Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 630
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 577(7792): 676-681, 2020 01.
Article in English | MEDLINE | ID: mdl-31969699

ABSTRACT

Empirical and anecdotal evidence has associated stress with accelerated hair greying (formation of unpigmented hairs)1,2, but so far there has been little scientific validation of this link. Here we report that, in mice, acute stress leads to hair greying through the fast depletion of melanocyte stem cells. Using a combination of adrenalectomy, denervation, chemogenetics3,4, cell ablation and knockout of the adrenergic receptor specifically in melanocyte stem cells, we find that the stress-induced loss of melanocyte stem cells is independent of immune attack or adrenal stress hormones. Instead, hair greying results from activation of the sympathetic nerves that innervate the melanocyte stem-cell niche. Under conditions of stress, the activation of these sympathetic nerves leads to burst release of the neurotransmitter noradrenaline (also known as norepinephrine). This causes quiescent melanocyte stem cells to proliferate rapidly, and is followed by their differentiation, migration and permanent depletion from the niche. Transient suppression of the proliferation of melanocyte stem cells prevents stress-induced hair greying. Our study demonstrates that neuronal activity that is induced by acute stress can drive a rapid and permanent loss of somatic stem cells, and illustrates an example in which the maintenance of somatic stem cells is directly influenced by the overall physiological state of the organism.


Subject(s)
Autonomic Pathways/physiopathology , Hair Color/physiology , Melanocytes/pathology , Stem Cell Niche/physiology , Stem Cells/pathology , Stress, Psychological/physiopathology , Sympathetic Nervous System/physiopathology , Adrenal Glands/metabolism , Adrenalectomy , Animals , Autonomic Pathways/pathology , Cell Proliferation , Cells, Cultured , Denervation , Female , Humans , Male , Melanocytes/cytology , Melanocytes/metabolism , Mice , Norepinephrine/metabolism , Psychological Trauma/pathology , Psychological Trauma/physiopathology , Receptors, Adrenergic, beta-2/deficiency , Receptors, Adrenergic, beta-2/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Stress, Psychological/pathology , Sympathetic Nervous System/pathology
2.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Article in English | MEDLINE | ID: mdl-35210363

ABSTRACT

Cancer-associated cachexia (CAC) is a hypermetabolic syndrome characterized by unintended weight loss due to the atrophy of adipose tissue and skeletal muscle. A phenotypic switch from white to beige adipocytes, a phenomenon called browning, accelerates CAC by increasing the dissipation of energy as heat. Addressing the mechanisms of white adipose tissue (WAT) browning in CAC, we now show that cachexigenic tumors activate type 2 immunity in cachectic WAT, generating a neuroprotective environment that increases peripheral sympathetic activity. Increased sympathetic activation, in turn, results in increased neuronal catecholamine synthesis and secretion, ß-adrenergic activation of adipocytes, and induction of WAT browning. Two genetic mouse models validated this progression of events. 1) Interleukin-4 receptor deficiency impeded the alternative activation of macrophages, reduced sympathetic activity, and restrained WAT browning, and 2) reduced catecholamine synthesis in peripheral dopamine ß-hydroxylase (DBH)-deficient mice prevented cancer-induced WAT browning and adipose atrophy. Targeting the intraadipose macrophage-sympathetic neuron cross-talk represents a promising therapeutic approach to ameliorate cachexia in cancer patients.


Subject(s)
Adipose Tissue, Brown/pathology , Cachexia/pathology , Cell Communication , Neoplasms/complications , Neurons/pathology , Sympathetic Nervous System/pathology , Animals , Cachexia/etiology , Cachexia/metabolism , Gene Expression , Heterografts , Humans , Mice , Neoplasms/metabolism , Receptors, Adrenergic, beta/metabolism , Thermogenesis
3.
J Hum Genet ; 69(9): 433-440, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38866925

ABSTRACT

BACKGROUND: Intronic GAA repeat expansion ([GAA] ≥250) in FGF14 is associated with the late-onset neurodegenerative disorder, spinocerebellar ataxia 27B (SCA27B, GAA-FGF14 ataxia). We aim to determine the prevalence of the GAA repeat expansion in FGF14 in Chinese populations presenting late-onset cerebellar ataxia (LOCA) and evaluate the characteristics of tandem repeat inheritance, radiological features and sympathetic nerve involvement. METHODS: GAA-FGF14 repeat expansion was screened in an undiagnosed LOCA cohort (n = 664) and variations in repeat-length were analyzed in families of confirmed GAA-FGF14 ataxia patients. Brain magnetic resonance imaging (MRI) was used to evaluate the radiological feature in GAA-FGF14 ataxia patients. Clinical examinations and sympathetic skin response (SSR) recordings in GAA-FGF14 patients (n = 16) were used to quantify sympathetic nerve involvement. RESULTS: Two unrelated probands (2/664) were identified. Genetic screening for GAA-FGF14 repeat expansion was performed in 39 family members, 16 of whom were genetically diagnosed with GAA-FGF14 ataxia. Familial screening revealed expansion of GAA repeats in maternal transmissions, but contraction upon paternal transmission. Brain MRI showed slight to moderate cerebellar atrophy. SSR amplitude was lower in GAA-FGF14 patients in pre-symptomatic stage compared to healthy controls, and further decreased in the symptomatic stage. CONCLUSIONS: GAA-FGF14 ataxia was rare among Chinese LOCA cases. Parental gender appears to affect variability in GAA repeat number between generations. Reduced SSR amplitude is a prominent feature in GAA-FGF14 patients, even in the pre-symptomatic stage.


Subject(s)
Fibroblast Growth Factors , Humans , Male , Female , Fibroblast Growth Factors/genetics , Middle Aged , Adult , Magnetic Resonance Imaging , Sympathetic Nervous System/physiopathology , Sympathetic Nervous System/pathology , Aged , Pedigree , Trinucleotide Repeat Expansion/genetics , Tandem Repeat Sequences/genetics , Spinocerebellar Degenerations
4.
J Mol Cell Cardiol ; 150: 12-22, 2021 01.
Article in English | MEDLINE | ID: mdl-33011158

ABSTRACT

Salt sensitivity of blood pressure (SSBP) is a trait carrying strong prognostic implications for various cardiovascular diseases. To test the hypothesis that excessive maternal salt intake causes SSBP in offspring through a mechanism dependent upon arginine-vasopressin (AVP), we performed a series of experiments using offspring of the rat dams salt-loaded during pregnancy and lactation with 1.5% saline drink ("experimental offspring") and those with normal perinatal salt exposure ("control offspring"). Salt challenge, given at 7-8 weeks of age with either 2% saline drink (3 days) or 8% NaCl-containing chow (4 weeks), had little or no effect on systolic blood pressure (SBP) in female offspring, whereas the salt challenge significantly raised SBP in male offspring, with the magnitude of increase being greater in experimental, than control, rats. Furthermore, the salt challenge not only raised plasma AVP level more and caused greater depressor responses to V1a and V2 AVP receptor antagonists to occur in experimental, than control, males, but it also made GABA excitatory in a significant proportion of magnocellular AVP neurons of experimental males by depolarizing GABA equilibrium potential. The effect of the maternal salt loading on the salt challenge-elicited SBP response in male offspring was precluded by maternal conivaptan treatment (non-selective AVP receptor antagonist) during the salt-loading period, whereas it was mimicked by neonatal AVP treatment. These results suggest that the excessive maternal salt intake brings about SSBP in male offspring, both the programming and the expression of which depend on increased AVP secretion that may partly result from excitatory GABAergic action.


Subject(s)
Blood Pressure , Prenatal Exposure Delayed Effects/pathology , Sodium Chloride, Dietary/adverse effects , Vasopressins/metabolism , Animals , Benzazepines/pharmacology , Benzazepines/therapeutic use , Female , Lactation/drug effects , Male , Neurons/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/blood , Prenatal Exposure Delayed Effects/cerebrospinal fluid , Rats, Sprague-Dawley , Receptors, GABA/metabolism , Sodium/blood , Sodium/cerebrospinal fluid , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/pathology , Systole/drug effects , Vasopressins/blood , gamma-Aminobutyric Acid/metabolism
5.
Horm Metab Res ; 53(2): 75-84, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33202429

ABSTRACT

SGLT2 inhibitors increase renal glucose excretion and thus decrease both fasting and postprandial plasma glucose levels. The effects of SGLT2 inhibition outweigh those on glycemic control and are also associated with the induction of hemodynamic changes that improve cardiovascular and renal function in people with type 2 diabetes. The exact mechanisms have not yet been completely clarified. This review is focused on the potential relationship between SGLT2 inhibition and sympathetic nerve activity. There is accumulating evidence for a suppressive effect of SGLT2 inhibitors on the sympathetic nerve tone, which might be a putative mechanism for cardiovascular protection in subjects with type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/pathology , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sympathetic Nervous System/pathology , Animals , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/pathology , Humans , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Sympathetic Nervous System/drug effects
6.
Mol Ther ; 28(7): 1731-1740, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32243833

ABSTRACT

VEGF-B gene therapy is a promising proangiogenic treatment for ischemic heart disease, but, unexpectedly, we found that high doses of VEGF-B promote ventricular arrhythmias (VAs). VEGF-B knockout, alpha myosin heavy-chain promoter (αMHC)-VEGF-B transgenic mice, and pigs transduced intramyocardially with adenoviral (Ad)VEGF- B186 were studied. Immunostaining showed a 2-fold increase in the number of nerves per field (76 vs. 39 in controls, p < 0.001) and an abnormal nerve distribution in the hypertrophic hearts of 11- to 20-month-old αMHC-VEGF-B mice. AdVEGF-B186 gene transfer (GT) led to local sprouting of nerve endings in pig myocardium (141 vs. 78 nerves per field in controls, p < 0.05). During dobutamine stress, 60% of the αMHC-VEGF-B hypertrophic mice had arrhythmias as compared to 7% in controls, and 20% of the AdVEGF-B186-transduced pigs and 100% of the combination of AdVEGF-B186- and AdsVEGFR-1-transduced pigs displayed VAs and even ventricular fibrillation. AdVEGF-B186 GT significantly increased the risk of sudden cardiac death in pigs when compared to any other GT with different VEGFs (hazard ratio, 500.5; 95% confidence interval [CI] 46.4-5,396.7; p < 0.0001). In gene expression analysis, VEGF-B induced the upregulation of Nr4a2, ATF6, and MANF in cardiomyocytes, molecules previously linked to nerve growth and differentiation. Thus, high AdVEGF-B186 overexpression induced nerve growth in the adult heart via a VEGFR-1 signaling-independent mechanism, leading to an increased risk of VA and sudden cardiac death.


Subject(s)
Arrhythmias, Cardiac/pathology , Myosin Heavy Chains/genetics , Sympathetic Nervous System/pathology , Up-Regulation , Vascular Endothelial Growth Factor B/genetics , Animals , Animals, Genetically Modified , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Dependovirus/genetics , Disease Notification , Female , Gene Knockout Techniques , Genetic Therapy , Genetic Vectors/administration & dosage , Male , Mice , Promoter Regions, Genetic , Recombinant Proteins/metabolism , Swine , Sympathetic Nervous System/metabolism , Transduction, Genetic , Vascular Endothelial Growth Factor B/adverse effects , Vascular Endothelial Growth Factor B/metabolism
7.
Lipids Health Dis ; 20(1): 5, 2021 Jan 12.
Article in English | MEDLINE | ID: mdl-33436015

ABSTRACT

BACKGROUND: Several large clinical trials have confirmed the cardioprotective role of sodium-glucose cotransporter 2 inhibitors (SGLT2i) in patients with type 2 diabetes. However, whether empagliflozin, as an SGLT2i, could alleviate atherosclerosis progression in non-diabetic states remain unknown. METHODS: ApoE-/- mice were fed a Western diet for 12 weeks to induce atherosclerosis. On the 7th week, a group of mice were treated with drinking water containing empagliflozin (10 mg/kg/day), while another group was given normal water. At the 12th week, the whole aortas of each group were harvested. Oil Red O, HE and Movat staining were performed for atherosclerotic lesion area and size. Mouse serum lipid profiles (total cholesterol [TC], triglyceride [TG], low-density lipoprotein-c [LDL], and high-density lipoprotein-c [HDL]), systemic inflammation levels (IL-1ß, IL-6 and IL-10), renin-angiotensin-aldosterone system (RAAS) components and sympathetic activity (norepinephrine and neuropeptide Y) indicators were measured by ELISA. RESULTS: Empagliflozin reduced the atherosclerotic lesion burden (-8.6 %, P = 0.004) at aortic root in ApoE-/- mice. In addition, empagliflozin decreased body weight (-3.27 g, P = 0.002), lipid profiles (TC: [-15.3 mmol/L, P = 0.011]; TG: [-2.4 mmol/L, P < 0.001]; LDL: [-2.9 mmol/L, P = 0.010]), RAAS (renin [-9.3 ng/L, P = 0.047]; aldosterone [-16.7 ng/L, P < 0.001]) and sympathetic activity (norepinephrine [-8.9 ng/L, P = 0.019]; neuropeptide Y [-8.8 ng/L, P = 0.002]). However, the anti-inflammatory effect of empagliflozin was not significantly evident. CONCLUSIONS: The early atherosclerotic lesion size was less visible in empagliflozin-treated mice. Empagliflozin could decrease lipid profiles and sympathetic activity in atherosclerosis.


Subject(s)
Atherosclerosis/drug therapy , Atherosclerosis/prevention & control , Benzhydryl Compounds/therapeutic use , Disease Progression , Glucosides/therapeutic use , Lipids/blood , Sympathetic Nervous System/pathology , Animals , Atherosclerosis/blood , Benzhydryl Compounds/pharmacology , Body Weight/drug effects , Glucosides/pharmacology , Inflammation/blood , Inflammation/pathology , Male , Mice , Neuropeptide Y/blood , Norepinephrine/blood , Renin-Angiotensin System/drug effects , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Sympathetic Nervous System/drug effects
8.
J Infect Dis ; 222(10): 1745-1755, 2020 10 13.
Article in English | MEDLINE | ID: mdl-32498080

ABSTRACT

Neurological signs and symptoms are the most common complications of Ebola virus disease. However, the mechanisms underlying the neurologic manifestations in Ebola patients are not known. In this study, peripheral ganglia were collected from 12 rhesus macaques that succumbed to Ebola virus (EBOV) disease from 5 to 8 days post exposure. Ganglionitis, characterized by neuronal degeneration, necrosis, and mononuclear leukocyte infiltrates, was observed in the dorsal root, autonomic, and enteric ganglia. By immunohistochemistry, RNAscope in situ hybridization, transmission electron microscopy, and confocal microscopy, we confirmed that CD68+ macrophages are the target cells for EBOV in affected ganglia. Further, we demonstrated that EBOV can induce satellite cell and neuronal apoptosis and microglial activation in infected ganglia. Our results demonstrate that EBOV can infect peripheral ganglia and results in ganglionopathy in rhesus macaques, which may contribute to the neurological signs and symptoms observed in acute and convalescent Ebola virus disease in human patients.


Subject(s)
Hemorrhagic Fever, Ebola/complications , Hemorrhagic Fever, Ebola/pathology , Nerve Degeneration/complications , Nerve Degeneration/pathology , Peripheral Nervous System Diseases/complications , Peripheral Nervous System Diseases/pathology , Animals , Antigens, CD , Antigens, Differentiation, Myelomonocytic , Disease Models, Animal , Ebolavirus , Female , Ganglia , Ganglia, Spinal/pathology , Ganglia, Spinal/virology , Ganglion Cysts/pathology , Hemorrhagic Fever, Ebola/virology , Humans , Immunohistochemistry , Leukocytes, Mononuclear , Macaca mulatta , Macrophages/pathology , Male , Microglia/pathology , Microglia/virology , Necrosis , Parasympathetic Nervous System/pathology , Peripheral Nervous System Diseases/virology , Sensory Receptor Cells/pathology , Sensory Receptor Cells/virology , Sympathetic Nervous System/pathology
9.
J Anat ; 236(1): 156-164, 2020 01.
Article in English | MEDLINE | ID: mdl-31498441

ABSTRACT

Omental milky spots (OMSs), small lymphoid structures positioned in the greater omentum, are involved in peritoneal immune homeostasis and the formation of omental metastases. Sympathetic nerve activity is known to regulate immune function in other lymphoid organs (e.g. spleen and lymph nodes) and to create a favourable microenvironment for various tumour types. However, it is still unknown whether OMSs receive sympathetic innervation. Therefore, the aim of this study was to establish whether OMSs of the adult human greater omentum receive sympathetic innervation. A total of 18 OMSs were isolated from five omenta, which were removed from 3% formaldehyde-perfused cadavers (with a median age of 84 years, ranging from 64 to 94). OMSs were embedded in paraffin, cut and stained with a general (PGP9.5) and sympathetic nerve marker (TH and DBH), and evaluated by bright field microscopy. A T-cell, B-cell, and macrophage staining was performed to confirm OMS identity. In 50% of the studied OMSs, sympathetic nerve fibres were observed at multiple levels of the same OMS. Nerve fibres were represented as dots or elongated structures and often observed in relation to small vessels and occasionally as individual structures residing between lymphoid cells. The current study shows that 50% of the investigated OMSs contain sympathetic nerve fibres. These findings may contribute to our understanding of neural regulation of peritoneal immune response and the involvement of OMSs in omental metastases.


Subject(s)
Lymphoid Tissue/pathology , Nerve Fibers/pathology , Omentum/pathology , Sympathetic Nervous System/pathology , Aged , Aged, 80 and over , Humans , Middle Aged
10.
Nature ; 512(7512): 78-81, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25043017

ABSTRACT

Myeloproliferative neoplasms (MPNs) are diseases caused by mutations in the haematopoietic stem cell (HSC) compartment. Most MPN patients have a common acquired mutation of Janus kinase 2 (JAK2) gene in HSCs that renders this kinase constitutively active, leading to uncontrolled cell expansion. The bone marrow microenvironment might contribute to the clinical outcomes of this common event. We previously showed that bone marrow nestin(+) mesenchymal stem cells (MSCs) innervated by sympathetic nerve fibres regulate normal HSCs. Here we demonstrate that abrogation of this regulatory circuit is essential for MPN pathogenesis. Sympathetic nerve fibres, supporting Schwann cells and nestin(+) MSCs are consistently reduced in the bone marrow of MPN patients and mice expressing the human JAK2(V617F) mutation in HSCs. Unexpectedly, MSC reduction is not due to differentiation but is caused by bone marrow neural damage and Schwann cell death triggered by interleukin-1ß produced by mutant HSCs. In turn, in vivo depletion of nestin(+) cells or their production of CXCL12 expanded mutant HSC number and accelerated MPN progression. In contrast, administration of neuroprotective or sympathomimetic drugs prevented mutant HSC expansion. Treatment with ß3-adrenergic agonists that restored the sympathetic regulation of nestin(+) MSCs prevented the loss of these cells and blocked MPN progression by indirectly reducing the number of leukaemic stem cells. Our results demonstrate that mutant-HSC-driven niche damage critically contributes to disease manifestation in MPN and identify niche-forming MSCs and their neural regulation as promising therapeutic targets.


Subject(s)
Hematopoietic Stem Cells/pathology , Myeloproliferative Disorders/pathology , Neoplasms/pathology , Nerve Fibers/pathology , Stem Cell Niche , Sympathetic Nervous System/pathology , Adrenergic beta-3 Receptor Agonists/pharmacology , Adrenergic beta-3 Receptor Agonists/therapeutic use , Animals , Apoptosis/drug effects , Disease Progression , Female , Hematopoietic Stem Cells/drug effects , Humans , Interleukin-1beta/metabolism , Janus Kinase 2/genetics , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/pathology , Mice , Myeloproliferative Disorders/drug therapy , Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Nerve Fibers/drug effects , Nestin/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Receptors, Adrenergic, beta-3/metabolism , Schwann Cells/drug effects , Schwann Cells/pathology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiopathology
11.
Acta Pharmacol Sin ; 41(3): 311-318, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31530901

ABSTRACT

Acute sympathetic stress causes excessive secretion of catecholamines and induces cardiac injuries, which are mainly mediated by ß-adrenergic receptors (ß-ARs). However, α1-adrenergic receptors (α1-ARs) are also expressed in the heart and are activated upon acute sympathetic stress. In the present study, we investigated whether α1-AR activation induced cardiac inflammation and the underlying mechanisms. Male C57BL/6 mice were injected with a single dose of α1-AR agonist phenylephrine (PE, 5 or 10 mg/kg, s.c.) with or without pretreatment with α-AR antagonist prazosin (5 mg/kg, s.c.). PE injection caused cardiac dysfunction and cardiac inflammation, evidenced by the increased expression of inflammatory cytokine IL-6 and chemokines MCP-1 and MCP-5, as well as macrophage infiltration in myocardium. These effects were blocked by prazosin pretreatment. Furthermore, PE injection significantly increased the expression of NOD-like receptor protein 3 (NLRP3) and the cleavage of caspase-1 (p20) and interleukin-18 in the heart; similar results were observed in both Langendorff-perfused hearts and cultured cardiomyocytes following the treatment with PE (10 µM). Moreover, PE-induced NLRP3 inflammasome activation and cardiac inflammation was blocked in Nlrp3-/- mice compared with wild-type mice. In conclusion, α1-AR overactivation induces cardiac inflammation by activating NLRP3 inflammasomes.


Subject(s)
Inflammasomes/metabolism , Inflammation/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Adrenergic alpha-1 Receptor Agonists/pharmacology , Animals , Dose-Response Relationship, Drug , Echocardiography , Heart/drug effects , Inflammasomes/drug effects , Inflammation/chemically induced , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Structure , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Phenylephrine/pharmacology , Structure-Activity Relationship , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/pathology
12.
Heart Lung Circ ; 29(4): 529-537, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31959550

ABSTRACT

Sympathetically triggered inherited arrhythmia syndromes, including the long QT syndrome (LQTS) and catecholaminergic polymorphic ventricular tachycardia (CPVT), can cause sudden cardiac death in young individuals with structurally normal hearts. With cardiac events typically triggered by physical or emotional stress, not surprisingly, two of the most common treatments are neuromodulators, including mainstay beta blocker pharmacotherapy, and surgical sympathetic cardiac denervation. This review updates the clinician on the relevant anatomy and physiology of the cardiac autonomic nervous system, outlines neurocardiac arrhythmia mechanisms, and discusses the latest rationale for a neurocardiac therapeutic approach to manage sympathetic-induced arrhythmia in patients with inherited cardiac disease.


Subject(s)
Brain , Heart/physiopathology , Long QT Syndrome , Psychological Distress , Sympathetic Nervous System , Tachycardia, Ventricular , Adrenergic beta-Antagonists/therapeutic use , Brain/pathology , Brain/physiopathology , Humans , Long QT Syndrome/pathology , Long QT Syndrome/physiopathology , Long QT Syndrome/therapy , Sympathetic Nervous System/pathology , Sympathetic Nervous System/physiopathology , Tachycardia, Ventricular/pathology , Tachycardia, Ventricular/physiopathology , Tachycardia, Ventricular/therapy
13.
J Cell Mol Med ; 23(2): 1001-1013, 2019 02.
Article in English | MEDLINE | ID: mdl-30411499

ABSTRACT

Reducing sympathetic neurohormone expression is a key therapeutic option in attenuating cardiac remodelling. Present study tested the feasibility of attenuating cardiac remodelling through reducing sympathetic neurohormone level by partial cardiac sympathetic denervation in a rat model of chronic volume overload. Male Sprague-Dawley rats were randomized into sham group (S, n = 7), aortocaval fistula group (AV, n = 7), and aortocaval fistula with bilateral sympathetic stellate ganglionectomy group (AD, n = 8). After 12 weeks, myocardial protein expression of sympathetic neurohormones, including tyrosine hydroxylase, neuropeptide Y, growth associated protein 43, and protein gene product 9.5, were significantly up-regulated in AV group compared to S group, and down-regulated in AD group. Cardiac remodelling was aggravated in AV group compared to S group and attenuated in AD group. The myocardial deposition of extracellular matrix, including collagen I and III, was enhanced in AV group, which was reduced in AD group. Myocardial angiotensin II and aldosterone expressions were significantly up-regulated in AV group and down-regulated in AD group. Our results show that bilateral sympathetic stellate ganglionectomy could attenuate cardiac remodelling and fibrosis by down-regulating sympathetic neurohormones expression in this rat model of chronic volume overload.


Subject(s)
Fibrosis/pathology , Myocardium/pathology , Sympathetic Nervous System/pathology , Ventricular Remodeling/physiology , Animals , Collagen/metabolism , Down-Regulation/physiology , Extracellular Matrix/metabolism , Fibrosis/metabolism , Ganglionectomy/methods , Heart/physiopathology , Heart Failure/metabolism , Heart Failure/pathology , Male , Rats , Rats, Sprague-Dawley , Sympathectomy/methods , Sympathetic Nervous System/metabolism , Up-Regulation/physiology
14.
J Cell Biochem ; 120(3): 3998-4008, 2019 03.
Article in English | MEDLINE | ID: mdl-30259991

ABSTRACT

Molecular hydrogen is reported to be used medically to ameliorate various systemic pathological conditions. This study aimed to investigate the effect of hydrogen (H2 ) gas on hypertension induced by intermittent hypoxia in rats. The adult rats were exposed to chronic intermittent hypoxia (CIH) 8 hours/day for 5 weeks and/or H 2 gas 2 hours/day. We found that the systolic and diastolic blood pressure (BP) increased significantly in rats exposed to intermittent hypoxia, both of which were markedly attenuated after H treatment. Furthermore, intermittent hypoxia exposure elevated renal sympathetic nerve activity, consistent with plasma norepinephrine. Additionally, H 2 gas significantly improved CIH-induced abnormal vascular relaxation. Nevertheless, inhalation of H 2 gas alone did not cause such changes. Moreover, H 2 gas-treated rats exposed to CIH showed a significant reduction in 8-hydroxy-2 deoxyguanosine content and increases in superoxide dismutase activity, indicating improved oxidative stress. Taken together, these results indicate that H 2 gas has significant effects on the reduction of BP without any side effects. Mechanistically, inhibition of sympathetic activity and reduction of systemic vascular resistance may participate in this process via the antioxidant activity of H 2 .


Subject(s)
Antioxidants/pharmacology , Hypertension/drug therapy , Oxidative Stress/drug effects , Sympathetic Nervous System/drug effects , Animals , Blood Pressure/drug effects , Deoxyadenosines/metabolism , Gases/pharmacology , Humans , Hydrogen/pharmacology , Hypertension/etiology , Hypertension/metabolism , Hypoxia/complications , Hypoxia/metabolism , Hypoxia/pathology , Rats , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Sympathetic Nervous System/pathology , Vasodilation/drug effects
15.
Kidney Int ; 96(1): 129-138, 2019 07.
Article in English | MEDLINE | ID: mdl-30979565

ABSTRACT

The sympathetic nervous system is critical in maintaining the homeostasis of renal functions. However, its three-dimensional (3D) structures in the kidney have not been elucidated due to limitation of conventional imaging methods. CUBIC (Clear, Unobstructed Brain/Body Imaging Cocktails and Computational analysis) is a newly developed tissue-clearing technique, which enables whole-organ 3D imaging without thin-sectioning. Comprehensive 3D imaging by CUBIC found that sympathetic nerves are primarily distributed around arteries in the mouse kidney. Notably, the sympathetic innervation density was significantly decreased 10 days after ischemia-reperfusion injury (voluminal ratio of innervation area to kidney) by about 70%. Moreover, norepinephrine levels in kidney tissue (output of sympathetic nerves) were significantly reduced in injured kidneys by 77%, confirming sympathetic denervation after ischemia-reperfusion injury. Time-course imaging indicated that innervation partially recovered although overall denervation persisted 28 days after injury, indicating a continuous sympathetic nervous abnormality during the progression of chronic kidney disease. Thus, CUBIC-kidney, the 3D imaging analysis, can be a strong imaging tool, providing comprehensive, macroscopic perspectives for kidney research.


Subject(s)
Acute Kidney Injury/pathology , Kidney/innervation , Renal Insufficiency, Chronic/pathology , Reperfusion Injury/complications , Sympathetic Nervous System/pathology , Acute Kidney Injury/etiology , Animals , Disease Models, Animal , Disease Progression , Fluorescent Antibody Technique , Histocytological Preparation Techniques , Humans , Imaging, Three-Dimensional , Kidney/blood supply , Kidney/chemistry , Kidney/pathology , Male , Mice , Norepinephrine/analysis , Norepinephrine/metabolism , Sympathetic Nervous System/diagnostic imaging , Sympathetic Nervous System/metabolism
16.
Clin Exp Immunol ; 197(3): 319-328, 2019 09.
Article in English | MEDLINE | ID: mdl-31319436

ABSTRACT

Increased peripheral levels of cytokines and central microglial activation have been reported in patients with psychiatric disorders. The degree of both innate and adaptive immune activation is also associated with worse clinical outcomes and poor treatment response in these patients. Understanding the possible causes and mechanisms leading to this immune activation is therefore an important and necessary step for the development of novel and more effective treatment strategies for these patients. In this work, we review the evidence of literature pointing to childhood trauma as one of the main causes behind the increased immune activation in patients with psychiatric disorders. We then discuss the potential mechanisms linking the experience of early life adversity (ELA) to innate immune activation. Specifically, we focus on the innervation of the bone marrow from sympathetic nervous system (SNS) as a new and emerging mechanism that has the potential to bridge the observed increases in both central and peripheral inflammatory markers in patients exposed to ELA. Experimental studies in laboratory rodents suggest that SNS activation following early life stress exposure causes a shift in the profile of innate immune cells, with an increase in proinflammatory monocytes. In turn, these cells traffic to the brain and influence neural circuitry, which manifests as increased anxiety and other relevant behavioural phenotypes. To date, however, very few studies have been conducted to explore this candidate mechanism in humans. Future research is also needed to clarify whether these pathways could be partially reversible to improve prevention and treatment strategies in the future.


Subject(s)
Immunity, Innate , Mental Disorders , Stress, Psychological , Sympathetic Nervous System , Humans , Mental Disorders/etiology , Mental Disorders/immunology , Mental Disorders/pathology , Stress, Psychological/complications , Stress, Psychological/immunology , Stress, Psychological/pathology , Sympathetic Nervous System/immunology , Sympathetic Nervous System/pathology
17.
J Transl Med ; 17(1): 256, 2019 08 07.
Article in English | MEDLINE | ID: mdl-31391086

ABSTRACT

BACKGROUND: The adipose afferent reflex (AAR), a sympatho-excitatory reflex, can promote the elevation of sympathetic nerve activity (SNA) and blood pressure (BP). Inflammation in the paraventricular nucleus (PVN) involves sympathetic abnormality in some cardiovascular diseases such as hypertension. This study was designed to explore the effects of tumor necrosis factor alpha (TNFα) in the PVN on the AAR and SNA in rats with obesity-related hypertension (OH) induced by a high-fat diet for 12 weeks. METHODS: Renal sympathetic nerve activity (RSNA) and mean arterial pressure (MAP) were continuously recorded in anesthetized rats, and their responses to capsaicin (CAP) stimulation of the right inguinal white adipose tissue were used to evaluate the AAR. RESULTS: Compared to the control rats, the systolic blood pressure (SBP), plasma norepinephrine (NE, indicating SNA) and TNFα levels, TNFα mRNA and protein levels, reactive oxygen species (ROS) content and NADPH oxidase activity in the PVN were significantly elevated in rats with OH. TNFα in the PVN markedly enhanced sympathoexcitation and AAR. Moreover, the enhancement of AAR caused by TNFα can be significantly strengthened by the pretreatment of diethyldithiocarbamate (DETC), a superoxide dismutase inhibitor, but attenuated by TNF-α receptor antagonist R-7050, superoxide scavenger PEG-SOD and NADPH oxidase inhibitor apocynin (Apo) in rats with OH. Acute microinjection of TNF-α into the PVN significantly increased the activity of NADPH oxidase and ROS levels in rats with OH, which were effectively blocked by R-7050. Furthermore, our results also showed that the increased levels of ROS, TNFα and NADPH oxidase subunits mRNA and protein in the PVN of rats with OH were significantly reversed by pentoxifylline (PTX, 30 mg/kg daily ip; in 10% ethanol) application, a cytokine blocker, for a period of 5 weeks. PTX administration also significantly decreased SBP, AAR and plasma NE levels in rats with OH. CONCLUSIONS: TNFα in the PVN modulates AAR and contributes to sympathoexcitation in OH possibly through NADPH oxidase-dependent ROS generation. TNFα blockade attenuates AAR and sympathoexcitation that unveils TNFα in the PVN may be a possible therapeutic target for the intervention of OH.


Subject(s)
Adipose Tissue/metabolism , Diet, High-Fat , Obesity/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adipose Tissue, White/pathology , Adiposity , Animals , Body Weight , Inflammation/metabolism , Male , NADPH Oxidases/metabolism , Neurons, Afferent/metabolism , Norepinephrine/blood , Rats , Rats, Inbred SHR , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Sympathetic Nervous System/pathology , Systole , Tumor Necrosis Factor-alpha/blood
18.
Cell Mol Life Sci ; 75(6): 1057-1070, 2018 03.
Article in English | MEDLINE | ID: mdl-29058015

ABSTRACT

Neuroblastoma (NB) is a tumor of the sympathoadrenal system arising in children under 15 years of age. In Germany, NB accounts for 7% of childhood cancer cases, but 11% of cancer deaths. It originates from highly migratory progenitor cells that leave the dorsal neural tube and contribute neurons and glial cells to sympathetic ganglia, and chromaffin and supportive cells to the adrenal medulla and paraganglia. Clinically, histologically and molecularly, NBs present as extremely heterogeneous, ranging from very good to very poor prognosis. The etiology of NB still remains unclear and needs to be elucidated, however, aberrant auto- and paracrine embryonic cell communications seem to be likely candidates to initiate or facilitate the emergence, progression and regression of NB. The wingless-type MMTV integration site (WNT) family of proteins represents an evolutionary highly conserved signaling system that orchestrates embryogenesis. At least 19 ligands in the human, numerous receptors and co-receptors are known, which control not only proliferation, but also cell polarity, migration and differentiation. Here we seek to interconnect aspects of WNT signaling with sympathoadrenal and paraganglionic development to define new WNT signaling cues in the etiology and progression of NB.


Subject(s)
Adrenal Gland Diseases/genetics , Gene Expression Regulation, Neoplastic , Neuroblastoma/genetics , Paraganglioma/genetics , Wnt Proteins/genetics , Wnt Signaling Pathway/genetics , Adolescent , Adrenal Gland Diseases/metabolism , Adrenal Gland Diseases/mortality , Adrenal Gland Diseases/pathology , Adrenal Glands/growth & development , Adrenal Glands/metabolism , Adrenal Glands/pathology , Child , Child, Preschool , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Crest/growth & development , Neural Crest/metabolism , Neural Crest/pathology , Neuroblastoma/metabolism , Neuroblastoma/mortality , Neuroblastoma/pathology , Neurons/metabolism , Neurons/pathology , Paraganglioma/metabolism , Paraganglioma/mortality , Paraganglioma/pathology , Stem Cells/metabolism , Stem Cells/pathology , Survival Analysis , Sympathetic Nervous System/growth & development , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/pathology , Wnt Proteins/metabolism
19.
Proc Natl Acad Sci U S A ; 113(3): 746-50, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26733679

ABSTRACT

The distribution and function of sympathetic innervation in skeletal muscle have largely remained elusive. Here we demonstrate that sympathetic neurons make close contact with neuromuscular junctions and form a network in skeletal muscle that may functionally couple different targets including blood vessels, motor neurons, and muscle fibers. Direct stimulation of sympathetic neurons led to activation of muscle postsynaptic ß2-adrenoreceptor (ADRB2), cAMP production, and import of the transcriptional coactivator peroxisome proliferator-activated receptor γ-coactivator 1α (PPARGC1A) into myonuclei. Electrophysiological and morphological deficits of neuromuscular junctions upon sympathectomy and in myasthenic mice were rescued by sympathicomimetic treatment. In conclusion, this study identifies the neuromuscular junction as a target of the sympathetic nervous system and shows that sympathetic input is crucial for synapse maintenance and function.


Subject(s)
Health , Homeostasis , Nervous System Diseases/pathology , Neuromuscular Junction/pathology , Sympathetic Nervous System/pathology , Active Transport, Cell Nucleus , Animals , Biosensing Techniques , Cell Nucleus/metabolism , Cyclic AMP/metabolism , Female , Male , Mice, Inbred C57BL , Models, Biological , Muscle, Skeletal/innervation , Neuromuscular Junction/metabolism , Neurons/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phenotype , Signal Transduction , Sympathectomy , Sympathetic Nervous System/metabolism , Transcription Factors/metabolism
20.
Proc Natl Acad Sci U S A ; 113(43): 12298-12303, 2016 10 25.
Article in English | MEDLINE | ID: mdl-27791019

ABSTRACT

Melanocortin-4 receptor (Mc4r)-expressing neurons in the autonomic nervous system, particularly in the paraventricular nucleus of the hypothalamus (PVH), play an essential role in blood pressure (BP) control. Mc4r-deficient (Mc4rKO) mice are severely obese but lack obesity-related hypertension; they also show a reduced pressor response to salt loading. We have previously reported that lean juvenile offspring born to diet-induced obese rats (OffOb) exhibit sympathetic-mediated hypertension, and we proposed a role for postnatally raised leptin in its etiology. Here, we test the hypothesis that neonatal hyperleptinemia due to maternal obesity induces persistent changes in the central melanocortin system, thereby contributing to offspring hypertension. Working on the OffOb paradigm in both sexes and using transgenic technology to restore Mc4r in the PVH of Mc4rKO (Mc4rPVH) mice, we have now shown that these mice develop higher BP than Mc4rKO or WT mice. We have also found that experimental hyperleptinemia induced in the neonatal period in Mc4rPVH and WT mice, but not in the Mc4rKO mice, leads to heightened BP and severe renal dysfunction. Thus, Mc4r in the PVH appears to be required for early-life programming of hypertension arising from either maternal obesity or neonatal hyperleptinemia. Early-life exposure of the PVH to maternal obesity through postnatal elevation of leptin may have long-term consequences for cardiovascular health.


Subject(s)
Hypertension/genetics , Leptin/metabolism , Obesity/genetics , Prenatal Exposure Delayed Effects/genetics , Receptor, Melanocortin, Type 4/genetics , Animals , Blood Pressure/genetics , Diet/adverse effects , Disease Models, Animal , Female , Humans , Hypertension/complications , Hypertension/physiopathology , Leptin/genetics , Male , Maternal-Fetal Relations/physiology , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Obesity/complications , Obesity/physiopathology , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/pathology , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/pathology
SELECTION OF CITATIONS
SEARCH DETAIL