agents/therapeutic use , Disease Models, Animal , Electrocardiography/drug effects , Ghrelin/therapeutic use , Male , Myocardial Infarction/complications , Rats, Sprague-Dawley , Sympathetic Nervous System/drug effects , Tyrosine 3-Monooxygenase/metabolism , Vagus Nerve/drug effects , Ventricular Fibrillation/drug therapy
12.
Diabetes ; 71(2): 249-263, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34732538

ABSTRACT

The increasing prevalence of obesity has resulted in demands for the development of new effective strategies for obesity treatment. Withaferin A (WA) shows a great potential for prevention of obesity by sensitizing leptin signaling in the hypothalamus. However, the mechanism underlying the weight- and adiposity-reducing effects of WA remains to be elucidated. In this study, we report that WA treatment induced white adipose tissue (WAT) browning, elevated energy expenditure, decreased respiratory exchange ratio, and prevented high-fat diet-induced obesity. The sympathetic chemical denervation dampened the WAT browning and also impeded the reduction of adiposity in WA-treated mice. WA markedly upregulated the levels of Prdm16 and FATP1 (Slc27a1) in the inguinal WAT (iWAT), and this was blocked by sympathetic denervation. Prdm16 or FATP1 knockdown in iWAT abrogated the WAT browning-inducing effects of WA and restored the weight gain and adiposity in WA-treated mice. Together, these findings suggest that WA induces WAT browning through the sympathetic nerve-adipose axis, and the adipocytic Prdm16-FATP1 pathway mediates the promotive effects of WA on white adipose browning.


Subject(s)
Adipose Tissue, Brown/drug effects , Adipose Tissue, White/drug effects , Obesity/prevention & control , Withanolides/pharmacology , Adipose Tissue, Brown/innervation , Adipose Tissue, Brown/physiology , Adipose Tissue, White/innervation , Adipose Tissue, White/physiology , Animals , Cell Transdifferentiation/drug effects , Cell Transdifferentiation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diet, High-Fat , Fatty Acid Transport Proteins/genetics , Fatty Acid Transport Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/genetics , Obesity/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Psychophysiology ; 59(2): e13955, 2022 02.
Article in English | MEDLINE | ID: mdl-34665890

ABSTRACT

Cannabis is the most commonly used psychotropic drug in the United States, after alcohol. Despite its apparent sedative and calming effects, cannabis and its main psychoactive constituent, ∆9 -tetrahydrocannabinol (THC) can produce serious adverse effects including tachycardia and anxiety. These effects can be especially pronounced in women, who remain underrepresented in clinical cannabinoid research. The present study is one of the first to characterize the effects of single doses of oral THC on autonomic nervous system function in healthy adult women. Occasional female cannabis users participated in three laboratory sessions in which they received oral THC (7.5 and 15 mg) and placebo. Autonomic measures included heart rate (HR), blood pressure (BP), pre-ejection period (PEP) a measure of cardiac sympathetic functioning, and high frequency heart rate variability (HF-HRV) a measure of parasympathetic cardiac control. Autonomic responses were examined in relation to subjective drug effects. THC dose-dependently increased HR, decreased HF-HRV, and increased ratings of feeling a drug effect, cannabis-like intoxication, and anxiety. Although the drug did not significantly affect BP or PEP, HR was negatively related to both PEP and HF-HRV. HF-HRV, the measure of parasympathetic activity, was significantly negatively related to subjective measures of cannabis intoxication (but not anxiety) at the 15 mg dose only. PEP was not significantly related to any subjective measure. These results extend our knowledge of the autonomic effects of THC in relation to subjective drug experience. This and future studies will help us to understand risk factors related to cannabis use.


Subject(s)
Anxiety/chemically induced , Blood Pressure/drug effects , Cannabinoid Receptor Agonists/pharmacology , Dronabinol/pharmacology , Heart Rate/drug effects , Parasympathetic Nervous System/drug effects , Sympathetic Nervous System/drug effects , Administration, Oral , Adult , Cannabinoid Receptor Agonists/administration & dosage , Cannabinoid Receptor Agonists/adverse effects , Dronabinol/administration & dosage , Dronabinol/adverse effects , Female , Humans , Young Adult
14.
J Pharmacol Sci ; 148(1): 86-92, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34924134

ABSTRACT

To clarify the effects of long-term administration of SGLT2 inhibitor, a hypoglycemic agent, on basal sympathetic nerve activity (SNA) and on SNA under development of insulin resistance, we measured peripheral SNA in response to a glucose load in standard chow- (SCF) and high-fat-fed (HFF) rats treated with or without dapagliflozin for 7 weeks. We conducted an intravenous glucose administration (IVGA), and evaluated SNA microneurographically recorded in the unilateral sciatic nerve. Dapagliflozin did not affect the steady state action potential (AP) rate just before the IVGA (baseline) in both the SCF and HFF rats. After the IVGA, in the SCF rats, the AP rate in dapagliflozin-treated group transiently decreased within 20 min after the IVGA, and was significantly lower (P < 0.05) than non-treated group for 60 min. In the HFF rats, no significant difference was seen in the AP rate between dapagliflozin-treated and non-treated groups. The rate in the dapagliflozin-treated group after the IVGA was significantly lower (P < 0.05) than the baseline whereas such difference was not found in the non-treated group. In conclusion, dapagliflozin attenuate SNA in response to glucose load, and that the SNA response is different between standard chow-fed- and high-fat-fed rats.


Subject(s)
Action Potentials/drug effects , Animal Feed/standards , Benzhydryl Compounds/pharmacology , Diet, High-Fat/adverse effects , Glucose/administration & dosage , Glucose/pharmacology , Glucosides/pharmacology , Hypoglycemic Agents/pharmacology , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiology , Administration, Intravenous , Animals , Glucose/metabolism , Insulin Resistance/physiology , Male , Rats, Wistar , Sciatic Nerve/drug effects , Sciatic Nerve/physiology , Time Factors
15.
Am J Physiol Regul Integr Comp Physiol ; 322(1): R64-R76, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34851729

ABSTRACT

Our knowledge about how low-dose (analgesic) fentanyl affects autonomic cardiovascular regulation is primarily limited to animal experiments. Notably, it is unknown if low-dose fentanyl influences human autonomic cardiovascular responses during painful stimuli in humans. Therefore, we tested the hypothesis that low-dose fentanyl reduces perceived pain and subsequent sympathetic and cardiovascular responses in humans during an experimental noxious stimulus. Twenty-three adults (10 females/13 males; 27 ± 7 yr; 26 ± 3 kg·m-2, means ± SD) completed this randomized, crossover, placebo-controlled trial during two laboratory visits. During each visit, participants completed a cold pressor test (CPT; hand in ∼0.4°C ice bath for 2 min) before and 5 min after drug/placebo administration (75 µg fentanyl or saline). We compared pain perception (100-mm visual analog scale), muscle sympathetic nerve activity (MSNA; microneurography, 11 paired recordings), and beat-to-beat blood pressure (BP; photoplethysmography) between trials (at both pre- and postdrug/placebo timepoints) using paired, two-tailed t tests. Before drug/placebo administration, perceived pain (P = 0.8287), ΔMSNA burst frequency (P = 0.7587), and Δmean BP (P = 0.8649) during the CPT were not different between trials. After the drug/placebo administration, fentanyl attenuated perceived pain (36 vs. 66 mm, P < 0.0001), ΔMSNA burst frequency (9 vs. 17 bursts/min, P = 0.0054), and Δmean BP (7 vs. 13 mmHg, P = 0.0174) during the CPT compared with placebo. Fentanyl-induced reductions in pain perception and Δmean BP were moderately related (r = 0.40, P = 0.0641). These data provide valuable information regarding how low-dose fentanyl reduces autonomic cardiovascular responses during an experimental painful stimulus.


Subject(s)
Analgesics, Opioid/administration & dosage , Blood Pressure/drug effects , Cardiovascular System/innervation , Fentanyl/administration & dosage , Muscle, Skeletal/innervation , Pain Perception/drug effects , Pain Threshold/drug effects , Pain/drug therapy , Sympathetic Nervous System/drug effects , Adult , Analgesics, Opioid/adverse effects , Cold Temperature , Cross-Over Studies , Female , Fentanyl/adverse effects , Humans , Immersion , Male , Pain/physiopathology , Pain/psychology , Sympathetic Nervous System/physiopathology , Time Factors , Water , Young Adult
16.
Am J Physiol Regul Integr Comp Physiol ; 322(1): R55-R63, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34851734

ABSTRACT

Hemorrhage is a leading cause of battlefield and civilian trauma deaths. Several pain medications, including fentanyl, are recommended for use in the prehospital (i.e., field setting) for a hemorrhaging solider. However, it is unknown whether fentanyl impairs arterial blood pressure (BP) regulation, which would compromise hemorrhagic tolerance. Thus, the purpose of this study was to test the hypothesis that an analgesic dose of fentanyl impairs hemorrhagic tolerance in conscious humans. Twenty-eight volunteers (13 females) participated in this double-blinded, randomized, placebo-controlled trial. We conducted a presyncopal limited progressive lower body negative pressure test (LBNP; a validated model to simulate hemorrhage) following intravenous administration of fentanyl (75 µg) or placebo (saline). We quantified tolerance as a cumulative stress index (mmHg·min), which was compared between trials using a paired, two-tailed t test. We also compared muscle sympathetic nerve activity (MSNA; microneurography) and beat-to-beat BP (photoplethysmography) during the LBNP test using a mixed effects model [time (LBNP stage) × trial]. LBNP tolerance was not different between trials (fentanyl: 647 ± 386 vs. placebo: 676 ± 295 mmHg·min, P = 0.61, Cohen's d = 0.08). Increases in MSNA burst frequency (time: P < 0.01, trial: P = 0.29, interaction: P = 0.94) and reductions in mean BP (time: P < 0.01, trial: P = 0.50, interaction: P = 0.16) during LBNP were not different between trials. These data, the first to be obtained in conscious humans, demonstrate that administration of an analgesic dose of fentanyl does not alter MSNA or BP during profound central hypovolemia, nor does it impair tolerance to this simulated hemorrhagic insult.


Subject(s)
Analgesics, Opioid/administration & dosage , Blood Pressure/drug effects , Cardiovascular System/innervation , Fentanyl/administration & dosage , Hemorrhage/physiopathology , Hypovolemia/physiopathology , Muscle, Skeletal/innervation , Sympathetic Nervous System/drug effects , Adult , Analgesics, Opioid/adverse effects , Cross-Over Studies , Double-Blind Method , Female , Fentanyl/adverse effects , Hemorrhage/diagnosis , Humans , Hypovolemia/diagnosis , Infusions, Intravenous , Lower Body Negative Pressure , Male , Middle Aged , Sympathetic Nervous System/physiopathology , Young Adult
17.
Nutrients ; 13(12)2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34959764

ABSTRACT

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.


Subject(s)
Adipose Tissue/metabolism , Flavonoids/administration & dosage , Sympathetic Nervous System/drug effects , Administration, Oral , Animals , Catecholamines/urine , Membrane Proteins/metabolism , Mice , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Positive Regulatory Domain I-Binding Factor 1/metabolism , Thermogenesis/drug effects , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Uncoupling Protein 1/metabolism
18.
Front Immunol ; 12: 749291, 2021.
Article in English | MEDLINE | ID: mdl-34867978

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a causative virus in the development of coronavirus disease 2019 (Covid-19) pandemic. Respiratory manifestations of SARS-CoV-2 infection such as acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) leads to hypoxia, oxidative stress, and sympatho-activation and in severe cases leads to sympathetic storm (SS). On the other hand, an exaggerated immune response to the SARS-CoV-2 invasion may lead to uncontrolled release of pro-inflammatory cytokine development of cytokine storm (CS). In Covid-19, there are interactive interactions between CS and SS in the development of multi-organ failure (MOF). Interestingly, cutting the bridge between CS and SS by anti-inflammatory and anti-adrenergic agents may mitigate complications that are induced by SARS-CoV-2 infection in severely affected Covid-19 patients. The potential mechanisms of SS in Covid-19 are through different pathways such as hypoxia, which activate the central sympathetic center through carotid bodies chemosensory input and induced pro-inflammatory cytokines, which cross the blood-brain barrier and activation of the sympathetic center. ß2-receptors signaling pathway play a crucial role in the production of pro-inflammatory cytokines, macrophage activation, and B-cells for the production of antibodies with inflammation exacerbation. ß-blockers have anti-inflammatory effects through reduction release of pro-inflammatory cytokines with inhibition of NF-κB. In conclusion, ß-blockers interrupt this interaction through inhibition of several mediators of CS and SS with prevention development of neural-cytokine loop in SARS-CoV-2 infection. Evidence from this study triggers an idea for future prospective studies to confirm the potential role of ß-blockers in the management of Covid-19.


Subject(s)
Adrenergic beta-Antagonists/therapeutic use , COVID-19 Drug Treatment , Cytokine Release Syndrome/drug therapy , Sympathetic Nervous System/drug effects , Anti-Inflammatory Agents/therapeutic use , COVID-19/complications , COVID-19/metabolism , COVID-19/physiopathology , Catecholamines/metabolism , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/metabolism , Cytokine Release Syndrome/physiopathology , Cytokines/metabolism , Humans , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/physiopathology , SARS-CoV-2/pathogenicity , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/physiopathology
19.
Circ Heart Fail ; 14(12): e008365, 2021 12.
Article in English | MEDLINE | ID: mdl-34789005

ABSTRACT

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.


Subject(s)
Heart Failure/metabolism , Heart Failure/physiopathology , Kidney/innervation , Sodium-Glucose Transporter 2/metabolism , Animals , Glucose/metabolism , Kidney/drug effects , Kidney/metabolism , Male , Rats, Sprague-Dawley , Sodium/metabolism , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiopathology
20.
Pflugers Arch ; 473(12): 1925-1938, 2021 12.
Article in English | MEDLINE | ID: mdl-34596752

ABSTRACT

Here we investigate mechanisms underlying spontaneous phasic contractions (SPCs) and sympathetic control of contractility in the rat epididymis, a long tubular duct involved in transportation and maturation of sperm. Longitudinal contractions of short segments (~ 1.5 mm) of rat proximal and distal caudal epididymal duct were measured + / - nerve stimulation. The extent of sympathetic innervation of these duct regions was determined by immunohistochemistry. Proximal caudal duct segments (150-300 µm dia.) exhibited SPCs, while distal segments (350-500 µm) were quiescent in ~ 80% of preparations. SPC amplitude and frequency were reduced by the L-type voltage-dependent Ca2+ channel (LVDCC) blocker nifedipine (1 µM), with the T-type voltage-dependent Ca2+ channel (TVDCC) blocker ML218 (1 µM) specifically decreasing SPC frequency. SPCs were inhibited upon blockade of the SR/ER Ca2+-ATPase (CPA 10 µM). SPCs were also inhibited by caffeine (1 µM), 2-APB (100 µM), niflumic acid (100 µM), or by lowering extracellular [Cl-] from 134.4 to 12.4 mM but not by ryanodine (25 µM) or tetracaine (100 µM). Electrical field stimulation (EFS) at 2 Hz for 60 s caused a sustained α1-adrenoceptor-sensitive contraction in distal segments and enhanced and/or induced α2-adrenoceptor-sensitive oscillatory phasic contractions in proximal and distal segments, the latter mimicked by application of the α2-adrenoceptor agonist clonidine. We hypothesise that SPCs in the proximal cauda are triggered by pacemaker mechanisms involving rhythmic IP3 receptor-operated SR/ER store Ca2+ release and resultant activation of CaCC with TVDCCs and possibly LVDCCs subserving in this process. Sympathetic nerve-released noradrenaline induces α2-adrenoceptor-mediated phasic contractions in the proximal and distal cauda. These findings provide new pharmacological targets for male infertility and contraception.


Subject(s)
Epididymis/physiology , Muscle Contraction/physiology , Muscle, Smooth/physiology , Sympathetic Nervous System/physiology , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Epididymis/drug effects , Epididymis/metabolism , Male , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Nifedipine/pharmacology , Norepinephrine/pharmacology , Phenylephrine/pharmacology , Rats , Rats, Wistar , Ryanodine/pharmacology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...