Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.884
Filter
Add more filters

Publication year range
1.
Immunity ; 49(1): 93-106.e7, 2018 07 17.
Article in English | MEDLINE | ID: mdl-29958804

ABSTRACT

There is a growing body of research on the neural control of immunity and inflammation. However, it is not known whether the nervous system can regulate the production of inflammatory myeloid cells from hematopoietic progenitor cells in disease conditions. Myeloid cell numbers in diabetic patients were strongly correlated with plasma concentrations of norepinephrine, suggesting the role of sympathetic neuronal activation in myeloid cell production. The spleens of diabetic patients and mice contained higher numbers of tyrosine hydroxylase (TH)-expressing leukocytes that produced catecholamines. Granulocyte macrophage progenitors (GMPs) expressed the ß2 adrenergic receptor, a target of catecholamines. Ablation of splenic sympathetic neuronal signaling using surgical, chemical, and genetic approaches diminished GMP proliferation and myeloid cell development. Finally, mice lacking TH-producing leukocytes had reduced GMP proliferation, resulting in diminished myelopoiesis. Taken together, our study demonstrates that catecholamines produced by leukocytes and sympathetic nerve termini promote GMP proliferation and myeloid cell development.


Subject(s)
Diabetes Mellitus/physiopathology , Granulocyte-Macrophage Progenitor Cells/cytology , Granulocyte-Macrophage Progenitor Cells/metabolism , Myelopoiesis , Neuroimmunomodulation , Sympathetic Nervous System/metabolism , Adrenergic beta-2 Receptor Antagonists/pharmacology , Animals , Cell Proliferation/drug effects , Diabetes Mellitus/blood , Disease Models, Animal , Female , Humans , Leukocytes/enzymology , Leukocytes/metabolism , Male , Mice , Myeloid Cells/cytology , Myelopoiesis/drug effects , Neuroimmunomodulation/drug effects , Norepinephrine/blood , Signal Transduction/drug effects , Spleen/cytology , Spleen/innervation , Spleen/metabolism , Sympathetic Nervous System/drug effects
2.
Nature ; 589(7843): 591-596, 2021 01.
Article in English | MEDLINE | ID: mdl-33361809

ABSTRACT

Haematopoietic stem cells (HSCs) reside in specialized microenvironments in the bone marrow-often referred to as 'niches'-that represent complex regulatory milieux influenced by multiple cellular constituents, including nerves1,2. Although sympathetic nerves are known to regulate the HSC niche3-6, the contribution of nociceptive neurons in the bone marrow remains unclear. Here we show that nociceptive nerves are required for enforced HSC mobilization and that they collaborate with sympathetic nerves to maintain HSCs in the bone marrow. Nociceptor neurons drive granulocyte colony-stimulating factor (G-CSF)-induced HSC mobilization via the secretion of calcitonin gene-related peptide (CGRP). Unlike sympathetic nerves, which regulate HSCs indirectly via the niche3,4,6, CGRP acts directly on HSCs via receptor activity modifying protein 1 (RAMP1) and the calcitonin receptor-like receptor (CALCRL) to promote egress by activating the Gαs/adenylyl cyclase/cAMP pathway. The ingestion of food containing capsaicin-a natural component of chili peppers that can trigger the activation of nociceptive neurons-significantly enhanced HSC mobilization in mice. Targeting the nociceptive nervous system could therefore represent a strategy to improve the yield of HSCs for stem cell-based therapeutic agents.


