Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Pflugers Arch ; 473(6): 953-968, 2021 06.
Article in English | MEDLINE | ID: mdl-33881614

ABSTRACT

Action potentials play an important role in neurotransmitter release in response to taste. Here, I have investigated voltage-gated Na+ channels, a primary component of action potentials, in respective cell types of mouse fungiform taste bud cells (TBCs) with in situ whole-cell clamping and single-cell RT-PCR techniques. The cell types of TBCs electrophysiologically examined were determined immunohistochemically using the type III inositol 1,4,5-triphoshate receptor as a type II cell marker and synaptosomal-associated protein 25 as a type III cell marker. I show that type II cells, type III cells, and TBCs not immunoreactive to these markers (likely type I cells) generate voltage-gated Na+ currents. The recovery following inactivation of these currents was well fitted with double exponential curves. The time constants in type III cells (~20 ms and ~ 1 s) were significantly slower than respective time constants in other cell types. RT-PCR analysis indicated the expression of Nav1.3, Nav1.5, Nav1.6, and ß1 subunit mRNAs in TBCs. Pharmacological inhibition and single-cell RT-PCR studies demonstrated that type II and type III cells principally express tetrodotoxin (TTX)-sensitive Nav1.3 channels and that ~ 30% of type I cells express TTX-resistant Nav1.5 channels. The auxiliary ß1 subunit that modulates gating kinetics was rarely detected in TBCs. As the ß1 subunit co-expressed with an α subunit is known to accelerate the recovery from inactivation, it is likely that voltage-gated Na+ channels in TBCs may function without ß subunits. Slow recovery from inactivation, especially in type III cells, may limit high-frequency firing in response to taste substances.


Subject(s)
Ion Channel Gating , NAV1.3 Voltage-Gated Sodium Channel/metabolism , Taste Buds/metabolism , Action Potentials , Animals , Mice , Protein Subunits/metabolism , Sodium Channel Blockers/pharmacology , Taste Buds/cytology , Taste Buds/physiology
2.
Cell Biol Int ; 45(11): 2294-2303, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34296787

ABSTRACT

This study aimed to investigate the functions of miR-214-3p in diabetic neuropathic rodents. The diabetic neuropathy was induced by intraperitoneal injection of streptozotocin (STZ) in rats, and miR-214-3p was delivered via tail vein injection of lentivirus. Hot or cold stimulus tests demonstrated that STZ induced thermal hyperalgesia. Neurophysiological measurements revealed that motor and sensory nerve conduction velocity and nerve blood flow were decreased in diabetic neuropathic rats. However, the STZ-induced hyperalgesia, and reduced nerve conduction velocity and nerve blood flow were all significantly reversed by miR-214-3p administration. HE staining, TUNEL, ELISA, and immunoblotting demonstrated that STZ led to obvious pathological lesion, cell apoptosis, and inflammation in dorsal root ganglion (DRG), evidenced by altered levels of apoptosis-related protein molecules and inflammatory factors, and activation of Toll-like receptor 4 (TLR4)/myeloid differentiation primary response gene 88/nuclear factor kappa B signaling. The pathological alterations in diabetic neuropathic rats in DRG were significantly ameliorated by miR-214-3p application. In addition, sodium channel protein type 3 subunit alpha isoform 1 (Nav1.3) and TLR4 were identified as targets of miR-214-3p via dual-luciferase reporter assay. MiR-214-3p may play its roles by downregulating Nav1.3 and TLR4. In summary, miR-214-3p alleviated diabetes-induced nerve injury, and pathological lesion, cell apoptosis, and inflammation in DRG by regulating Nav1.3 and TLR4 in STZ-induced rats. These findings may provide novel therapeutic targets for clinical treatment of diabetic neuropathy.


Subject(s)
Diabetic Neuropathies/genetics , MicroRNAs/genetics , NAV1.3 Voltage-Gated Sodium Channel/metabolism , Toll-Like Receptor 4/metabolism , Animals , Apoptosis , Diabetes Mellitus, Experimental/genetics , Diabetic Neuropathies/physiopathology , Disease Models, Animal , Ganglia, Spinal/metabolism , Hyperalgesia/physiopathology , Inflammation/metabolism , Male , MicroRNAs/metabolism , NAV1.3 Voltage-Gated Sodium Channel/genetics , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Streptozocin/pharmacology , Toll-Like Receptor 4/genetics
3.
Epilepsia ; 61(3): 387-399, 2020 03.
Article in English | MEDLINE | ID: mdl-32090326

ABSTRACT

OBJECTIVE: Voltage-gated sodium channels (SCNs) share similar amino acid sequence, structure, and function. Genetic variants in the four human brain-expressed SCN genes SCN1A/2A/3A/8A have been associated with heterogeneous epilepsy phenotypes and neurodevelopmental disorders. To better understand the biology of seizure susceptibility in SCN-related epilepsies, our aim was to determine similarities and differences between sodium channel disorders, allowing us to develop a broader perspective on precision treatment than on an individual gene level alone. METHODS: We analyzed genotype-phenotype correlations in large SCN-patient cohorts and applied variant constraint analysis to identify severe sodium channel disease. We examined temporal patterns of human SCN expression and correlated functional data from in vitro studies with clinical phenotypes across different sodium channel disorders. RESULTS: Comparing 865 epilepsy patients (504 SCN1A, 140 SCN2A, 171 SCN8A, four SCN3A, 46 copy number variation [CNV] cases) and analysis of 114 functional studies allowed us to identify common patterns of presentation. All four epilepsy-associated SCN genes demonstrated significant constraint in both protein truncating and missense variation when compared to other SCN genes. We observed that age at seizure onset is related to SCN gene expression over time. Individuals with gain-of-function SCN2A/3A/8A missense variants or CNV duplications share similar characteristics, most frequently present with early onset epilepsy (<3 months), and demonstrate good response to sodium channel blockers (SCBs). Direct comparison of corresponding SCN variants across different SCN subtypes illustrates that the functional effects of variants in corresponding channel locations are similar; however, their clinical manifestation differs, depending on their role in different types of neurons in which they are expressed. SIGNIFICANCE: Variant function and location within one channel can serve as a surrogate for variant effects across related sodium channels. Taking a broader view on precision treatment suggests that in those patients with a suspected underlying genetic epilepsy presenting with neonatal or early onset seizures (<3 months), SCBs should be considered.


Subject(s)
Epileptic Syndromes/genetics , NAV1.1 Voltage-Gated Sodium Channel/genetics , NAV1.2 Voltage-Gated Sodium Channel/genetics , NAV1.3 Voltage-Gated Sodium Channel/genetics , NAV1.6 Voltage-Gated Sodium Channel/genetics , Sodium Channels/genetics , Age of Onset , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/physiopathology , Child , Child, Preschool , Codon, Nonsense , DNA Copy Number Variations , Electroencephalography , Epileptic Syndromes/drug therapy , Epileptic Syndromes/physiopathology , Female , Gain of Function Mutation , Gene Deletion , Gene Duplication , Gene Expression , Gene Expression Regulation, Developmental , Genotype , Humans , Infant , Infant, Newborn , Loss of Function Mutation , Male , Mutation, Missense , NAV1.1 Voltage-Gated Sodium Channel/metabolism , NAV1.2 Voltage-Gated Sodium Channel/metabolism , NAV1.3 Voltage-Gated Sodium Channel/metabolism , NAV1.6 Voltage-Gated Sodium Channel/metabolism , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/physiopathology , Phenotype , Sodium Channel Blockers/therapeutic use , Sodium Channels/metabolism
4.
J Physiol ; 596(9): 1601-1626, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29441586

ABSTRACT

KEY POINTS: Na+ current inactivation is biphasic in insulin-secreting cells, proceeding with two voltage dependences that are half-maximal at ∼-100 mV and -60 mV. Inactivation of voltage-gated Na+ (NaV ) channels occurs at ∼30 mV more negative voltages in insulin-secreting Ins1 and primary ß-cells than in HEK, CHO or glucagon-secreting αTC1-6 cells. The difference in inactivation between Ins1 and non-ß-cells persists in the inside-out patch configuration, discounting an involvement of a diffusible factor. In Ins1 cells and primary ß-cells, but not in HEK cells, inactivation of a single NaV subtype is biphasic and follows two voltage dependences separated by 30-40 mV. We propose that NaV channels adopt different inactivation behaviours depending on the local membrane environment. ABSTRACT: Pancreatic ß-cells are equipped with voltage-gated Na+ channels that undergo biphasic voltage-dependent steady-state inactivation. A small Na+ current component (10-15%) inactivates over physiological membrane potentials and contributes to action potential firing. However, the major Na+ channel component is completely inactivated at -90 to -80 mV and is therefore inactive in the ß-cell. It has been proposed that the biphasic inactivation reflects the contribution of different NaV α-subunits. We tested this possibility by expression of TTX-resistant variants of the NaV subunits found in ß-cells (NaV 1.3, NaV 1.6 and NaV 1.7) in insulin-secreting Ins1 cells and in non-ß-cells (including HEK and CHO cells). We found that all NaV subunits inactivated at 20-30 mV more negative membrane potentials in Ins1 cells than in HEK or CHO cells. The more negative inactivation in Ins1 cells does not involve a diffusible intracellular factor because the difference between Ins1 and CHO persisted after excision of the membrane. NaV 1.7 inactivated at 15--20 mV more negative membrane potentials than NaV 1.3 and NaV 1.6 in Ins1 cells but this small difference is insufficient to solely explain the biphasic inactivation in Ins1 cells. In Ins1 cells, but never in the other cell types, widely different components of NaV inactivation (separated by 30 mV) were also observed following expression of a single type of NaV α-subunit. The more positive component exhibited a voltage dependence of inactivation similar to that found in HEK and CHO cells. We propose that biphasic NaV inactivation in insulin-secreting cells reflects insertion of channels in membrane domains that differ with regard to lipid and/or membrane protein composition.


Subject(s)
Gene Expression Regulation , Insulin-Secreting Cells/metabolism , Insulinoma/metabolism , NAV1.3 Voltage-Gated Sodium Channel/chemistry , NAV1.6 Voltage-Gated Sodium Channel/chemistry , NAV1.7 Voltage-Gated Sodium Channel/chemistry , Sodium Channel Blockers/pharmacology , Action Potentials , Amino Acid Sequence , Animals , Cricetinae , Cricetulus , Electrophysiological Phenomena , HEK293 Cells , Humans , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Insulinoma/drug therapy , Insulinoma/pathology , Membrane Potentials , Mice , Mice, Knockout , NAV1.3 Voltage-Gated Sodium Channel/metabolism , NAV1.6 Voltage-Gated Sodium Channel/metabolism , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Rats , Sequence Homology , Sodium/metabolism
5.
Mol Pain ; 14: 1744806918778491, 2018.
Article in English | MEDLINE | ID: mdl-29783906

ABSTRACT

Background Oral administration of Bulleyaconitine A, an extracted diterpenoid alkaloid from Aconitum bulleyanum plants, is effective for treating chronic pain in rats and in human patients, but the underlying mechanisms are poorly understood. Results As the hyperexcitability of dorsal root ganglion neurons resulting from the upregulation of voltage-gated sodium (Nav) channels has been proved critical for development of chronic pain, we tested the effects of Bulleyaconitine A on Nav channels in rat spared nerve injury model of neuropathic pain. We found that Bulleyaconitine A at 5 nM increased the threshold of action potentials and reduced the firing rate of dorsal root ganglion neurons in spared nerve injury rats but not in sham rats. Bulleyaconitine A preferably blocked tetrodotoxin-sensitive Nav channels over tetrodotoxin-resistant ones in dorsal root ganglion neurons of spared nerve injury rats. Bulleyaconitine A was more potent for blocking Nav1.3 and Nav1.7 than Nav1.8 in cell lines. The half maximal inhibitory concentration (IC50) values for resting Nav1.3, Nav1.7, and Nav1.8 were 995.6 ± 139.1 nM, 125.7 ± 18.6 nM, and 151.2 ± 15.4 µM, respectively, which were much higher than those for inactivated Nav1.3 (20.3 ± 3.4 pM), Nav1.7 (132.9 ± 25.5 pM), and Nav1.8 (18.0 ± 2.5 µM). The most profound use-dependent blocking effect of Bulleyaconitine A was observed on Nav1.7, less on Nav1.3, and least on Nav1.8 at IC50 concentrations. Bulleyaconitine A facilitated the inactivation of Nav channels in each subtype. Conclusions Preferably blocking tetrodotoxin-sensitive Nav1.7 and Nav1.3 in dorsal root ganglion neurons may contribute to Bulleyaconitine A's antineuropathic pain effect.


Subject(s)
Aconitine/analogs & derivatives , Ganglia, Spinal/pathology , NAV1.3 Voltage-Gated Sodium Channel/metabolism , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Nerve Tissue/injuries , Neurons/metabolism , Aconitine/pharmacology , Animals , Cell Line , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Humans , Ion Channel Gating/drug effects , Male , Nerve Tissue/drug effects , Nerve Tissue/metabolism , Nerve Tissue/pathology , Neurons/drug effects , Neurons/pathology , Rats, Sprague-Dawley
6.
J Pharmacol Sci ; 137(1): 93-97, 2018 May.
Article in English | MEDLINE | ID: mdl-29773519

ABSTRACT

The neurosteroid allopregnanolone has potent analgesic effects, and its potential use for neuropathic pain is supported by recent reports. However, the analgesic mechanisms are obscure. The voltage-gated sodium channels (Nav) α subunit Nav1.3 is thought to play an essential role in neuropathic pain. Here, we report the effects of allopregnanolone sulfate (APAS) on sodium currents (INa) in Xenopus oocytes expressing Nav1.3 with ß1 or ß3 subunits. APAS suppressed INa of Nav1.3 with ß1 and ß3 in a concentration-dependent manner (IC50 values; 75 and 26 µmol/L). These results suggest the possible importance of Nav1.3 inhibition for the analgesic mechanisms of allopregnanolone.


Subject(s)
Analgesics , NAV1.3 Voltage-Gated Sodium Channel/metabolism , Neurotransmitter Agents/pharmacology , Oocytes/metabolism , Pregnanolone/pharmacology , Voltage-Gated Sodium Channel Blockers , Animals , Dose-Response Relationship, Drug , Female , Neuralgia/drug therapy , Neurotransmitter Agents/therapeutic use , Pregnanolone/therapeutic use , Xenopus
7.
Chin J Physiol ; 61(2): 124-129, 2018 Apr 30.
Article in English | MEDLINE | ID: mdl-29689688

ABSTRACT

Diabetes is a common metabolic disease which leads to diabetic peripheral neuropathy. Recently, the role of microRNA-96 (miR-96) in alleviating neuropathic pain by inhibiting the expression of NaV1.3, an isoform of voltage-gated sodium channels, has been shown. Peripheral nerve injuries result in NaV1.3 elevation. Exercise has beneficial role in diabetes management and peripheral neuropathy. However, the effects of exercise on miR-96 and its target gene NaV1.3 in diabetic rats are unknown. Therefore, the present study investigated the effects of exercise training on the expression of miR-96 and NaV1.3 in diabetic rats. For this purpose, rats were randomly divided into four groups: control, exercise, diabetic and diabetic-exercise groups. Type 2 diabetes was induced by a high-fat diet and the administration of streptozotocin (STZ) (35 mg/kg, i.p.). The exercise groups were subjected to swimming exercise 5 days/week for 10 weeks. At the end of the treatment period, thermal pain threshold, determined through the tail-flick test, and the expression levels of miR-96 and its target gene NaV1.3 were determined by reverse transcription (RT)-PCR in the sciatic nerve tissues of the rats. Data of the present study indicated that diabetes diminished miR-96 expression levels, but significantly upregulated NaV1.3 expression in the sciatic nerve. On exercise training, miR-96 expression was reversed with concurrent down-regulation of the NaV1.3 expression. This study introduced a new and potential miRNA-dependent mechanism for exerciseinduced protective effects against diabetic thermal hyperalgesia.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 2/therapy , Diabetic Neuropathies/therapy , Exercise Therapy/methods , MicroRNAs/metabolism , NAV1.3 Voltage-Gated Sodium Channel/metabolism , Sciatic Nerve/metabolism , Swimming , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Diabetic Neuropathies/genetics , Diabetic Neuropathies/metabolism , Diabetic Neuropathies/physiopathology , Diet, High-Fat , Gene Expression Regulation , Hyperalgesia/genetics , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Hyperalgesia/therapy , Male , MicroRNAs/genetics , NAV1.3 Voltage-Gated Sodium Channel/genetics , Pain Threshold , Rats, Wistar , Sciatic Nerve/physiopathology , Streptozocin , Time Factors
8.
Biochim Biophys Acta ; 1848(7): 1545-51, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25838126

ABSTRACT

With the ultimate goal of detailed structural analysis of mammalian and particularly human voltage-gated sodium channels (VGSCs), we have investigated the relative stability of human and rat VGSCs and compared them with electric eel VGSC. We found that NaV1.3 from rat was the most stable after detergent solubilisation. The order of stability was rNaV1.3>hNaV1.2>hNaV1.1>hNaV1.6>hNaV1.3>hNaV1.4. However, a comparison with the VGSC from Electrophorus electricus, which is most similar to NaV1.4, shows that the eel VGSC is considerably more stable in detergent than the human VGSCs examined. We conclude that current methods of structural analysis, such as single particle electron cryomicroscopy (cryoEM), may be most usefully targeted to eel VGSC or rNaV1.3, but that structural analysis on the full spectrum of VGSCs, by methods that require greater stability such as crystallisation and X-ray crystallography, will require further stabilisation of the channel.


Subject(s)
Benchmarking/methods , Detergents/chemistry , Membrane Proteins/chemistry , Voltage-Gated Sodium Channels/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Cryoelectron Microscopy , Crystallography, X-Ray , Eels/metabolism , HEK293 Cells , Humans , Membrane Proteins/metabolism , Membrane Proteins/ultrastructure , NAV1.1 Voltage-Gated Sodium Channel/chemistry , NAV1.1 Voltage-Gated Sodium Channel/metabolism , NAV1.1 Voltage-Gated Sodium Channel/ultrastructure , NAV1.2 Voltage-Gated Sodium Channel/chemistry , NAV1.2 Voltage-Gated Sodium Channel/metabolism , NAV1.2 Voltage-Gated Sodium Channel/ultrastructure , NAV1.3 Voltage-Gated Sodium Channel/chemistry , NAV1.3 Voltage-Gated Sodium Channel/metabolism , NAV1.3 Voltage-Gated Sodium Channel/ultrastructure , Protein Stability/drug effects , Rats , Solubility , Temperature , Toxins, Biological/metabolism , Toxins, Biological/pharmacology , Voltage-Gated Sodium Channels/metabolism , Voltage-Gated Sodium Channels/ultrastructure
9.
Proc Natl Acad Sci U S A ; 110(29): E2724-32, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23818614

ABSTRACT

Voltage-gated sodium (Nav) channels play a fundamental role in the generation and propagation of electrical impulses in excitable cells. Here we describe two unique structurally related nanomolar potent small molecule Nav channel inhibitors that exhibit up to 1,000-fold selectivity for human Nav1.3/Nav1.1 (ICA-121431, IC50, 19 nM) or Nav1.7 (PF-04856264, IC50, 28 nM) vs. other TTX-sensitive or resistant (i.e., Nav1.5) sodium channels. Using both chimeras and single point mutations, we demonstrate that this unique class of sodium channel inhibitor interacts with the S1-S4 voltage sensor segment of homologous Domain 4. Amino acid residues in the "extracellular" facing regions of the S2 and S3 transmembrane segments of Nav1.3 and Nav1.7 seem to be major determinants of Nav subtype selectivity and to confer differences in species sensitivity to these inhibitors. The unique interaction region on the Domain 4 voltage sensor segment is distinct from the structural domains forming the channel pore, as well as previously characterized interaction sites for other small molecule inhibitors, including local anesthetics and TTX. However, this interaction region does include at least one amino acid residue [E1559 (Nav1.3)/D1586 (Nav1.7)] that is important for Site 3 α-scorpion and anemone polypeptide toxin modulators of Nav channel inactivation. The present study provides a potential framework for identifying subtype selective small molecule sodium channel inhibitors targeting interaction sites away from the pore region.


Subject(s)
Acetamides/pharmacology , Electrophysiological Phenomena/physiology , NAV1.3 Voltage-Gated Sodium Channel/metabolism , Thiazoles/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacology , Amino Acid Motifs/genetics , Binding Sites/genetics , HEK293 Cells , Humans , Inhibitory Concentration 50 , Molecular Sequence Data , NAV1.3 Voltage-Gated Sodium Channel/genetics , Patch-Clamp Techniques , Sequence Alignment
10.
J Biol Phys ; 42(3): 351-70, 2016 06.
Article in English | MEDLINE | ID: mdl-27072680

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a lethal paralytic disease caused by the degeneration of motor neurons in the spinal cord, brain stem, and motor cortex. Mutations in the gene encoding copper/zinc superoxide dismutase (SOD1) are present in ~20% of familial ALS and ~2% of all ALS cases. The most common SOD1 gene mutation in North America is a missense mutation substituting valine for alanine (A4V). In this study, we analyze sodium channel currents in oocytes expressing either wild-type or mutant (A4V) SOD1 protein. We demonstrate that the A4V mutation confers a propensity to hyperexcitability on a voltage-dependent sodium channel (Nav1.3) mediated by heightened total Na(+) conductance and a hyperpolarizing shift in the voltage dependence of Nav1.3 activation. To estimate the impact of these channel effects on excitability in an intact neuron, we simulated these changes in the program NEURON; this shows that the changes induced by mutant SOD1 increase the spontaneous firing frequency of the simulated neuron. These findings are consistent with the view that excessive excitability of neurons is one component in the pathogenesis of this disease.


Subject(s)
Electrophysiological Phenomena/genetics , Mutation , NAV1.3 Voltage-Gated Sodium Channel/metabolism , Superoxide Dismutase-1/genetics , Animals , Humans , Neurons/cytology , Sodium/metabolism , Xenopus laevis
11.
Int J Mol Sci ; 17(9)2016 Sep 05.
Article in English | MEDLINE | ID: mdl-27608006

ABSTRACT

Diabetes mellitus (DM) is a common chronic medical problem worldwide; one of its complications is painful peripheral neuropathy, which can substantially erode quality of life and increase the cost of management. Despite its clinical importance, the pathogenesis of painful diabetic neuropathy (PDN) is complex and incompletely understood. Voltage-gated sodium channels (VGSCs) link many physiological processes to electrical activity by controlling action potentials in all types of excitable cells. Two isoforms of VGSCs, NaV1.3 and NaV1.7, which are encoded by the sodium voltage-gated channel alpha subunit 3 and 9 (Scn3A and Scn9A) genes, respectively, have been identified in both peripheral nociceptive neurons of dorsal root ganglion (DRG) and pancreatic islet cells. Recent advances in our understanding of tetrodotoxin-sensitive (TTX-S) sodium channels NaV1.3 and NaV1.7 lead to the rational doubt about the cause-effect relation between diabetes and painful neuropathy. In this review, we summarize the roles of NaV1.3 and NaV1.7 in islet cells and DRG neurons, discuss the link between DM and painful neuropathy, and present a model, which may provide a starting point for further studies aimed at identifying the mechanisms underlying diabetes and painful neuropathy.


Subject(s)
Diabetes Mellitus/metabolism , Diabetic Neuropathies/metabolism , NAV1.3 Voltage-Gated Sodium Channel/metabolism , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Animals , Humans , Islets of Langerhans/metabolism , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology
12.
J Physiol ; 593(14): 3109-22, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25969124

ABSTRACT

Voltage-gated sodium channels (VGSC) play a key role for initiating action potentials (AP) in excitable cells. VGSC in human lymphatic vessels have not been investigated. In the present study, we report the electrical activity and APs of small human lymphatic collecting vessels, as well as mRNA expression and function of VGSC in small and large human lymphatic vessels. The VGSC blocker TTX inhibited spontaneous contractions in six of 10 spontaneously active vessels, whereas ranolazine, which has a narrower VGSC blocking profile, had no influence on spontaneous activity. TTX did not affect noradrenaline-induced contractions. The VGSC opener veratridine induced contractions in a concentration-dependent manner (0.1-30 µm) eliciting a stable tonic contraction and membrane depolarization to -18 ± 0.6 mV. Veratridine-induced depolarizations and contractions were reversed ∼80% by TTX, and were dependent on Ca(2+) influx via L-type calcium channels and the sodium-calcium exchanger in reverse mode. Molecular analysis determined NaV 1.3 to be the predominantly expressed VGSC isoform. Electrophysiology of mesenteric lymphatics determined the resting membrane potential to be -45 ± 1.7 mV. Spontaneous APs were preceded by a slow depolarization of 5.3 ± 0.6 mV after which a spike was elicited that almost completely repolarized before immediately depolarizing again to plateau. Vessels transiently hyperpolarized prior to returning to the resting membrane potential. TTX application blocked APs. We have shown that VGSC are necessary for initiating and maintaining APs and spontaneous contractions in human lymphatic vessels and our data suggest the main contribution from comes NaV 1.3. We have also shown that activation of these channels augments the contractile activity of the vessels.


Subject(s)
Action Potentials , Lymphatic Vessels/physiology , Muscle Contraction , NAV1.3 Voltage-Gated Sodium Channel/metabolism , Sodium Channels/metabolism , Adult , Aged , Aged, 80 and over , Female , Humans , Lymphatic Vessels/drug effects , Lymphatic Vessels/metabolism , Male , Middle Aged , NAV1.3 Voltage-Gated Sodium Channel/genetics , Sodium Channel Blockers/pharmacology , Sodium Channels/genetics
13.
J Neurophysiol ; 114(3): 1554-64, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26156380

ABSTRACT

Gain-of-function missense mutations in voltage-gated sodium channel Nav1.7 have been linked to small-fiber neuropathy, which is characterized by burning pain, dysautonomia and a loss of intraepidermal nerve fibers. However, the mechanistic cascades linking Nav1.7 mutations to axonal degeneration are incompletely understood. The G856D mutation in Nav1.7 produces robust changes in channel biophysical properties, including hyperpolarized activation, depolarized inactivation, and enhanced ramp and persistent currents, which contribute to the hyperexcitability exhibited by neurons containing Nav1.8. We report here that cell bodies and neurites of dorsal root ganglion (DRG) neurons transfected with G856D display increased levels of intracellular Na(+) concentration ([Na(+)]) and intracellular [Ca(2+)] following stimulation with high [K(+)] compared with wild-type (WT) Nav1.7-expressing neurons. Blockade of reverse mode of the sodium/calcium exchanger (NCX) or of sodium channels attenuates [Ca(2+)] transients evoked by high [K(+)] in G856D-expressing DRG cell bodies and neurites. We also show that treatment of WT or G856D-expressing neurites with high [K(+)] or 2-deoxyglucose (2-DG) does not elicit degeneration of these neurites, but that high [K(+)] and 2-DG in combination evokes degeneration of G856D neurites but not WT neurites. Our results also demonstrate that 0 Ca(2+) or blockade of reverse mode of NCX protects G856D-expressing neurites from degeneration when exposed to high [K(+)] and 2-DG. These results point to [Na(+)] overload in DRG neurons expressing mutant G856D Nav1.7, which triggers reverse mode of NCX and contributes to Ca(2+) toxicity, and suggest subtype-specific blockade of Nav1.7 or inhibition of reverse NCX as strategies that might slow or prevent axon degeneration in small-fiber neuropathy.


Subject(s)
Calcium/metabolism , Erythromelalgia/metabolism , Ganglia, Spinal/metabolism , Mutation, Missense , NAV1.3 Voltage-Gated Sodium Channel/metabolism , Neurites/metabolism , Sodium Channels/metabolism , Sodium-Calcium Exchanger/metabolism , Animals , Calcium/toxicity , Cells, Cultured , Ganglia, Spinal/cytology , Humans , NAV1.3 Voltage-Gated Sodium Channel/genetics , Neurites/pathology , Potassium/metabolism , Rats , Rats, Sprague-Dawley , Sodium/metabolism , Sodium Channels/genetics , Sodium-Calcium Exchanger/antagonists & inhibitors
14.
J Biochem Mol Toxicol ; 29(3): 129-34, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25358543

ABSTRACT

Voltage-gated sodium channels (Na(v)) are essential for initiation and propagation of action potentials. Previous in vitro studies reported that exposure to the Na(v) toxins veratridine and α scorpion toxin cause persistent downregulation of Na(v) mRNA in vitro. However the mechanism of this downregulation is not well characterized. Here, we report that the type-II pyrethroid deltamethrin, which has a similar mechanism as these toxins, elicited an approximate 25% reduction in Na(v) 1.2 and Na(v) 1.3 mRNA in SK-N-AS cells. Deltamethrin-induced decreases of Na(v) mRNA were blocked with the Na(v) antagonist tetrodotoxin, demonstrating a primary role for interaction with Na(v). Pre-treatment with the intracellular calcium chelator BAPTA-AM and the calpain inhibitor PD-150606 also prevented these decreases, identifying a role for intracellular calcium and calpain activation. Because alterations in Na(v) expression and function can result in neurotoxicity, additional studies are warranted to determine whether or not such effects occur in vivo.


Subject(s)
Calcium/physiology , Calpain/physiology , Insecticides/pharmacology , NAV1.2 Voltage-Gated Sodium Channel/metabolism , NAV1.3 Voltage-Gated Sodium Channel/metabolism , Nitriles/pharmacology , Pyrethrins/pharmacology , Cell Line, Tumor , Down-Regulation , Humans , NAV1.2 Voltage-Gated Sodium Channel/genetics , NAV1.3 Voltage-Gated Sodium Channel/genetics , RNA, Messenger/metabolism
15.
J Physiol ; 592(21): 4677-96, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25172946

ABSTRACT

Mouse pancreatic ß- and α-cells are equipped with voltage-gated Na(+) currents that inactivate over widely different membrane potentials (half-maximal inactivation (V0.5) at -100 mV and -50 mV in ß- and α-cells, respectively). Single-cell PCR analyses show that both α- and ß-cells have Nav1.3 (Scn3) and Nav1.7 (Scn9a) α subunits, but their relative proportions differ: ß-cells principally express Nav1.7 and α-cells Nav1.3. In α-cells, genetically ablating Scn3a reduces the Na(+) current by 80%. In ß-cells, knockout of Scn9a lowers the Na(+) current by >85%, unveiling a small Scn3a-dependent component. Glucagon and insulin secretion are inhibited in Scn3a(-/-) islets but unaffected in Scn9a-deficient islets. Thus, Nav1.3 is the functionally important Na(+) channel α subunit in both α- and ß-cells because Nav1.7 is largely inactive at physiological membrane potentials due to its unusually negative voltage dependence of inactivation. Interestingly, the Nav1.7 sequence in brain and islets is identical and yet the V0.5 for inactivation is >30 mV more negative in ß-cells. This may indicate the presence of an intracellular factor that modulates the voltage dependence of inactivation.


Subject(s)
Glucagon-Secreting Cells/metabolism , Insulin-Secreting Cells/metabolism , NAV1.3 Voltage-Gated Sodium Channel/metabolism , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Sodium/physiology , Animals , Gene Expression Regulation , Glucagon-Secreting Cells/drug effects , Glucose , HEK293 Cells , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , NAV1.3 Voltage-Gated Sodium Channel/genetics , NAV1.7 Voltage-Gated Sodium Channel/genetics , Neurotoxins/pharmacology , Protein Isoforms , Protein Subunits
16.
J Biol Chem ; 288(34): 24316-31, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23836888

ABSTRACT

Voltage-gated sodium channel (NaV) trafficking is incompletely understood. Post-translational modifications of NaVs and/or auxiliary subunits and protein-protein interactions have been posited as NaV-trafficking mechanisms. Here, we tested if modification of the axonal collapsin response mediator protein 2 (CRMP2) by a small ubiquitin-like modifier (SUMO) could affect NaV trafficking; CRMP2 alters the extent of NaV slow inactivation conferred by the anti-epileptic (R)-lacosamide, implying NaV-CRMP2 functional coupling. Expression of a CRMP2 SUMOylation-incompetent mutant (CRMP2-K374A) in neuronal model catecholamine A differentiated (CAD) cells did not alter lacosamide-induced NaV slow inactivation compared with CAD cells expressing wild type CRMP2. Like wild type CRMP2, CRMP2-K374A expressed robustly in CAD cells. Neurite outgrowth, a canonical CRMP2 function, was moderately reduced by the mutation but was still significantly higher than enhanced GFP-transfected cortical neurons. Notably, huwentoxin-IV-sensitive NaV1.7 currents, which predominate in CAD cells, were significantly reduced in CAD cells expressing CRMP2-K374A. Increasing deSUMOylation with sentrin/SUMO-specific protease SENP1 or SENP2 in wild type CRMP2-expressing CAD cells decreased NaV1.7 currents. Consistent with a reduction in current density, biotinylation revealed a significant reduction in surface NaV1.7 levels in CAD cells expressing CRMP2-K374A; surface NaV1.7 expression was also decreased by SENP1 + SENP2 overexpression. Currents in HEK293 cells stably expressing NaV1.7 were reduced by CRMP2-K374A in a manner dependent on the E2-conjugating enzyme Ubc9. No decrement in current density was observed in HEK293 cells co-expressing CRMP2-K374A and NaV1.1 or NaV1.3. Diminution of sodium currents, largely NaV1.7, was recapitulated in sensory neurons expressing CRMP2-K374A. Our study elucidates a novel regulatory mechanism that utilizes CRMP2 SUMOylation to choreograph NaV1.7 trafficking.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Nerve Tissue Proteins/metabolism , Sensory Receptor Cells/metabolism , Sumoylation/physiology , Amino Acid Substitution , Animals , Catecholamines/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Endopeptidases/genetics , Endopeptidases/metabolism , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Mutation, Missense , NAV1.1 Voltage-Gated Sodium Channel/genetics , NAV1.1 Voltage-Gated Sodium Channel/metabolism , NAV1.3 Voltage-Gated Sodium Channel/genetics , NAV1.3 Voltage-Gated Sodium Channel/metabolism , NAV1.7 Voltage-Gated Sodium Channel/genetics , Nerve Tissue Proteins/genetics , Protein Transport/drug effects , Protein Transport/physiology , SUMO-1 Protein/genetics , SUMO-1 Protein/metabolism , Sensory Receptor Cells/cytology , Sodium Channels/genetics , Sodium Channels/metabolism , Sumoylation/drug effects , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
17.
Eur J Neurosci ; 39(10): 1682-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24612023

ABSTRACT

Members of the miR-183 family are unique in that they are highly abundant in sensory organs. In a recent study, significant downregulation was observed for miR-96 and miR-183 in the L5 dorsal root ganglion (DRG) 2 weeks after spinal nerve ligation (SNL). In this study, we focused on miR-183, which is the most regulated member of the miR-183 family, to look at the specific role on neuropathic pain. Persistent mechanical allodynia was induced with the L5 SNL model in 8-week-old male Sprague-Dawley rats. Paw withdrawal thresholds in response to mechanical stimuli were assessed with Von Frey filaments. Expression of miR-183 in the L5 DRG was assessed with quantitative real-time polymerase chain reaction (qPCR) analysis. Lentivirions expressing miR-183 were injected intrathecally into SNL rats. Changes in mechanical allodynia were assessed with Von Frey filaments. In addition, changes in the predicted target genes of miR-183 were assessed with qPCR. L5 SNL produced marked mechanical allodynia in the ipsilateral hindpaws of adult rats, beginning at postoperative day 1 and continuing to day 14. L5 SNL caused significant downregulation of miR-183 in adult DRG cells. Intrathecal administration of lentivirions expressing miR-183 downregulated SNL-induced increases in the expression of Nav1.3 and brain-derived neurotrophic factor (BDNF), which correlated with the significant attenuation of SNL-induced mechanical allodynia. Our results show that SNL-induced mechanical allodynia is significantly correlated with the decreased expression of miR-183 in DRG cells. Replacement of miR-183 downregulates SNL-induced increases in Nav1.3 and BDNF expression, and attenuates SNL-induced mechanical allodynia.


Subject(s)
Ganglia, Spinal/physiopathology , Hyperalgesia/physiopathology , MicroRNAs/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Down-Regulation , Gene Transfer Techniques , Genetic Vectors , Lentivirus/genetics , Male , MicroRNAs/genetics , NAV1.3 Voltage-Gated Sodium Channel/metabolism , Neuralgia/physiopathology , Pain Threshold/physiology , Physical Stimulation , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Spinal Nerves/injuries , Touch
18.
Brain ; 136(Pt 9): 2738-50, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23861446

ABSTRACT

Neuronal damage in the somatosensory system causes intractable chronic neuropathic pain. Plastic changes in sensory neuron excitability are considered the cellular basis of persistent pain. Non-coding microRNAs modulate specific gene translation to impact on diverse cellular functions and their dysregulation causes various diseases. However, their significance in adult neuronal functions and disorders is still poorly understood. Here, we show that miR-7a is a key functional RNA sustaining the late phase of neuropathic pain through regulation of neuronal excitability in rats. In the late phase of neuropathic pain, microarray analysis identified miR-7a as the most robustly decreased microRNA in the injured dorsal root ganglion. Moreover, local induction of miR-7a, using an adeno-associated virus vector, in sensory neurons of injured dorsal root ganglion, suppressed established neuropathic pain. In contrast, miR-7a overexpression had no effect on acute physiological or inflammatory pain. Furthermore, miR-7a downregulation was sufficient to cause pain-related behaviours in intact rats. miR-7a targeted the ß2 subunit of the voltage-gated sodium channel, and decreased miR-7a associated with neuropathic pain caused increased ß2 subunit protein expression, independent of messenger RNA levels. Consistently, miR-7a overexpression in primary sensory neurons of injured dorsal root ganglion suppressed increased ß2 subunit expression and normalized long-lasting hyperexcitability of nociceptive neurons. These findings demonstrate miR-7a downregulation is causally involved in maintenance of neuropathic pain through regulation of neuronal excitability, and miR-7a replenishment offers a novel therapeutic strategy specific for chronic neuropathic pain.


Subject(s)
MicroRNAs/genetics , MicroRNAs/metabolism , Neuralgia/pathology , Neuralgia/therapy , Nociceptors/metabolism , Nociceptors/physiology , Action Potentials/drug effects , Action Potentials/genetics , Analysis of Variance , Animals , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Down-Regulation/genetics , Down-Regulation/physiology , Ganglia, Spinal/pathology , Green Fluorescent Proteins/metabolism , Hyperalgesia/metabolism , Hyperalgesia/therapy , Male , MicroRNAs/therapeutic use , Microarray Analysis , NAV1.3 Voltage-Gated Sodium Channel/genetics , NAV1.3 Voltage-Gated Sodium Channel/metabolism , Neuralgia/metabolism , Nociceptors/drug effects , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Transfection
19.
Neurosci Lett ; 832: 137806, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38714229

ABSTRACT

BACKGROUND: Trigeminal neuralgia (TN) is a common and difficult-to-treat neuropathic pain disorder in clinical practice. Previous studies have shown that Toll-like receptor 4 (TLR4) modulates the activation of the NF-κB pathway to affect neuropathic pain in rats. Voltage-gated sodium channels (VGSCs) are known to play an important role in neuropathic pain electrical activity. OBJECTIVE: To investigate whether TLR4 can regulate Nav1.3 through the TRAF6/NF-κB p65 pathway after infraorbital nerve chronic constriction injury (ION-CCI). STUDY DESIGN: ION-CCI modeling was performed on SD (Sprague Dawley) rats. To verify the success of the modeling, we need to detect the mechanical pain threshold and ATF3. Then, detecting the expression of TLR4, TRAF6, NF-κB p65, p-p65, and Nav1.3 in rat TG. Subsequently, investigate the role of TLR4/TRAF6/NF-κB pathway in ION-CCI model by intrathecal injections of LPS-rs (TLR4 antagonist), C25-140 (TRAF6 inhibitor), and PDTC (NF-κB p65 inhibitor). RESULTS: ION-CCI surgery decreased the mechanical pain threshold of rats and increased the expression of ATF3, TLR4, TRAF6, NF-κB p-p65 and Nav1.3, but there was no difference in NF-κB p65 expression. After inject antagonist or inhibitor of the TLR4/TRAF6/NF-κB pathway, the expression of Nav1.3 was decreased and mechanical pain threshold was increased. CONCLUSION: In the rat model of ION-CCI, TLR4 in the rat trigeminal ganglion regulates Nav1.3 through the TRAF6/NF-κB p65 pathway, and TLR4 antagonist alleviates neuropathic pain in ION-CCI rats.


Subject(s)
NAV1.3 Voltage-Gated Sodium Channel , Rats, Sprague-Dawley , Signal Transduction , TNF Receptor-Associated Factor 6 , Toll-Like Receptor 4 , Animals , Toll-Like Receptor 4/metabolism , TNF Receptor-Associated Factor 6/metabolism , Male , NAV1.3 Voltage-Gated Sodium Channel/metabolism , Signal Transduction/physiology , NF-kappa B/metabolism , Trigeminal Neuralgia/metabolism , Rats , Disease Models, Animal , Transcription Factor RelA/metabolism , Activating Transcription Factor 3/metabolism , Pain Threshold/physiology
20.
Am J Physiol Cell Physiol ; 304(8): C739-47, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23364266

ABSTRACT

This study investigated the molecular identity and impact of enhancing voltage-gated Na(+) (Na(V)) channels in the control of vascular tone. In rat isolated mesenteric and femoral arteries mounted for isometric tension recording, the vascular actions of the Na(V) channel activator veratridine were examined. Na(V) channel expression was probed by molecular techniques and immunocytochemistry. In mesenteric arteries, veratridine induced potent contractions (pEC(50) = 5.19 ± 0.20, E(max) = 12.0 ± 2.7 mN), which were inhibited by 1 µM TTX (a blocker of all Na(V) channel isoforms, except Na(V)1.5, Na(V)1.8, and Na(V)1.9), but not by selective blockers of Na(V)1.7 (ProTx-II, 10 nM) or Na(V)1.8 (A-80347, 1 µM) channels. The responses were insensitive to endothelium removal but were partly (~60%) reduced by chemical destruction of sympathetic nerves by 6-hydroxydopamine (2 mM) or antagonism at the α1-adrenoceptor by prazosin (1 µM). KB-R7943, a blocker of the reverse mode of the Na(+)/Ca(2+) exchanger (3 µM), inhibited veratridine contractions in the absence or presence of prazosin. T16A(inh)-A01, a Ca(2+)-activated Cl(-) channel blocker (10 µM), also inhibited the prazosin-resistant contraction to veratridine. Na(V) channel immunoreactivity was detected in freshly isolated mesenteric myocytes, with apparent colocalization with the Na(+)/Ca(2+) exchanger. Veratridine induced similar contractile effects in the femoral artery, and mRNA transcripts for Na(V)1.2 and Na(V)1.3 channels were evident in both vessel types. We conclude that, in addition to sympathetic nerves, NaV channels are expressed in vascular myocytes, where they are functionally coupled to the reverse mode of Na(+)/Ca(2+) exchanger and subsequent activation of Ca(2+)-activated Cl(-) channels, causing contraction. The TTX-sensitive Na(V)1.2 and Na(V)1.3 channels are likely involved in vascular control.


Subject(s)
NAV1.2 Voltage-Gated Sodium Channel/metabolism , NAV1.3 Voltage-Gated Sodium Channel/metabolism , Vasoconstriction/physiology , Animals , Male , Mesenteric Arteries/physiology , Muscle, Smooth, Vascular/metabolism , Rats , Rats, Wistar , Sodium Channel Blockers/pharmacology , Veratridine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL