Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 483
Filter
1.
Clin Auton Res ; 34(1): 45-77, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38393672

ABSTRACT

PURPOSE: The heart receives cervical and thoracic sympathetic contributions. Although the stellate ganglion is considered the main contributor to cardiac sympathetic innervation, the superior cervical ganglia (SCG) is used in many experimental studies. The clinical relevance of the SCG to cardiac innervation is controversial. We investigated current morphological and functional evidence as well as controversies on the contribution of the SCG to cardiac innervation. METHODS: A systematic literature review was conducted in PubMed, Embase, Web of Science, and COCHRANE Library. Included studies received a full/text review and quality appraisal. RESULTS: Seventy-six eligible studies performed between 1976 and 2023 were identified. In all species studied, morphological evidence of direct or indirect SCG contribution to cardiac innervation was found, but its contribution was limited. Morphologically, SCG sidedness may be relevant. There is indirect functional evidence that the SCG contributes to cardiac innervation as shown by its involvement in sympathetic overdrive reactions in cardiac disease states. A direct functional contribution was not found. Functional data on SCG sidedness was largely unavailable. Information about sex differences and pre- and postnatal differences was lacking. CONCLUSION: Current literature mainly supports an indirect involvement of the SCG in cardiac innervation, via other structures and plexuses or via sympathetic overdrive in response to cardiac diseases. Morphological evidence of a direct involvement was found, but its contribution seems limited. The relevance of SCG sidedness, sex, and developmental stage in health and disease remains unclear and warrants further exploration.


Subject(s)
Heart , Superior Cervical Ganglion , Humans , Superior Cervical Ganglion/physiology , Heart/innervation , Heart/physiology , Animals , Autonomic Nervous System/physiology , Heart Diseases/physiopathology
2.
J Pharmacol Sci ; 148(1): 103-107, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34924113

ABSTRACT

We have reported that nicotine has a neurotrophic action on peripheral adrenergic nerves in vivo, which is mediated by α7 nicotinic acetylcholine receptors (nAChRs). To clarify the possible mechanisms, the present study further investigated the effect of nicotine on neurite outgrowth in tyrosine hydroxylase (TH)-positive superior cervical ganglia (SCG) cells isolated from neonatal rats in vitro. Nicotine at low concentrations (0.01-0.3 mM) increased the number of neurite outgrowths in TH-immunopositive SCG cells, while high concentrations of nicotine (1-10 mM) gradually reduced it, and only 10 mM nicotine was markedly inhibited compared to the control. A 100 µM of nicotine-induced increase in neurite numbers depended on the exposure time and was inhibited by treatment with the nAChR antagonist hexamethonium (Hex) and α7 nAChR antagonist α-bungarotoxin (α-Bgtx). The nicotine (10 mM)-induced a significant decrease in neurite outgrowth in SCG, which was perfectly canceled by Hex to the control level but not by α-Bgtx. These results suggest that nicotine has a regulatory neurotrophic action mediated by both α7 nAChR and other subtypes in TH-positive SCG cells of rats.


Subject(s)
Nerve Growth Factors , Neurites/drug effects , Neurites/physiology , Neuronal Outgrowth/drug effects , Nicotine/pharmacology , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/physiology , Animals , Cells, Cultured , Rats , alpha7 Nicotinic Acetylcholine Receptor/physiology
3.
BMB Rep ; 54(6): 311-316, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33408002

ABSTRACT

Ethanol often causes critical health problems by altering the neuronal activities of the central and peripheral nerve systems. One of the cellular targets of ethanol is the plasma membrane proteins including ion channels and receptors. Recently, we reported that ethanol elevates membrane excitability in sympathetic neurons by inhibiting Kv7.2/7.3 channels in a cell type-specific manner. Even though our studies revealed that the inhibitory effects of ethanol on the Kv7.2/7.3 channel was diminished by the increase of plasma membrane phosphatidylinositol 4,5-bisphosphate (PI (4,5)P2), the molecular mechanism of ethanol on Kv7.2/7.3 channel inhibition remains unclear. By investigating the kinetics of Kv7.2/7.3 current in high K+ solution, we found that ethanol inhibited Kv7.2/7.3 channels through a mechanism distinct from that of tetraethylammonium (TEA) which enters into the pore and blocks the gate of the channels. Using a non-stationary noise analysis (NSNA), we demonstrated that the inhibitory effect of ethanol is the result of reduction of open probability (PO) of the Kv7.2/7.3 channel, but not of a single channel current (i) or channel number (N). Finally, ethanol selectively facilitated the kinetics of Kv7.2 current suppression by voltage-sensing phosphatase (VSP)-induced PI(4,5)P2 depletion, while it slowed down Kv7.2 current recovery from the VSP-induced inhibition. Together our results suggest that ethanol regulates neuronal activity through the reduction of open probability and PI(4,5)P2 sensitivity of Kv7.2/7.3 channels. [BMB Reports 2021; 54(6): 311-316].


Subject(s)
Ethanol/pharmacology , Ion Channel Gating , KCNQ2 Potassium Channel/metabolism , KCNQ3 Potassium Channel/metabolism , Kidney/physiology , Neurons/physiology , Phosphatidylinositol 4,5-Diphosphate/metabolism , Animals , Central Nervous System Depressants/pharmacology , Humans , Kidney/drug effects , Mice , Neurons/drug effects , Superior Cervical Ganglion/drug effects , Superior Cervical Ganglion/physiology
4.
Histol Histopathol ; 35(12): 1493-1502, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33179759

ABSTRACT

Orthodontic tooth movement (OTM) is a specific treatment of malocclusion, whose regulation mechanism is still not clear. This study aimed to reveal the relationship between the sympathetic nervous system (SNS) and OTM through the construction of an OTM rat model through the utilization of orthodontic nickeltitanium coiled springs. The results indicated that the stimulation of SNS by dopamine significantly promote the OTM process represented by the much larger distance between the first and second molar compared with mere exertion of orthodontic force. Superior cervical ganglionectomy (SCGx) can alleviate this promotion effect, further proving the role of SNS in the process of OTM. Subsequently, the ability of orthodontic force to stimulate the center of the SNS was visualized by the tyrosin hydroxylase (TH) staining of neurons in ventromedial hypothalamic nucleus (VMH) and arcuate nucleus (ARC) of the hypothalamus, as well as the up-regulated expression of norepinephrine in local alveolar bone. Moreover, we also elucidated that the stimulation of SNS can promote osteoclast differentiation in periodontal ligament cells (PDLCs) and bone marrow-derived cells (BMCs) through regulation of receptor activator of nuclear factor-κB ligand (RANKL)/osteoprotegerin (OPG) system, thus promoting the OTM process. In conclusion, this study provided the first evidence for the involvement of the hypothalamus in the promotion effect of SNS on OTM. This work could provide a novel theoretical and experimental basis for further understanding of the molecular mechanism of OTM.


Subject(s)
Alveolar Process/physiology , Periodontal Ligament/physiology , Superior Cervical Ganglion/physiology , Tooth Migration , Tooth Mobility , Tooth Movement Techniques , Ventromedial Hypothalamic Nucleus/physiology , Alveolar Process/innervation , Alveolar Process/metabolism , Animals , Cells, Cultured , Dopamine/pharmacology , Ganglionectomy , Male , Mechanotransduction, Cellular , Norepinephrine/metabolism , Osteoclasts/physiology , Osteogenesis , Osteoprotegerin/metabolism , Periodontal Ligament/innervation , Periodontal Ligament/metabolism , RANK Ligand/metabolism , Rats, Sprague-Dawley , Superior Cervical Ganglion/surgery , Ventromedial Hypothalamic Nucleus/drug effects
5.
Int J Mol Sci ; 21(16)2020 Aug 12.
Article in English | MEDLINE | ID: mdl-32806753

ABSTRACT

The ionic mechanisms controlling the resting membrane potential (RMP) in superior cervical ganglion (SCG) neurons have been widely studied and the M-current (IM, KCNQ) is one of the key players. Recently, with the discovery of the presence of functional TREK-2 (TWIK-related K+ channel 2) channels in SCG neurons, another potential main contributor for setting the value of the resting membrane potential has appeared. In the present work, we quantified the contribution of TREK-2 channels to the resting membrane potential at physiological temperature and studied its role in excitability using patch-clamp techniques. In the process we have discovered that TREK-2 channels are sensitive to the classic M-current blockers linopirdine and XE991 (IC50 = 0.310 ± 0.06 µM and 0.044 ± 0.013 µM, respectively). An increase from room temperature (23 °C) to physiological temperature (37 °C) enhanced both IM and TREK-2 currents. Likewise, inhibition of IM by tetraethylammonium (TEA) and TREK-2 current by XE991 depolarized the RMP at room and physiological temperatures. Temperature rise also enhanced adaptation in SCG neurons which was reduced due to TREK-2 and IM inhibition by XE991 application. In summary, TREK-2 and M currents contribute to the resting membrane potential and excitability at room and physiological temperature in the primary culture of mouse SCG neurons.


Subject(s)
KCNQ Potassium Channels/metabolism , Membrane Potentials , Neurons/physiology , Potassium Channels, Tandem Pore Domain/metabolism , Sympathetic Nervous System/physiology , Temperature , Adaptation, Physiological/drug effects , Animals , Anthracenes/pharmacology , HEK293 Cells , Humans , Indoles/pharmacology , Ion Channel Gating/drug effects , Membrane Potentials/drug effects , Mice , Neurons/drug effects , Pyridines/pharmacology , Riluzole/pharmacology , Superior Cervical Ganglion/drug effects , Superior Cervical Ganglion/physiology , Tetraethylammonium/pharmacology , Tetrahydronaphthalenes/pharmacology , Tetrazoles/pharmacology
6.
Invest Ophthalmol Vis Sci ; 60(13): 4303-4309, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31618767

ABSTRACT

Purpose: To investigate specific effects of denervation and stimulation of the internal carotid nerve (ICN) on the choroid and retina. Methods: Female Sprague Dawley rats underwent unilateral ICN transection (n = 20) or acute ICN electrical stimulation (n = 7). Rats in the denervation group were euthanized 6 weeks after nerve transection, and eyes were analyzed for changes in choroidal vascularity (via histomorphometry) or angiogenic growth factors and inflammatory markers (via ELISA). Rats in the stimulation group received acute ICN electrical stimulation with a bipolar cuff electrode over a range of stimulus amplitudes, frequencies, and pulse widths. Choroidal blood flow and pupil diameter were monitored before, during, and after stimulation. Results: Six weeks after unilateral ICN transection, sympathectomized choroids exhibited increased vascularity, defined as the percentage of choroidal surface area occupied by blood vessel lumina. Vascular endothelial growth factor (VEGF) and VEGF receptor-2 (VEGFR-2) protein levels in denervated choroids were 61% and 124% higher than in contralateral choroids, respectively. TNF-α levels in denervated retinas increased by 3.3-fold relative to levels in contralateral retinas. In animals undergoing acute ICN electrical stimulation, mydriasis and reduced choroidal blood flow were observed in the ipsilateral eye. The magnitude of the reduction in blood flow correlated positively with stimulus frequency. Conclusions: Modulation of ICN activity reveals a potential role of the ocular sympathetic system in regulating endpoints related to neovascular diseases of the eye.


Subject(s)
Carotid Artery, Internal/innervation , Choroid/blood supply , Sympathectomy , Sympathetic Nervous System/surgery , Animals , Biomarkers/metabolism , Choroid/metabolism , Electric Stimulation , Enzyme-Linked Immunosorbent Assay , Female , Pupil/physiology , Rats , Rats, Sprague-Dawley , Retina/metabolism , Superior Cervical Ganglion/physiology , Sympathetic Nervous System/physiology , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
7.
Anat Rec (Hoboken) ; 301(11): 1906-1916, 2018 11.
Article in English | MEDLINE | ID: mdl-30338669

ABSTRACT

Wheat germ agglutinin-horseradish peroxidase was injected into the entire (0.8 µL) or partial (rostral or caudal, 0.1-0.3 µL) superior cervical ganglion (SCG) of the rat (male Sprague-Dawley, N = 35) to examine the distribution of neurons in the middle (MCG) and inferior (ICG) cervical ganglion that send axons bypass the SCG. Whole-mounts of the SCG, cervical sympathetic trunk (CST), MCG, ICG, and sections of the brainstem and spinal cord were prepared. With entire SCG tracer injection, neurons were labeled evenly in the MCG (left: 258, right: 121), ICG (left: 848, right: 681), and CST (up to 770). Some neurons grouped in a single bulge just rostral to the MCG, which we termed as the "premiddle cervical ganglion" (pMCG). The left pMCG (120) is larger and has more neurons than the right pMCG (82). Centrally, neurons were labeled in lamina IX of cervical segments (C1: 18%, C2: 46%, C3: 33%, C4: 3%), intermediate zone of thoracic segments (T1: 31%, T2: 35%, T3: 27%, T4: 7%), and intermediate reticular nuclei (96%) and perifacial zone (4%) of brainstem. The rostral and caudal SCG injection selectively labeled neurons mainly in brainstem, C1-C2 and in T1-T2, respectively. Before projecting to their peripheral targets, many neurons in pMCG, MCG and ICG run rostrally within the CST rather than segmentally through the closest rami, from the level of SCG or above. Neurons in pMCG and MCG may have similar or complementary function and those in brainstem may be involved in the vestibulo-autonomic interaction. Anat Rec, 301:1906-1916, 2018. © 2018 Wiley Periodicals, Inc.


Subject(s)
Axons/physiology , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/physiology , Animals , Axons/chemistry , Brain Stem/chemistry , Brain Stem/cytology , Brain Stem/physiology , Male , Rats , Rats, Sprague-Dawley , Spinal Cord/chemistry , Spinal Cord/cytology , Spinal Cord/physiology , Superior Cervical Ganglion/chemistry
8.
Biomed Res ; 38(2): 99-109, 2017.
Article in English | MEDLINE | ID: mdl-28442666

ABSTRACT

Pituitary adenylyl cyclase-activating polypeptide (PACAP) is a bioactive peptide with diverse effects in the nervous system. The present study investigated whether stimulation of PACAP receptors (PACAPRs) induces responses in neurons and satellite cells of the superior cervical ganglia (SCG), with special reference to intracellular Ca2+ ([Ca2+]i) changes. The expression of PACAPRs in SCG was detected by reverse transcription-PCR. PACAP type 1 receptor (PAC1R), vasoactive intestinal peptide receptor type (VPAC)1R, and VPAC2R transcripts were expressed in SCG, with PAC1R showing the highest levels. Confocal microscopy analysis revealed that PACAP38 and PACAP27 induced an increase in [Ca2+]i in SCG, first in satellite cells and subsequently in neurons. Neither extracellular Ca2+ removal nor Ca2+ channel blockade affected the PACAP38-induced increase in [Ca2+]i in satellite cells; however, this was partly inhibited in neurons. U73122 or xestospongin C treatment completely and partly abrogated [Ca2+]i changes in satellite cells and in neurons, respectively, whereas VPAC1R and VPAC2R agonists increased [Ca2+]i in satellite cells only. This is the first report demonstrating the expression of PACAPRs specifically, VPAC1 and VPAC2 in SCG and providing evidence for PACAP38-induced [Ca2+]i changes in both satellite cells and neurons via Ca2+ mobilization.


Subject(s)
Calcium Signaling , Calcium/metabolism , Neurons/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Satellite Cells, Perineuronal/physiology , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/physiology , Animals , Biomarkers , Calcium Signaling/drug effects , Gene Expression , Microscopy, Confocal , Molecular Imaging , Neurons/drug effects , Neurons/ultrastructure , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/agonists , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Satellite Cells, Perineuronal/drug effects , Satellite Cells, Perineuronal/ultrastructure
9.
Traffic ; 17(11): 1155-1167, 2016 11.
Article in English | MEDLINE | ID: mdl-27484852

ABSTRACT

Neurons rely heavily on axonal transport to deliver materials from the sites of synthesis to the axon terminals over distances that can be many centimetres long. KIF1A is the neuron-specific kinesin with the fastest reported anterograde motor activity. Previous studies have shown that KIF1A transports a subset of synaptic proteins, neurofilaments and dense-core vesicles. Using two-colour live imaging, we showed that beta-secretase 1 (BACE1)-mCherry moves together with KIF1A-GFP in both the anterograde and retrograde directions in superior cervical ganglions (SCG) neurons. We confirmed that KIF1A is functionally required for BACE1 transport by using KIF1A siRNA and a KIF1A mutant construct (KIF1A-T312M) to impair its motor activity. We further identified several cargoes that have little or no co-migration with KIF1A-GFP and also move independently from BACE1-mCherry. Together, these findings support a primary role for KIF1A in the anterograde transport of BACE1 and suggest that axonally transported cargoes are sorted into different classes of carrier vesicles in the cell body and are transported by cargo-specific motor proteins through the axon.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Axonal Transport/physiology , Kinesins/physiology , Motor Neurons/physiology , Protein Transport/physiology , Superior Cervical Ganglion/physiology , Amyloid Precursor Protein Secretases/genetics , Animals , Aspartic Acid Endopeptidases/genetics , Cells, Cultured , Green Fluorescent Proteins/genetics , Kinesins/genetics , Luminescent Proteins/genetics , Mice, Inbred C57BL , Microscopy, Fluorescence , Motor Neurons/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Signal Transduction , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/metabolism , Red Fluorescent Protein
10.
Toxicol Appl Pharmacol ; 305: 75-82, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27260673

ABSTRACT

The abuse of ketamine and amphetamine analogs is associated with incidence of hypertension and strokes involving activation of sympathetic activities. Large cerebral arteries at the base of the brain from several species receive dense sympathetic innervation which upon activation causes parasympathetic-nitrergic vasodilation with increased regional blood flow via axo-axonal interaction mechanism, serving as a protective mechanism to meet O2 demand in an acutely stressful situation. The present study was designed to examine effects of ketamine and amphetamine analogs on axo-axonal interaction-mediated neurogenic nitrergic vasodilation in porcine basilar arteries using techniques of blood-vessel myography, patch clamp and two-electrode voltage clamp, and calcium imaging. In U46619-contracted basilar arterial rings, nicotine (100µM) and electrical depolarization of nitrergic nerves by transmural nerve stimulation (TNS, 8Hz) elicited neurogenic nitrergic vasodilations. Ketamine and amphetamine analogs concentration-dependently inhibited nicotine-induced parasympathetic-nitrergic vasodilation without affecting that induced by TNS, nitroprusside or isoproterenol. Ketamine and amphetamine analogs also concentration-dependently blocked nicotine-induced inward currents in Xenopus oocytes expressing α3ß2-nicotinic acetylcholine receptors (nAChRs), and nicotine-induced inward currents as well as calcium influxes in rat superior cervical ganglion neurons. The potency in inhibiting both inward-currents and calcium influxes is ketamine>methamphetamine>hydroxyamphetamine. These results indicate that ketamine and amphetamine analogs, by blocking nAChRs located on cerebral perivascular sympathetic nerves, reduce nicotine-induced, axo-axonal interaction mechanism-mediated neurogenic dilation of the basilar arteries. Chronic abuse of these drugs, therefore, may interfere with normal sympathetic-parasympathetic interaction mechanism resulting in diminished neurogenic vasodilation and, possibly, normal blood flow in the brainstem.


Subject(s)
Amphetamines/pharmacology , Basilar Artery/drug effects , Ketamine/pharmacology , Receptors, Nicotinic/physiology , Vasoconstrictor Agents/pharmacology , Animals , Basilar Artery/metabolism , Basilar Artery/physiology , Calcium/metabolism , Circle of Willis/drug effects , Circle of Willis/physiology , In Vitro Techniques , Ketamine/analogs & derivatives , Nicotine/pharmacology , Oocytes , Rats, Sprague-Dawley , Receptors, Nicotinic/genetics , Superior Cervical Ganglion/drug effects , Superior Cervical Ganglion/metabolism , Superior Cervical Ganglion/physiology , Swine , Vasodilation/drug effects , Xenopus
11.
Development ; 143(9): 1560-70, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27143756

ABSTRACT

During development of the peripheral nervous system, excess neurons are generated, most of which will be lost by programmed cell death due to a limited supply of neurotrophic factors from their targets. Other environmental factors, such as 'competition factors' produced by neurons themselves, and axon guidance molecules have also been implicated in developmental cell death. Semaphorin 3A (Sema3A), in addition to its function as a chemorepulsive guidance cue, can also induce death of sensory neurons in vitro The extent to which Sema3A regulates developmental cell death in vivo, however, is debated. We show that in compartmentalized cultures of rat sympathetic neurons, a Sema3A-initiated apoptosis signal is retrogradely transported from axon terminals to cell bodies to induce cell death. Sema3A-mediated apoptosis utilizes the extrinsic pathway and requires both neuropilin 1 and plexin A3. Sema3A is not retrogradely transported in older, survival factor-independent sympathetic neurons, and is much less effective at inducing apoptosis in these neurons. Importantly, deletion of either neuropilin 1 or plexin A3 significantly reduces developmental cell death in the superior cervical ganglia. Taken together, a Sema3A-initiated apoptotic signaling complex regulates the apoptosis of sympathetic neurons during the period of naturally occurring cell death.


Subject(s)
Apoptosis/physiology , Nerve Tissue Proteins/metabolism , Neuropilin-1/metabolism , Receptors, Cell Surface/metabolism , Semaphorin-3A/metabolism , Superior Cervical Ganglion/embryology , Sympathetic Nervous System/embryology , Animals , Axons/metabolism , Caspase 3/metabolism , Cells, Cultured , Mice , Mice, Knockout , Microtubules/metabolism , Nerve Tissue Proteins/genetics , Neuropilin-1/genetics , RNA Interference , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/genetics , Signal Transduction , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/physiology
12.
Mol Pharmacol ; 89(4): 476-83, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26869400

ABSTRACT

Tetrodotoxin-sensitive Na(+) currents have been extensively studied because they play a major role in neuronal firing and bursting. In this study, we showed that voltage-dependent Na(+) currents are regulated in a slow manner by oxotremorine (oxo-M) and angiotensin II in rat sympathetic neurons. We found that these currents can be readily inhibited through a signaling pathway mediated by G proteins and phospholipase C (PLC) ß1. This inhibition is slowly established, pertussis toxin-insensitive, partially reversed within tens of seconds after oxo-M washout, and not relieved by a strong depolarization, suggesting a voltage-insensitive mechanism of inhibition. Specificity of the M1 receptor was tested by the MT-7 toxin. Activation and inactivation curves showed no shift in the voltage dependency under the inhibition by oxo-M. This inhibition is blocked by a PLC inhibitor (U73122, 1-(6-{[(17ß)-3-Methoxyestra-1,3,5(10)-trien-17-yl]amino}hexyl)-1H-pyrrole-2,5-dione), and recovery from inhibition is prevented by wortmannin, a PI3/4 kinase inhibitor. Hence, the pathway involves Gq/11 and is mediated by a diffusible second messenger. Oxo-M inhibition is occluded by screening phosphatidylinositol 4,5-bisphosphate (PIP2)-negative charges with poly-l-lysine and prevented by intracellular dialysis with a PIP2 analog. In addition, bisindolylmaleimide I, a specific ATP-competitive protein kinase C (PKC) inhibitor, rules out that this inhibition may be mediated by this protein kinase. Furthermore, oxo-M-induced suppression of Na(+) currents remains unchanged when neurons are treated with calphostin C, a PKC inhibitor that targets the diacylglycerol-binding site of the kinase. These results support a general mechanism of Na(+) current inhibition that is widely present in excitable cells through modulation of ion channels by specific G protein-coupled receptors.


Subject(s)
Angiotensin II/pharmacology , Oxotremorine/pharmacology , Sodium Channel Blockers/pharmacology , Sodium Channels/physiology , Superior Cervical Ganglion/physiology , Tetrodotoxin/pharmacology , Animals , Ganglia, Sympathetic/drug effects , Ganglia, Sympathetic/physiology , Male , Neurons/drug effects , Neurons/physiology , Rats , Rats, Wistar , Superior Cervical Ganglion/drug effects
13.
Philos Trans R Soc Lond B Biol Sci ; 370(1672)2015 Jul 05.
Article in English | MEDLINE | ID: mdl-26009767

ABSTRACT

Little is known about the interactions between nicotinic and muscarinic acetylcholine receptors (nAChRs and mAChRs). Here we report that methacholine (MCh), a selective agonist of mAChRs, inhibited up to 80% of nicotine-induced nAChR currents in sympathetic superior cervical ganglion neurons and adrenal chromaffin cells. The muscarine-induced inhibition (MiI) substantially reduced ACh-induced membrane currents through nAChRs and quantal neurotransmitter release. The MiI was time- and temperature-dependent. The slow recovery of nAChR current after washout of MCh, as well as the high value of Q10 (3.2), suggested, instead of a direct open-channel blockade, an intracellular metabotropic process. The effects of GTP-γ-S, GDP-ß-S and pertussis toxin suggested that MiI was mediated by G-protein signalling. Inhibitors of protein kinase C (bisindolymaleimide-Bis), protein kinase A (H89) and PIP2 depletion attenuated the MiI, indicating that a second messenger pathway is involved in this process. Taken together, these data suggest that mAChRs negatively modulated nAChRs via a G-protein-mediated second messenger pathway. The time dependence suggests that MiI may provide a novel mechanism for post-synaptic adaptation in all cells/neurons and synapses expressing both types of AChRs.


Subject(s)
Chromaffin Cells/physiology , Methacholine Chloride/pharmacology , Neurons/physiology , Nicotinic Antagonists/pharmacology , Superior Cervical Ganglion/cytology , Synaptic Transmission/physiology , Animals , Chromaffin Cells/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , GTP-Binding Proteins/metabolism , Muscarinic Agonists/pharmacology , Neurons/metabolism , Patch-Clamp Techniques , Protein Kinase C/antagonists & inhibitors , Rats , Second Messenger Systems/physiology , Superior Cervical Ganglion/physiology , Temperature , Time Factors
14.
Neurosci Lett ; 595: 45-9, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25849529

ABSTRACT

1,8-Cineole is a terpenoid present in many essential oil of plants with several pharmacological and biological effects, including antinociceptive, smooth muscle relaxant and ion channel activation. Also, 1,8-cineole blocked action potentials, reducing excitability of peripheral neurons. The objective of this work was to investigate effects of 1,8-cineole on Na(+) currents (INa(+)) in dissociated superior cervical ganglion neurons (SCG). Wistar rats of both sexes were used (10-12 weeks old, 200-300g). SCG's were dissected and neurons were enzymatically treated. To study 1,8-cineole effect on INa(+), the patch-clamp technique in whole-cell mode was employed. 1,8-Cineole (6.0mM) partially blocked INa(+) in SCG neurons. The effect stabilized within ∼150s and there was a partial recovery of INa(+) after washout. Current density was reduced from -105.8 to -83.7pA/pF, corresponding to a decrease to ∼20% of control. 1,8-Cineole also reduced the time-to-peak of INa(+) activation and the amplitude and decay time constants of INa(+) inactivation. Current-voltage plots revealed that 1,8-cineole left-shifted the V1/2 of both activation and inactivation curves by ∼10 and ∼20mV, respectively. In conclusion, we demonstrate that 1,8-cineole directly affects Na(+) channels of the SCG by modifying several gating parameters that are likely to be the major cause of excitability blockade.


Subject(s)
Cyclohexanols/pharmacology , Monoterpenes/pharmacology , Neurons/drug effects , Sodium Channel Blockers/pharmacology , Sodium Channels/physiology , Superior Cervical Ganglion/drug effects , Animals , Eucalyptol , Female , Ion Channel Gating/drug effects , Male , Neurons/physiology , Rats, Wistar , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/physiology
15.
J Physiol ; 593(4): 755-6, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25708916
16.
Neuroscience ; 289: 9-18, 2015 Mar 19.
Article in English | MEDLINE | ID: mdl-25583636

ABSTRACT

Both central and peripheral sympathetic nervous systems contribute to the cardiovascular effects of dexmedetomidine (DMED), a highly selective and widely used a2-adrenoceptor agonist for sedation, analgesia, and stress management. The central sympatholytic effects are augmented by peripheral inhibition of sympathetic ganglion transmission. The mechanism is not clear. In this research, using conventional patch-clamp recordings we investigated the direct effects of DMED on sodium (Na(+)) channel currents (INa) and nicotinic acetylcholine (ACh) receptor (nAChRs) channel currents (IACh) in rat superior cervical ganglion (SCG) neurons to explore the possible mechanisms of sympathetic ganglion transmission inhibition by DMED. DMED voltage-dependently suppressed INa with half maximal inhibitory concentration (IC50) values of 67.2±9.6µM and 26.1±5.3µM at holding potentials of -80mV and -60mV, respectively. The inhibition of Na(+) channels by DMED was also frequency dependent. 100µM DMED shifted the Na(+) channel inactivation curves to the hyperpolarizing direction by 9.8mV (P<0.01) and slowed the recovery from inactivation by 8.9ms (P<0.01), but no effects were seen on the shape of the current-voltage relationship or Na(+) channels activation curves. DMED dose-dependently inhibited IACh with an IC50 value of 5.5±2.4µM in SCG neurons, and this inhibition was voltage-independent. DMED pretreatment followed by fast co-application of DMED and ACh produced a significantly larger IACh inhibition than without DMED pretreatment. Yohimbine, phentolamine, and atropine pretreatment did not alter the inhibitory effects of DMED on INa and IACh. In conclusion, DMED dose-dependently inhibits INa and IACh in rat SCG neurons by preferential binding to the inactivated state of the Na(+) channels and the closed state (resting) of nAChR channels respectively. Both inhibitions are a2-adrenoceptor independent. Furthermore, the nAChR channels in rat SCG neurons are much more sensitive to inhibition by DMED than Na(+) channels.


Subject(s)
Dexmedetomidine/pharmacology , Membrane Transport Modulators/pharmacology , Neurons/drug effects , Superior Cervical Ganglion/drug effects , Animals , Atropine/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/physiology , Patch-Clamp Techniques , Phentolamine/pharmacology , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Nicotinic/metabolism , Sodium Channels/metabolism , Superior Cervical Ganglion/physiology , Yohimbine/pharmacology
17.
Eur J Pharmacol ; 748: 1-9, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25514605

ABSTRACT

Nicotine has been shown to have neuroprotective and neurotrophic actions in the central nervous system. To elucidate the peripheral neurotrophic effects of nicotine, we determined whether nicotine affected the reinnervation of mesenteric perivascular nerves following a topical phenol treatment. A topical phenol treatment was applied to the superior mesenteric artery proximal to the abdominal aorta in Wistar rats. We examined the immunohistochemistry of the distal small arteries 7 days after the treatment. The topical phenol treatment markedly reduced the density of tyrosine hydroxylase (TH)-LI and calcitonin gene-related peptide (CGRP)-LI fibers in these arteries. The administration of nicotine at a dose of 3 mg/kg/day (1.5 mg/kg/injection, twice a day), but not once a day or its continuous infusion using a mini-pump significantly increased the density of TH-LI nerves without affecting CGRP-LI nerves. A pretreatment with nicotinic acetylcholine receptor antagonists hexamethonium, mecamylamine, and methyllycaconitine, but not dextrometorphan, canceled the TH-LI nerve reinnervation induced by nicotine. Nicotine significantly increased NGF levels in the superior cervical ganglia (SCG) and mesenteric arteries, but not in the dorsal root ganglia, and also up-regulated the expression of NGF receptors (TrkA) in the SCG, which were canceled by hexamethonium. These results suggested that nicotine exhibited neurotrophic effects that facilitated the reinnervation of adrenergic TH-LI nerves by activating α7 nicotinic acetylcholine receptor and NGF in the SCG.


Subject(s)
Mesenteric Arteries/innervation , Nerve Fibers/drug effects , Nerve Fibers/physiology , Nerve Regeneration/drug effects , Nicotine/pharmacology , Phenol/adverse effects , Animals , Calcitonin Gene-Related Peptide/metabolism , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , Gene Expression Regulation/drug effects , Nerve Growth Factors/metabolism , Rats , Rats, Wistar , Receptor, trkA/metabolism , Receptors, Nicotinic/metabolism , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/drug effects , Superior Cervical Ganglion/physiology , Tyrosine 3-Monooxygenase/metabolism
18.
J Physiol ; 593(4): 803-23, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25398531

ABSTRACT

KEY POINTS: The synaptic organization of paravertebral sympathetic ganglia enables them to relay activity from the spinal cord to the periphery and thereby control autonomic functions, including blood pressure and body temperature. The present experiments were done to reconcile conflicting observations in tissue culture, intact isolated ganglia and living animals. By recording intracellularly from dissociated neurons and intact ganglia, we found that when electrode damage makes cells leaky it could profoundly distort cellular excitability and the integration of synaptic potentials. The experiments relied on the dynamic clamp method, which allows the creation of virtual ion channels by injecting current into a cell based upon a mathematical model and using rapid feedback between the model and cell. The results support the hypothesis that sympathetic ganglia can produce a 2.4-fold amplification of presynaptic activity. This could aid understanding of the neural hyperactivity that is believed to drive high blood pressure in some patients. ABSTRACT: The excitability of rat sympathetic neurons and integration of nicotinic EPSPs were compared in primary cell culture and in the acutely isolated intact superior cervical ganglion using whole cell patch electrode recordings. When repetitive firing was classified by Hodgkin's criteria in cultured cells, 18% displayed tonic class 1 excitability, 36% displayed adapting class 2 excitability and 46% displayed phasic class 3 excitability. In the intact ganglion, 71% of cells were class 1 and 29% were class 2. This diverges from microelectrode reports that nearly 100% of superior cervical ganglion neurons show phasic class 3 firing. The hypothesis that the disparity between patch and microelectrode data arises from a shunt conductance was tested using the dynamic clamp in cell culture. Non-depolarizing shunts of 3-10 nS converted cells from classes 1 and 2 to class 3 dynamics with current-voltage relations that replicated microelectrode data. Primary and secondary EPSPs recorded from the intact superior cervical ganglion were modelled as virtual synapses in cell culture using the dynamic clamp. Stimulating sympathetic neurons with virtual synaptic activity, designed to replicate in vivo recordings of EPSPs in muscle vasoconstrictor neurons, produced a 2.4-fold amplification of presynaptic activity. This gain in postsynaptic output did not differ between neurons displaying the three classes of excitability. Mimicry of microelectrode damage by virtual leak channels reduced and eventually obliterated synaptic gain by inhibiting summation of subthreshold EPSPs. These results provide a framework for interpreting sympathetic activity recorded from intact animals and support the hypothesis that paravertebral ganglia function as activity-dependent amplifiers of spinal output from preganglionic circuitry.


Subject(s)
Neurons/physiology , Superior Cervical Ganglion/physiology , Action Potentials/physiology , Animals , Cells, Cultured , Excitatory Postsynaptic Potentials/physiology , In Vitro Techniques , Male , Rats , Rats, Sprague-Dawley , Synapses/physiology
19.
Mol Cell Neurosci ; 63: 124-31, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25447945

ABSTRACT

Facilitation and inactivation of P/Q-type Ca2+ currents mediated by Ca2+/calmodulin binding to Ca(V)2.1 channels contribute to facilitation and rapid depression of synaptic transmission, respectively. Other calcium sensor proteins displace calmodulin from its binding site and differentially modulate P/Q-type Ca2 + currents, resulting in diverse patterns of short-term synaptic plasticity. Neuronal calcium sensor-1 (NCS-1, frequenin) has been shown to enhance synaptic facilitation, but the underlying mechanism is unclear. We report here that NCS-1 directly interacts with IQ-like motif and calmodulin-binding domain in the C-terminal domain of Ca(V)2.1 channel. NCS-1 reduces Ca2 +-dependent inactivation of P/Q-type Ca2+ current through interaction with the IQ-like motif and calmodulin-binding domain without affecting peak current or activation kinetics. Expression of NCS-1 in presynaptic superior cervical ganglion neurons has no effect on synaptic transmission, eliminating effects of this calcium sensor protein on endogenous N-type Ca2+ currents and the endogenous neurotransmitter release machinery. However, in superior cervical ganglion neurons expressing wild-type Ca(V)2.1 channels, co-expression of NCS-1 induces facilitation of synaptic transmission in response to paired pulses and trains of depolarizing stimuli, and this effect is lost in Ca(V)2.1 channels with mutations in the IQ-like motif and calmodulin-binding domain. These results reveal that NCS-1 directly modulates Ca(V)2.1 channels to induce short-term synaptic facilitation and further demonstrate that CaS proteins are crucial in fine-tuning short-term synaptic plasticity.


Subject(s)
Calcium Channels, N-Type/metabolism , Neuronal Calcium-Sensor Proteins/metabolism , Neuropeptides/metabolism , Synapses/metabolism , Synaptic Transmission , Amino Acid Motifs , Animals , Binding Sites , Calcium Channels, N-Type/chemistry , Cells, Cultured , HEK293 Cells , Humans , Mice , Neuronal Calcium-Sensor Proteins/genetics , Neuropeptides/genetics , Protein Binding , Rats , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/metabolism , Superior Cervical Ganglion/physiology , Synapses/physiology
20.
J Dent Res ; 93(11): 1163-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25252876

ABSTRACT

The sympathetic nervous system (SNS) regulates bone resorption through ß-2 adrenergic receptor (Adrb2). In orthodontic tooth movement (OTM), mechanical force induces and regulates alveolar bone remodeling. Compressive force-associated osteoclast differentiation and alveolar bone resorption are the rate-limiting steps of tooth movement. However, whether mechanical force can activate Adrb2 and thus contribute to OTM remains unknown. In this study, orthodontic nickel-titanium springs were applied to the upper first molars of rats and Adrb1/2(-/-) mice to confirm the role of SNS and Adrb2 in OTM. The results showed that blockage of SNS activity in the jawbones of rats by means of superior cervical ganglion ectomy reduced OTM distance from 860 to 540 µm after 14 d of force application. In addition, the injection of nonselective Adrb2 agonist isoproterenol activated the downstream signaling of SNS to accelerate OTM from 300 to 540 µm after 7 d of force application. Adrb1/2(-/-) mice showed significantly reduced OTM distance (19.5 µm) compared with the wild-type mice (107.6 µm) after 7 d of force application. Histopathologic analysis showed that the number of Adrb2-positive cells increased in the compressive region of periodontal ligament after orthodontic force was applied on rats. Mechanistically, mechanical compressive force upregulated Adrb2 expression in primary-cultured human periodontal ligament cells (PDLCs) through the elevation of intracellular Ca(2+) concentration. Activation of Adrb2 in PDLCs increased the RANKL/OPG ratio and promoted the peripheral blood mononuclear cell differentiation to osteoclasts in the cocultured system. Upregulation of Adrb2 in PDLCs promoted osteoclastogenesis, which accelerated OTM through Adrb2-enhanced bone resorption. In summary, this study suggests that mechanical force-induced Adrb2 activation in PDLCs contributes to SNS-regulated OTM.


Subject(s)
Periodontal Ligament/metabolism , Receptors, Adrenergic, beta-2/metabolism , Tooth Movement Techniques , Adrenergic beta-Agonists/pharmacology , Alveolar Process/cytology , Alveolar Process/physiology , Animals , Biomechanical Phenomena , Bone Remodeling/physiology , Bone Resorption/pathology , Bone Resorption/physiopathology , Calcium Signaling/physiology , Cell Differentiation/physiology , Cells, Cultured , Coculture Techniques , Ganglionectomy , Humans , Isoproterenol/pharmacology , Male , Mice , Mice, Knockout , Orthodontic Wires , Osteoclasts/physiology , Osteoprotegerin/analysis , Periodontal Ligament/cytology , RANK Ligand/analysis , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Stress, Mechanical , Superior Cervical Ganglion/physiology , Sympathetic Nervous System/physiology
SELECTION OF CITATIONS
SEARCH DETAIL