Subject(s)
Autonomic Pathways , Cell Movement , Hematopoietic Stem Cells/cytology , Nociception/physiology , Nociceptors/physiology , Sympathetic Nervous System/cytology , Adenylyl Cyclases/metabolism , Animals , Autonomic Pathways/drug effects , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Receptor-Like Protein/metabolism , Capsaicin/pharmacology , Cell Movement/drug effects , Cyclic AMP/metabolism , Female , GTP-Binding Protein alpha Subunits, Gs/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Nociception/drug effects , Nociceptors/drug effects , Receptor Activity-Modifying Protein 1/metabolism , Signal Transduction/drug effects , Stem Cell Niche , Sympathetic Nervous System/drug effects
3.
J Neurosci ; 44(21)2024 May 22.
Article in English | MEDLINE | ID: mdl-38565292

ABSTRACT

Glucagon-like peptide-1 (GLP-1) and its analogs are widely used for diabetes treatment. The paraventricular nucleus (PVN) is crucial for regulating cardiovascular activity. This study aims to determine the roles of GLP-1 and its receptors (GLP-1R) in the PVN in regulating sympathetic outflow and blood pressure. Experiments were carried out in male normotensive rats and spontaneously hypertensive rats (SHR). Renal sympathetic nerve activity (RSNA) and mean arterial pressure (MAP) were recorded. GLP-1 and GLP-1R expressions were present in the PVN. PVN microinjection of GLP-1R agonist recombinant human GLP-1 (rhGLP-1) or EX-4 increased RSNA and MAP, which were prevented by GLP-1R antagonist exendin 9-39 (EX9-39) or GLP-1R antagonist 1, superoxide scavenger tempol, antioxidant N-acetylcysteine, NADPH oxidase (NOX) inhibitor apocynin, adenylyl cyclase (AC) inhibitor SQ22536 or protein kinase A (PKA) inhibitor H89. PVN microinjection of rhGLP-1 increased superoxide production, NADPH oxidase activity, cAMP level, AC, and PKA activity, which were prevented by SQ22536 or H89. GLP-1 and GLP-1R were upregulated in the PVN of SHR. PVN microinjection of GLP-1 agonist increased RSNA and MAP in both WKY and SHR, but GLP-1 antagonists caused greater effects in reducing RSNA and MAP in SHR than in WKY. The increased superoxide production and NADPH oxidase activity in the PVN of SHR were augmented by GLP-1R agonists but attenuated by GLP-1R antagonists. These results indicate that activation of GLP-1R in the PVN increased sympathetic outflow and blood pressure via cAMP-PKA-mediated NADPH oxidase activation and subsequent superoxide production. GLP-1 and GLP-1R upregulation in the PVN partially contributes to sympathetic overactivity and hypertension.


Subject(s)
Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor , Hypertension , Paraventricular Hypothalamic Nucleus , Rats, Inbred SHR , Sympathetic Nervous System , Animals , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Male , Hypertension/physiopathology , Hypertension/metabolism , Rats , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiology , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Blood Pressure/drug effects , Blood Pressure/physiology , Rats, Inbred WKY , Rats, Sprague-Dawley
4.
J Physiol ; 602(14): 3505-3518, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38743485

ABSTRACT

NaV1.7 plays a crucial role in inducing and conducting action potentials in pain-transducing sensory nociceptor fibres, suggesting that NaV1.7 blockers could be effective non-opioid analgesics. While SCN9A is expressed in both sensory and autonomic neurons, its functional role in the autonomic system remains less established. Our single neuron rt-PCR analysis revealed that 82% of sympathetic neurons isolated from guinea-pig stellate ganglia expressed NaV1.7 mRNA, with NaV1.3 being the only other tetrodotoxin-sensitive channel expressed in approximately 50% of neurons. We investigated the role of NaV1.7 in conducting action potentials in postganglionic sympathetic nerves and in the sympathetic adrenergic contractions of blood vessels using selective NaV1.7 inhibitors. Two highly selective NaV1.7 blockers, GNE8493 and PF 05089771, significantly inhibited postganglionic compound action potentials by approximately 70% (P < 0.01), with residual activity being blocked by the NaV1.3 inhibitor, ICA 121431. Electrical field stimulation (EFS) induced rapid contractions in guinea-pig isolated aorta, pulmonary arteries, and human isolated pulmonary arteries via stimulation of intrinsic nerves, which were inhibited by prazosin or the NaV1 blocker tetrodotoxin. Our results demonstrated that blocking NaV1.7 with GNE8493, PF 05089771, or ST2262 abolished or strongly inhibited sympathetic adrenergic responses in guinea-pigs and human vascular smooth muscle. These findings support the hypothesis that pharmacologically inhibiting NaV1.7 could potentially reduce sympathetic and parasympathetic function in specific vascular beds and airways. KEY POINTS: 82% of sympathetic neurons isolated from the stellate ganglion predominantly express NaV1.7 mRNA. NaV1.7 blockers inhibit action potential conduction in postganglionic sympathetic nerves. NaV1.7 blockade substantially inhibits sympathetic nerve-mediated adrenergic contractions in human and guinea-pig blood vessels. Pharmacologically blocking NaV1.7 profoundly affects sympathetic and parasympathetic responses in addition to sensory fibres, prompting exploration into the broader physiological consequences of NaV1.7 mutations on autonomic nerve activity.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel , Animals , Guinea Pigs , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.7 Voltage-Gated Sodium Channel/physiology , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Humans , Male , Action Potentials/drug effects , Action Potentials/physiology , Sympathetic Fibers, Postganglionic/physiology , Sympathetic Fibers, Postganglionic/drug effects , Female , Arteries/physiology , Arteries/drug effects , Arteries/innervation , Sodium Channel Blockers/pharmacology , Stellate Ganglion/physiology , Sympathetic Nervous System/physiology , Sympathetic Nervous System/drug effects
5.
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
6.
J Physiol ; 602(16): 4053-4071, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39058701

ABSTRACT

The present study investigated the impact of central α2-adrenergic mechanisms on sympathetic action potential (AP) discharge, recruitment and latency strategies. We used the microneurographic technique to record muscle sympathetic nerve activity and a continuous wavelet transform to investigate postganglionic sympathetic AP firing during a baseline condition and an infusion of a α2-adrenergic receptor agonist, dexmedetomidine (10 min loading infusion of 0.225 µg kg-1; maintenance infusion of 0.1-0.5 µg kg h-1) in eight healthy individuals (28 ± 7 years, five females). Dexmedetomidine reduced mean pressure (92 ± 7 to 80 ± 8 mmHg, P < 0.001) but did not alter heart rate (61 ± 13 to 60 ± 14 bpm; P = 0.748). Dexmedetomidine reduced sympathetic AP discharge (126 ± 73 to 27 ± 24 AP 100 beats-1, P = 0.003) most strongly for medium-sized APs (normalized cluster 2: 21 ± 10 to 5 ± 5 AP 100 beats-1; P < 0.001). Dexmedetomidine progressively de-recruited sympathetic APs beginning with the largest AP clusters (12 ± 3 to 7 ± 2 clusters, P = 0.002). Despite de-recruiting large AP clusters with shorter latencies, dexmedetomidine reduced AP latency across remaining clusters (1.18 ± 0.12 to 1.13 ± 0.13 s, P = 0.002). A subset of six participants performed a Valsalva manoeuvre (20 s, 40 mmHg) during baseline and the dexmedetomidine infusion. Compared to baseline, AP discharge (Δ 361 ± 292 to Δ 113 ± 155 AP 100 beats-1, P = 0.011) and AP cluster recruitment elicited by the Valsalva manoeuvre were lower during dexmedetomidine (Δ 2 ± 1 to Δ 0 ± 2 AP clusters, P = 0.041). The reduction in sympathetic AP latency elicited by the Valsalva manoeuvre was not affected by dexmedetomidine (Δ -0.09 ± 0.07 to Δ -0.07 ± 0.14 s, P = 0.606). Dexmedetomidine reduced baroreflex gain, most strongly for medium-sized APs (normalized cluster 2: -6.0 ± 5 to -1.6 ± 2 % mmHg-1; P = 0.008). These data suggest that α2-adrenergic mechanisms within the central nervous system modulate sympathetic postganglionic neuronal discharge, recruitment and latency strategies in humans. KEY POINTS: Sympathetic postganglionic neuronal subpopulations innervating the human circulation exhibit complex patterns of discharge, recruitment and latency. However, the central neural mechanisms governing sympathetic postganglionic discharge remain unclear. This microneurographic study investigated the impact of a dexmedetomidine infusion (α2-adrenergic receptor agonist) on muscle sympathetic postganglionic action potential (AP) discharge, recruitment and latency patterns. Dexmedetomidine infusion inhibited the recruitment of large and fast conducting sympathetic APs and attenuated the discharge of medium sized sympathetic APs that fired during resting conditions and the Valsalva manoeuvre. Dexmedetomidine infusion elicited shorter sympathetic AP latencies during resting conditions but did not affect the reductions in latency that occurred during the Valsalva manoeuvre. These data suggest that α2-adrenergic mechanisms within the central nervous system modulate sympathetic postganglionic neuronal discharge, recruitment and latency strategies in humans.


Subject(s)
Action Potentials , Adrenergic alpha-2 Receptor Agonists , Dexmedetomidine , Sympathetic Nervous System , Humans , Dexmedetomidine/pharmacology , Female , Adult , Male , Adrenergic alpha-2 Receptor Agonists/pharmacology , Sympathetic Nervous System/physiology , Sympathetic Nervous System/drug effects , Action Potentials/drug effects , Action Potentials/physiology , Young Adult , Heart Rate/drug effects , Heart Rate/physiology , Blood Pressure/physiology , Blood Pressure/drug effects , Muscle, Skeletal/physiology , Muscle, Skeletal/innervation , Muscle, Skeletal/drug effects , Receptors, Adrenergic, alpha-2/physiology , Receptors, Adrenergic, alpha-2/metabolism
7.
Am J Physiol Heart Circ Physiol ; 327(1): H000, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38787381

ABSTRACT

Systemic insulin increases muscle sympathetic nerve activity (MSNA) via both central actions within the brainstem and peripheral activation of the arterial baroreflex. Augmented MSNA during hyperinsulinemia likely restrains peripheral vasodilation and contributes to the maintenance of blood pressure (BP). However, in the absence of insulin action within the peripheral vasculature, whether central insulin stimulation increases MSNA and influences peripheral hemodynamics in humans remains unknown. Herein, we hypothesized intranasal insulin administration would increase MSNA and BP in healthy young adults. Participants were assigned to time control [TC, n = 13 (5 females/8 males), 28 ± 1 yr] or 160 IU of intranasal insulin administered over 5 min [n = 15 (5 females/10 males), 26 ± 2 yr]; five (1 female/4 males) participants completed both conditions. MSNA (fibular microneurography), BP (finger photoplethysmography), and leg blood flow (LBF, femoral Doppler ultrasound) were assessed at baseline, and 15 and 30 min following insulin administration. Leg vascular conductance [LVC = (LBF ÷ mean BP) × 100] was calculated. Venous insulin and glucose concentrations remained unchanged throughout (P > 0.05). Following intranasal insulin administration, MSNA (burst frequency; baseline = 100%; minute 15, 121 ± 8%; minute 30, 118 ± 6%; P = 0.009, n = 7) and mean BP (baseline = 100%; minute 15, 103 ± 1%; minute 30, 102 ± 1%; P = 0.003) increased, whereas LVC decreased (baseline = 100%; minute 15, 93 ± 3%; minute 30, 99 ± 3%; P = 0.03). In contrast, MSNA, mean BP, and LVC were unchanged in TC participants (P > 0.05). We provide the first evidence that intranasal insulin administration in healthy young adults acutely increases MSNA and BP and decreases LVC. These results enhance mechanistic understanding of the sympathetic and peripheral hemodynamic response to insulin.NEW & NOTEWORTHY Systemic insulin increases muscle sympathetic nerve activity (MSNA) via central actions within the brainstem and peripheral activation of the arterial baroreflex. In the absence of peripheral insulin action, whether central insulin stimulation increases MSNA and influences peripheral hemodynamics in humans was unknown. We provide the first evidence that intranasal insulin administration increases MSNA and blood pressure and reduces leg vascular conductance. These results enhance mechanistic understanding of the sympathetic and hemodynamic response to insulin.


Subject(s)
Administration, Intranasal , Insulin , Muscle, Skeletal , Sympathetic Nervous System , Humans , Male , Female , Insulin/administration & dosage , Insulin/blood , Sympathetic Nervous System/drug effects , Adult , Muscle, Skeletal/innervation , Muscle, Skeletal/blood supply , Muscle, Skeletal/drug effects , Blood Pressure/drug effects , Regional Blood Flow/drug effects , Blood Glucose/metabolism , Blood Glucose/drug effects , Healthy Volunteers , Young Adult , Baroreflex/drug effects
8.
Am J Physiol Heart Circ Physiol ; 326(6): H1337-H1349, 2024 06 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
9.
J Pharmacol Exp Ther ; 390(2): 240-249, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-38902033

ABSTRACT

Sympathetic hyperinnervation is the leading cause of fatal ventricular arrhythmia (VA) after myocardial infarction (MI). Cardiac mast cells cause arrhythmias directly through degranulation. However, the role and mechanism of mast cell degranulation in sympathetic remodeling remain unknown. We investigated the role of oxytocin (OT) in stabilizing cardiac mast cells and improving sympathetic innervation in rats. MI was induced by coronary artery ligation. Western blotting, immunofluorescence, and toluidine staining of mast cells were performed to determine the expression and location of target protein. Mast cells accumulated significantly in peri-infarcted tissues and were present in a degranulated state. They expressed OT receptor (OTR), and OT infusion reduced the number of degranulated cardiac mast cells post-MI. Sympathetic hyperinnervation was attenuated as assessed by immunofluorescence for tyrosine hydroxylase (TH). Seven days post-MI, the arrhythmia score of programmed electrical stimulation was higher in vehicle-treated rats with MI than in rats treated with OT. An in vitro study showed that OT stabilized mast cells via the phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) signaling pathway. Further in vivo studies on OTR-deficient mice showed worsening mast cell degranulation and worsening sympathetic innervation. OT pretreatment inhibited cardiac mast cell degranulation post-MI and prevented sympathetic hyperinnervation, along with mast cell stabilization via the PI3K/Akt pathway. SIGNIFICANCE STATEMENT: This is the first study to elucidate the role and mechanism of oxytocin (OT) in inflammatory-sympathetic communication mediated sympathetic hyperinnervation after myocardial infarction (MI), providing new approaches to prevent fatal arrhythmias.


Subject(s)
Cell Degranulation , Mast Cells , Myocardial Infarction , Oxytocin , Rats, Sprague-Dawley , Receptors, Oxytocin , Sympathetic Nervous System , Animals , Oxytocin/pharmacology , Oxytocin/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Mast Cells/drug effects , Mast Cells/metabolism , Rats , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , Male , Cell Degranulation/drug effects , Receptors, Oxytocin/metabolism , Receptors, Oxytocin/antagonists & inhibitors , Mice , Signal Transduction/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/etiology
10.
Brain Behav Immun ; 119: 105-119, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38548186

ABSTRACT

The sympathetic arm of the inflammatory reflex is the efferent pathway through which the central nervous system (CNS) can control peripheral immune responses. Diminazene aceturate (DIZE) is an antiparasitic drug that has been reported to exert protective effects on various experimental models of inflammation. However, the pathways by which DIZE promotes a protective immunomodulatory effects still need to be well established, and no studies demonstrate the capacity of DIZE to modulate a neural reflex to control inflammation. C57BL/6 male mice received intraperitoneal administration of DIZE (2 mg/Kg) followed by lipopolysaccharide (LPS, 5 mg/Kg, i.p.). Endotoxemic animals showed hyperresponsiveness to inflammatory signals, while those treated with DIZE promoted the activation of the inflammatory reflex to attenuate the inflammatory response during endotoxemia. The unilateral cervical vagotomy did not affect the anti-inflammatory effect of DIZE in the spleen and serum. At the same time, splenic denervation attenuated tumor necrosis factor (TNF) synthesis in the spleen and serum. Using broad-spectrum antibiotics for two weeks showed that LPS modulated the microbiota to induce a pro-inflammatory profile in the intestine and reduced the serum concentration of tryptophan and serotonin (5-HT), while DIZE restored serum tryptophan and increased the hypothalamic 5-HT levels. Furthermore, the treatment with 4-Chloro-DL-phenylalanine (pcpa, an inhibitor of 5-HT synthesis) abolished the anti-inflammatory effects of the DIZE in the spleen. Our results indicate that DIZE promotes microbiota modulation to increase central 5-HT levels and activates the efferent sympathetic arm of the inflammatory reflex to control splenic TNF production in endotoxemic mice.


Subject(s)
Diminazene , Endotoxemia , Gastrointestinal Microbiome , Inflammation , Lipopolysaccharides , Mice, Inbred C57BL , Serotonin , Spleen , Sympathetic Nervous System , Animals , Male , Mice , Spleen/metabolism , Spleen/drug effects , Diminazene/analogs & derivatives , Diminazene/pharmacology , Lipopolysaccharides/pharmacology , Gastrointestinal Microbiome/drug effects , Inflammation/metabolism , Serotonin/metabolism , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , Endotoxemia/metabolism , Brain/metabolism , Brain/drug effects , Brain-Gut Axis/drug effects , Anti-Inflammatory Agents/pharmacology , Tumor Necrosis Factor-alpha/metabolism
11.
J Cardiovasc Pharmacol ; 84(2): 227-238, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39115721

ABSTRACT

ABSTRACT: Previous studies have found that anxiety disorders may increase the incidence of atrial fibrillation (AF). More and more studies have shown that α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) are involved in the occurrence and development of cardiovascular diseases. However, the role of AMPARs in AF associated with anxiety disorder remains unclear. The aim of this study was to investigate the effect of AMPARs on AF susceptibility in rats with anxiety disorder and its possible mechanism. The anxiety disorder rat model was established by unpredictable empty bottle stimulation and was treated with AMPARs agonist and antagonist. Our results showed that AMPARs antagonist treatment significantly reduced sympathetic activity, improved heart rate variability, shortened action potential duration, prolonged effective refractory period, reduced AF induction rate, and improved cardiac electrical remodeling and the expression of inflammatory factors. In addition, inhibition of AMPARs reduced the phosphorylation of IκBα and p65. Our experimental results suggest that inhibition of AMPARs can reduce autonomic remodeling, improve atrial electrical remodeling, and suppress myocardial inflammation, which provides a potential therapeutic strategy for the treatment of AF associated with anxiety disorder.


Subject(s)
Anxiety Disorders , Atrial Fibrillation , Disease Models, Animal , Heart Atria , Rats, Sprague-Dawley , Receptors, AMPA , Animals , Atrial Fibrillation/physiopathology , Atrial Fibrillation/drug therapy , Atrial Fibrillation/metabolism , Male , Anxiety Disorders/drug therapy , Anxiety Disorders/metabolism , Anxiety Disorders/physiopathology , Heart Atria/drug effects , Heart Atria/physiopathology , Heart Atria/metabolism , Heart Atria/pathology , Receptors, AMPA/metabolism , Atrial Remodeling/drug effects , Heart Rate/drug effects , Inflammation Mediators/metabolism , Action Potentials/drug effects , Phosphorylation , Signal Transduction , Sympathetic Nervous System/physiopathology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , Transcription Factor RelA/metabolism , Rats , Anti-Inflammatory Agents/pharmacology , Refractory Period, Electrophysiological/drug effects , NF-KappaB Inhibitor alpha/metabolism
12.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Article in English | MEDLINE | ID: mdl-33972441

ABSTRACT

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.


Subject(s)
Immune System/innervation , Immunomodulation/drug effects , Spleen/immunology , Sympathetic Nervous System/immunology , Vagus Nerve/immunology , Animals , Female , Gene Expression , Humans , Immune System/drug effects , Inflammation , Interleukin-6/genetics , Interleukin-6/immunology , Lipopolysaccharides/pharmacology , Mice , Microcirculation/drug effects , Microcirculation/genetics , Microcirculation/immunology , Norepinephrine/pharmacology , Rats , Species Specificity , Spleen/drug effects , Spleen/innervation , Spleen/pathology , Swine , Sympathetic Nervous System/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Vagus Nerve/drug effects , Vagus Nerve Stimulation/methods
13.
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
14.
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
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.
Circ Res ; 127(2): e1-e13, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32268833

ABSTRACT

RATIONALE: Chronic exposure to hypoxia is associated with elevated sympathetic nervous activity and reduced vascular function in lowlanders, and Andean highlanders suffering from excessive erythrocytosis (EE); however, the mechanistic link between chronically elevated sympathetic nervous activity and hypoxia-induced vascular dysfunction has not been determined. OBJECTIVE: To determine the impact of heightened sympathetic nervous activity on resistance artery endothelial-dependent dilation (EDD), and endothelial-independent dilation, in lowlanders and Andean highlanders with and without EE. METHODS AND RESULTS: We tested healthy lowlanders (n=9) at sea level (344 m) and following 14 to 21 days at high altitude (4300 m), and permanent Andean highlanders with (n=6) and without (n=9) EE at high altitude. Vascular function was assessed using intraarterial infusions (3 progressive doses) of acetylcholine (ACh; EDD) and sodium nitroprusside (endothelial-independent dilation) before and after local α+ß adrenergic receptor blockade (phentolamine and propranolol). Intraarterial blood pressure, heart rate, and simultaneous brachial artery diameter and blood velocity were recorded at rest and during drug infusion. Changes in forearm vascular conductance were calculated. The main findings were (1) chronic hypoxia reduced EDD in lowlanders (changes in forearm vascular conductance from sea level: ACh1: -52.7±19.6%, ACh2: -25.4±38.7%, ACh3: -35.1±34.7%, all P≤0.02); and in Andeans with EE compared with non-EE (changes in forearm vascular conductance at ACh3: -36.4%, P=0.007). Adrenergic blockade fully restored EDD in lowlanders at high altitude, and normalized EDD between EE and non-EE Andeans. (2) Chronic hypoxia had no effect on endothelial-independent dilation in lowlanders, and no differences were detected between EE and non-EE Andeans; however, EID was increased in the non-EE Andeans after adrenergic blockade (P=0.012), but this effect was not observed in the EE Andeans. CONCLUSIONS: These data indicate that chronic hypoxia reduces EDD via heightened α-adrenergic signaling in lowlanders and in Andeans with EE. These vascular mechanisms have important implications for understanding the physiological consequences of acute and chronic high altitude adaptation.


Subject(s)
Adaptation, Physiological , Altitude Sickness/metabolism , Polycythemia/metabolism , Receptors, Adrenergic/metabolism , Vasodilation , Acetylcholine/metabolism , Acetylcholine/pharmacology , Adrenergic Agents/pharmacology , Adult , Altitude , Altitude Sickness/blood , Altitude Sickness/physiopathology , Blood Pressure , Blood Vessels/drug effects , Blood Vessels/metabolism , Blood Vessels/physiopathology , Heart Rate , Humans , Male , Nitroprusside/pharmacology , Phentolamine/pharmacology , Polycythemia/etiology , Polycythemia/physiopathology , Propranolol/pharmacology , Signal Transduction , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/physiopathology , Vasodilator Agents/pharmacology
17.
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
18.
Cereb Cortex ; 31(9): 4115-4139, 2021 07 29.
Article in English | MEDLINE | ID: mdl-34003210

ABSTRACT

Noradrenaline (NA) in the thalamus has important roles in physiological, pharmacological, and pathological neuromodulation. In this work, a complete characterization of NA axons and Alpha adrenoceptors distributions is provided. NA axons, revealed by immunohistochemistry against the synthesizing enzyme and the NA transporter, are present in all thalamic nuclei. The most densely innervated ones are the midline nuclei, intralaminar nuclei (paracentral and parafascicular), and the medial sector of the mediodorsal nucleus (MDm). The ventral motor nuclei and most somatosensory relay nuclei receive a moderate NA innervation. The pulvinar complex receives a heterogeneous innervation. The lateral geniculate nucleus (GL) has the lowest NA innervation. Alpha adrenoceptors were analyzed by in vitro quantitative autoradiography. Alpha-1 receptor densities are higher than Alpha-2 densities. Overall, axonal densities and Alpha adrenoceptor densities coincide; although some mismatches were identified. The nuclei with the highest Alpha-1 values are MDm, the parvocellular part of the ventral posterior medial nucleus, medial pulvinar, and midline nuclei. The nucleus with the lowest Alpha-1 receptor density is GL. Alpha-2 receptor densities are highest in the lateral dorsal, centromedian, medial and inferior pulvinar, and midline nuclei. These results suggest a role for NA in modulating thalamic involvement in consciousness, limbic, cognitive, and executive functions.


Subject(s)
Norepinephrine/physiology , Receptors, Adrenergic/physiology , Sympathetic Nervous System/physiology , Thalamus/physiology , Animals , Autoradiography , Axons/physiology , Dopamine beta-Hydroxylase/metabolism , Electrophysiological Phenomena , Female , Macaca mulatta , Macaca nemestrina , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Receptors, Adrenergic/drug effects , Receptors, Adrenergic, alpha-1/drug effects , Receptors, Adrenergic, alpha-1/genetics , Receptors, Adrenergic, alpha-1/metabolism , Sympathetic Nervous System/diagnostic imaging , Sympathetic Nervous System/drug effects
19.
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
20.
Proc Natl Acad Sci U S A ; 116(44): 22282-22287, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31619570

ABSTRACT

Sympathetic activation of ß-adrenoreceptors (ß-AR) represents a hallmark in the development of heart failure (HF). However, little is known about the underlying mechanisms of gene regulation. In human ventricular myocardium from patients with end-stage HF, we found high levels of phosphorylated histone 3 at serine-28 (H3S28p). H3S28p was increased by inhibition of the catecholamine-sensitive protein phosphatase 1 and decreased by ß-blocker pretreatment. By a series of in vitro and in vivo experiments, we show that the ß-AR downstream protein kinase CaM kinase II (CaMKII) directly binds and phosphorylates H3S28. Whereas, in CaMKII-deficient myocytes, acute catecholaminergic stimulation resulted in some degree of H3S28p, sustained catecholaminergic stimulation almost entirely failed to induce H3S28p. Genome-wide analysis of CaMKII-mediated H3S28p in response to chronic ß-AR stress by chromatin immunoprecipitation followed by massive genomic sequencing led to the identification of CaMKII-dependent H3S28p target genes. Forty percent of differentially H3S28p-enriched genomic regions were associated with differential, mostly increased expression of the nearest genes, pointing to CaMKII-dependent H3S28p as an activating histone mark. Remarkably, the adult hemoglobin genes showed an H3S28p enrichment close to their transcriptional start or end sites, which was associated with increased messenger RNA and protein expression. In summary, we demonstrate that chronic ß-AR activation leads to CaMKII-mediated H3S28p in cardiomyocytes. Thus, H3S28p-dependent changes may play an unexpected role for cardiac hemoglobin regulation in the context of sympathetic activation. These data also imply that CaMKII may be a yet unrecognized stress-responsive regulator of hematopoesis.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Heart Failure/metabolism , Hemoglobins/genetics , Histone Code , Histones/metabolism , Myocardium/metabolism , Sympathetic Nervous System/physiology , Adrenergic beta-Antagonists/pharmacology , Adult , Animals , Catecholamines/pharmacology , Cells, Cultured , Female , Heart Failure/genetics , Hemoglobins/metabolism , Humans , Male , Mice , Middle Aged , Phosphorylation , Rats , Sympathetic Nervous System/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL