Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
J Neurophysiol ; 126(3): 827-839, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34320850

ABSTRACT

Small fiber neuropathy (SFN) is a common condition affecting thinly myelinated Aδ and unmyelinated C fibers, often resulting in excruciating pain and dysautonomia. SFN has been associated with several conditions, but a significant number of cases have no discernible cause. Recent genetic studies have identified potentially pathogenic gain-of-function mutations in several pore-forming voltage-gated sodium channel α subunits (NaV) in a subset of patients with SFN, but the auxiliary sodium channel ß subunits have been less implicated in the development of the disease. ß subunits modulate NaV trafficking and gating, and several mutations have been linked to epilepsy and cardiac dysfunction. Recently, we provided the first evidence for the contribution of a mutation in the ß2 subunit to pain in human painful diabetic neuropathy. Here, we provide the first evidence for the involvement of a sodium channel ß subunit mutation in the pathogenesis of SFN with no other known causes. We show, through current-clamp analysis, that the newly identified Y69H variant of the ß2 subunit induces neuronal hyperexcitability in dorsal root ganglion neurons, lowering the threshold for action potential firing and allowing for increased repetitive action potential spiking. Underlying the hyperexcitability induced by the ß2-Y69H variant, we demonstrate an upregulation in tetrodotoxin-sensitive, but not tetrodotoxin-resistant sodium currents. This provides the first evidence for the involvement of ß2 subunits in SFN and strengthens the link between sodium channel ß subunits and the development of neuropathic pain in humans.NEW & NOTEWORTHY Small fiber neuropathy (SFN) often has no discernible cause, although mutations in the voltage-gated sodium channel α subunits have been implicated in some cases. We identify a patient suffering from SFN with a mutation in the auxiliary ß2 subunit and no other discernible causes for SFN. Functional assessment confirms this mutation renders dorsal root ganglion neurons hyperexcitable and upregulates tetrodotoxin-sensitive sodium currents. This study strengthens a newly emerging link between sodium channel ß2 subunit mutations and human pain disorders.


Subject(s)
Gain of Function Mutation , Small Fiber Neuropathy/genetics , Voltage-Gated Sodium Channel beta-2 Subunit/genetics , Action Potentials , Animals , Cells, Cultured , Ganglia, Spinal/cytology , HEK293 Cells , Humans , Mutation, Missense , Neurons/metabolism , Neurons/physiology , Rats , Rats, Sprague-Dawley , Small Fiber Neuropathy/metabolism , Voltage-Gated Sodium Channel beta-2 Subunit/metabolism
2.
J Cell Sci ; 134(6)2021 03 26.
Article in English | MEDLINE | ID: mdl-33602743

ABSTRACT

The voltage-gated sodium channel is critical for cardiomyocyte function. It consists of a protein complex comprising a pore-forming α subunit and associated ß subunits. In polarized Madin-Darby canine kidney cells, we show evidence by acyl-biotin exchange that ß2 is S-acylated at Cys-182. Interestingly, we found that palmitoylation increases ß2 association with detergent-resistant membranes. ß2 localizes exclusively to the apical surface. However, depletion of plasma membrane cholesterol, or blocking intracellular cholesterol transport, caused mislocalization of ß2, as well as of the non-palmitoylable C182S mutant, to the basolateral domain. Apical ß2 did not undergo endocytosis and displayed limited diffusion within the plane of the membrane; such behavior suggests that, at least in part, it is cytoskeleton anchored. Upon acute cholesterol depletion, its mobility was greatly reduced, and a slight reduction was also measured as a result of lack of palmitoylation, supporting ß2 association with cholesterol-rich lipid rafts. Indeed, lipid raft labeling confirmed a partial overlap with apical ß2. Although ß2 palmitoylation was not required to promote surface localization of the α subunit, our data suggest that it is likely implicated in lipid raft association and the polarized localization of ß2.


Subject(s)
Lipoylation , Voltage-Gated Sodium Channel beta-2 Subunit , Animals , Cell Membrane/metabolism , Dogs , Madin Darby Canine Kidney Cells , Membrane Microdomains/metabolism , Voltage-Gated Sodium Channel beta-2 Subunit/metabolism
3.
Int J Mol Med ; 45(4): 1091-1102, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32124967

ABSTRACT

Our previous study demonstrated that the expression of sodium channel voltage­gated beta 2 (SCN2B) increased with aging in senescence­accelerated mouse prone 8 (SAMP8) mice, and was identified to be associated with a decline in learning and memory, while the underlying mechanism is unclear. In the present study, multiple differentially expressed miRNAs, which may be involved in the process of aging by regulating target genes, were identified in the prefrontal cortex and hippocampus of SAMP8 mice though miRNA microarray analysis. Using bioinformatics prediction, SCN2B was identified to be one of the potential target genes of miR­449a, which was downregulated in the hippocampus. Previous studies demonstrated that miR­449a is involved in the occurrence and progression of aging by regulating a variety of target genes. Therefore, it was hypothesized that miR­449a may be involved in the process of brain aging by targeting SCN2B. To verify this hypothesis, the following experiments were conducted: A reverse transcription­quantitative polymerase chain reaction assay revealed that the expression level of miR­449a was significantly decreased in the prefrontal cortex and hippocampus of 12­month old SAMP8 mice; a dual­luciferase reporter assay verified that miR­449a regulated SCN2B expression by binding to the 3'­UTR 'seed region'; an anti­Ago co­immunoprecipitation combined with Affymetrix microarray analyses demonstrated that the target mRNA highly enriched with Ago­miRNPs was confirmed to be SCN2B. Finally, overexpression of miR­449a or inhibition of SCN2B promoted the extension of hippocampal neurons in vitro. The results of the present study suggested that miR­449a was downregulated in the prefrontal cortex and hippocampus of SAMP8 mice and may regulate the process of brain aging by targeting SCN2B.


Subject(s)
Aging/metabolism , Brain/metabolism , Gene Expression Regulation , MicroRNAs/metabolism , Voltage-Gated Sodium Channel beta-2 Subunit/biosynthesis , Aging/genetics , Animals , Male , Mice , Mice, Transgenic , MicroRNAs/genetics , Voltage-Gated Sodium Channel beta-2 Subunit/genetics
4.
Biomolecules ; 9(10)2019 10 13.
Article in English | MEDLINE | ID: mdl-31614896

ABSTRACT

The voltage-gated sodium channel is vital for cardiomyocyte function, and consists of a protein complex containing a pore-forming α subunit and two associated ß subunits. A fundamental, yet unsolved, question is to define the precise function of ß subunits. While their location in vivo remains unclear, large evidence shows that they regulate localization of α and the biophysical properties of the channel. The current data support that one of these subunits, ß2, promotes cell surface expression of α. The main α isoform in an adult heart is NaV1.5, and mutations in SCN5A, the gene encoding NaV1.5, often lead to hereditary arrhythmias and sudden death. The association of ß2 with cardiac arrhythmias has also been described, which could be due to alterations in trafficking, anchoring, and localization of NaV1.5 at the cardiomyocyte surface. Here, we will discuss research dealing with mechanisms that regulate ß2 trafficking, and how ß2 could be pivotal for the correct localization of NaV1.5, which influences cellular excitability and electrical coupling of the heart. Moreover, ß2 may have yet to be discovered roles on cell adhesion and signaling, implying that diverse defects leading to human disease may arise due to ß2 mutations.


Subject(s)
Voltage-Gated Sodium Channel beta-2 Subunit/metabolism , Humans , Mutation , Protein Transport , Voltage-Gated Sodium Channel beta-2 Subunit/genetics
5.
J Biol Chem ; 294(44): 16123-16140, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31511323

ABSTRACT

The voltage-gated sodium channel is critical for cardiomyocyte function and consists of a protein complex comprising a pore-forming α subunit and two associated ß subunits. It has been shown previously that the associated ß2 subunits promote cell surface expression of the α subunit. The major α isoform in the adult human heart is NaV1.5, and germline mutations in the NaV1.5-encoding gene, sodium voltage-gated channel α subunit 5 (SCN5A), often cause inherited arrhythmias. Here, we investigated the mechanisms that regulate ß2 trafficking and how they may determine proper NaV1.5 cell surface localization. Using heterologous expression in polarized Madin-Darby canine kidney cells, we show that ß2 is N-glycosylated in vivo and in vitro at residues 42, 66, and 74, becoming sialylated only at Asn-42. We found that fully nonglycosylated ß2 was mostly retained in the endoplasmic reticulum, indicating that N-linked glycosylation is required for efficient ß2 trafficking to the apical plasma membrane. The nonglycosylated variant reached the cell surface by bypassing the Golgi compartment at a rate of only approximately one-third of that of WT ß2. YFP-tagged, nonglycosylated ß2 displayed mobility kinetics in the plane of the membrane similar to that of WT ß2. However, it was defective in promoting surface localization of NaV1.5. Interestingly, ß2 with a single intact glycosylation site was as effective as the WT in promoting NaV1.5 surface localization. In conclusion, our results indicate that N-linked glycosylation of ß2 is required for surface localization of NaV1.5, a property that is often defective in inherited cardiac arrhythmias.


Subject(s)
NAV1.5 Voltage-Gated Sodium Channel/metabolism , Voltage-Gated Sodium Channel beta-2 Subunit/metabolism , Animals , Cell Membrane/metabolism , Dogs , Glycosylation , Madin Darby Canine Kidney Cells , Membrane Potentials/physiology , Mutation , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , Protein Transport/physiology , Voltage-Gated Sodium Channel beta-2 Subunit/physiology
6.
Neurosci Lett ; 708: 134365, 2019 08 24.
Article in English | MEDLINE | ID: mdl-31278961

ABSTRACT

INTRODUCTION: Recent evidence shows that numerous microRNAs (miRNAs) regulate pain-related genes in chronic pain. The aim of the present study was to further explore the regulation of miRNAs and their effect on the expression of pain-associated target genes in experimental neuropathic pain. METHODS: Male Wistar rats underwent chronic constriction injury (CCI) of the sciatic nerve or Sham procedure. After assessment of mechanical allodynia, the ipsilateral dorsal root ganglia (DRG) were harvested. MiRNA expression levels were analysed with Agilent microRNA microarrays and real time quantitative PCR. An interaction between miRNAs and pain-relevant genes was confirmed by luciferase assays. Western Blot analysis and ELISA were performed to evaluate protein expression, respectively. RESULTS: Mechanical allodynia developed within 6 days after CCI. MiRNA-arrays revealed the differential expression of 49 miRNAs after 4 h, of 3 miRNAs after 1 d, of 26 miRNAs after 6 d and of 28 miRNAs after 12 d in the CCI group versus Sham. Time-dependent down regulation of miR-34a was verified by qPCR. Bioinformatic prediction revealed an interaction with several pain-relevant targets including voltage-gated sodium channel ß2 subunit (SCN2B) and vesicle-associated membrane protein 2 (VAMP-2), both of which were subsequently confirmed by luciferase assay. VAMP-2 expression was statistically significantly increased 12 d after CCI. A non-significant upregulation of SCN2B in the DRG after CCI was confirmed by ELISA. DISCUSSION: Peripheral mononeuropathic pain in rats was associated with distinct alterations of miRNA expression in the ipsilateral DRG. Notably, miR-34a was time-dependently down regulated. We validated SCN2B and VAMP-2 as new targets of miR-34a. While SCN2B expression was only marginally altered, VAMP-2 expression was increased. The present study underlines that the induction and maintenance of neuropathic pain is accompanied by expression changes of miRNAs in the peripheral nervous system, adding several previously unreported miRNAs, including miR-34a.


Subject(s)
Ganglia, Spinal/metabolism , MicroRNAs/metabolism , Neuralgia/metabolism , Sciatic Nerve/injuries , Animals , Chronic Disease , Constriction , Hyperalgesia/physiopathology , Male , Neuralgia/physiopathology , Rats, Wistar , Time Factors , Vesicle-Associated Membrane Protein 2/genetics , Vesicle-Associated Membrane Protein 2/metabolism , Voltage-Gated Sodium Channel beta-2 Subunit/genetics , Voltage-Gated Sodium Channel beta-2 Subunit/metabolism
7.
Mol Med Rep ; 20(2): 2002-2011, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31257483

ABSTRACT

Voltage­gated sodium channel ß2 (Navß2), as an unconventional substrate of ß­site amyloid precursor protein cleaving enzyme 1, is involved in regulating the neuronal surface expression of sodium channels. A previous study demonstrated that knockdown of Navß2 protected neurons and induced spatial cognition improvement by partially reducing pathological amyloidogenic processing of amyloid precursor protein (APP) in aged APP/presenilin 1 (PS1) transgenic mice. The present study aimed to investigate whether Navß2 knockdown altered APP metabolism via regulation of the Aß­degrading enzyme neprilysin (NEP). APPswe/PS1ΔE9 mice (APP/PS1 transgenic mice with a C57BL/6J genetic background) carrying a Navß2­knockdown mutation (APP/PS1/Navß2­kd) or without Navß2 knockdown (APP/PS1) were used for cell culture and further analysis. The present results demonstrated that in APP/PS1 mouse­derived neurons, Navß2 knockdown partially reversed the reduction in pathological APP cleavage, and the recovery of neurite extension and neuron area. Additionally, Navß2 knockdown increased NEP activity and levels, and the levels of intracellular domain fragment binding to the NEP promoter. The present findings suggested that knockdown of Navß2 reversed the APP/PS1 mutation­induced deficiency in amyloid ß degradation by regulating NEP.


Subject(s)
Neurons/metabolism , Neuroprotection/genetics , Presenilin-1/genetics , Voltage-Gated Sodium Channel beta-2 Subunit/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Humans , Mice , Mice, Transgenic , Mutation , Neprilysin/genetics , Neurons/pathology , Promoter Regions, Genetic/genetics
8.
Medicine (Baltimore) ; 98(13): e14974, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30921204

ABSTRACT

Previous research identified SCN1B variants in some cases of Dravet syndrome (DS). We investigated whether SCN1B and SCN2B variants are commonly happened in DS patients without SCN1A variants. A total of 22 DS patients without SCN1A variants and 100 healthy controls were enrolled in this genetic study. DNA from DS patients was sequenced by Sanger method in whole exons of SCN1B and SCN2B genes. We identified two exon variants (c.351C>T, p.G117G and c.467C>T, p.T156M), which were present both in 1000 egenomes database and in healthy controls with a frequency of 0.54% and 4%, 0.06% and 0%, respectively. Additionally, eight intron or 3 prime UTR variants showing benign clinical significance have also been identified. Our results suggest that variants of SCN1B and SCN2B may not be common causes of DS according to our data. Further large sample-size cohort studies are needed to confirm our conclusion.


Subject(s)
Epilepsies, Myoclonic/genetics , NAV1.1 Voltage-Gated Sodium Channel/genetics , Voltage-Gated Sodium Channel beta-1 Subunit/genetics , Voltage-Gated Sodium Channel beta-2 Subunit/genetics , Adolescent , Child , Child, Preschool , Female , Humans , Male , Mutation , Young Adult
9.
Hum Mutat ; 40(6): 749-764, 2019 06.
Article in English | MEDLINE | ID: mdl-30821013

ABSTRACT

Brugada syndrome (BrS) is an inherited arrhythmogenic disease associated with sudden cardiac death. The main gene is SCN5A. Additional variants in 42 other genes have been reported as deleterious, although these variants have not yet received comprehensive pathogenic analysis. Our aim was to clarify the role of all currently reported variants in minor genes associated with BrS. We performed a comprehensive analysis according to the American College of Medical Genetics and Genomics guidelines of published clinical and basic data on all genes (other than SCN5A) related to BrS. Our results identified 133 rare variants potentially associated with BrS. After applying current recommendations, only six variants (4.51%) show a conclusive pathogenic role. All definitively pathogenic variants were located in four genes encoding sodium channels or related proteins: SLMAP, SEMA3A, SCNN1A, and SCN2B. In total, 33.83% of variants in 19 additional genes were potentially pathogenic. Beyond SCN5A, we conclude definitive pathogenic variants associated with BrS in four minor genes. The current list of genes associated with BrS, therefore, should include SCN5A, SLMAP, SEMA3A, SCNN1A, and SCN2B. Comprehensive genetic interpretation and careful clinical translation should be done for all variants currently classified as potentially deleterious for BrS.


Subject(s)
Brugada Syndrome/genetics , Computational Biology/methods , Gene Regulatory Networks , Mutation , Epithelial Sodium Channels/genetics , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Male , Membrane Proteins/genetics , NAV1.5 Voltage-Gated Sodium Channel/genetics , Semaphorin-3A/genetics , Voltage-Gated Sodium Channel beta-2 Subunit/genetics
10.
Science ; 363(6433): 1309-1313, 2019 03 22.
Article in English | MEDLINE | ID: mdl-30765605

ABSTRACT

The voltage-gated sodium channel Nav1.2 is responsible for the initiation and propagation of action potentials in the central nervous system. We report the cryo-electron microscopy structure of human Nav1.2 bound to a peptidic pore blocker, the µ-conotoxin KIIIA, in the presence of an auxiliary subunit, ß2, to an overall resolution of 3.0 angstroms. The immunoglobulin domain of ß2 interacts with the shoulder of the pore domain through a disulfide bond. The 16-residue KIIIA interacts with the extracellular segments in repeats I to III, placing Lys7 at the entrance to the selectivity filter. Many interacting residues are specific to Nav1.2, revealing a molecular basis for KIIIA specificity. The structure establishes a framework for the rational design of subtype-specific blockers for Nav channels.


Subject(s)
Conotoxins/chemistry , NAV1.2 Voltage-Gated Sodium Channel/chemistry , Voltage-Gated Sodium Channel Blockers/chemistry , Amino Acid Sequence , Cryoelectron Microscopy , HEK293 Cells , Humans , Protein Conformation , Voltage-Gated Sodium Channel beta-2 Subunit/chemistry
11.
Science ; 363(6433): 1303-1308, 2019 Mar 22.
Article in English | MEDLINE | ID: mdl-30765606

ABSTRACT

Voltage-gated sodium channel Nav1.7 represents a promising target for pain relief. Here we report the cryo-electron microscopy structures of the human Nav1.7-ß1-ß2 complex bound to two combinations of pore blockers and gating modifier toxins (GMTs), tetrodotoxin with protoxin-II and saxitoxin with huwentoxin-IV, both determined at overall resolutions of 3.2 angstroms. The two structures are nearly identical except for minor shifts of voltage-sensing domain II (VSDII), whose S3-S4 linker accommodates the two GMTs in a similar manner. One additional protoxin-II sits on top of the S3-S4 linker in VSDIV The structures may represent an inactivated state with all four VSDs "up" and the intracellular gate closed. The structures illuminate the path toward mechanistic understanding of the function and disease of Nav1.7 and establish the foundation for structure-aided development of analgesics.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel/chemistry , Peptides/chemistry , Saxitoxin/chemistry , Spider Venoms/chemistry , Tetrodotoxin/chemistry , Voltage-Gated Sodium Channel Blockers/chemistry , Voltage-Gated Sodium Channel beta-1 Subunit/chemistry , Voltage-Gated Sodium Channel beta-2 Subunit/chemistry , Amino Acid Sequence , Animals , Binding Sites , Cryoelectron Microscopy , HEK293 Cells , Humans , Protein Conformation
12.
J Neurosci ; 37(39): 9519-9533, 2017 09 27.
Article in English | MEDLINE | ID: mdl-28871036

ABSTRACT

Neurotransmitter release depends on voltage-gated Na+ channels (Navs) to propagate an action potential (AP) successfully from the axon hillock to a synaptic terminal. Unmyelinated sections of axon are very diverse structures encompassing branch points and numerous presynaptic terminals with undefined molecular partners of Na+ channels. Using optical recordings of Ca2+ and membrane voltage, we demonstrate here that Na+ channel ß2 subunits (Navß2s) are required to prevent AP propagation failures across the axonal arborization of cultured rat hippocampal neurons (mixed male and female). When Navß2 expression was reduced, we identified two specific phenotypes: (1) membrane excitability and AP-evoked Ca2+ entry were impaired at synapses and (2) AP propagation was severely compromised with >40% of axonal branches no longer responding to AP-stimulation. We went on to show that a great deal of electrical signaling heterogeneity exists in AP waveforms across the axonal arborization independent of axon morphology. Therefore, Navß2 is a critical regulator of axonal excitability and synaptic function in unmyelinated axons.SIGNIFICANCE STATEMENT Voltage-gated Ca2+ channels are fulcrums of neurotransmission that convert electrical inputs into chemical outputs in the form of vesicle fusion at synaptic terminals. However, the role of the electrical signal, the presynaptic action potential (AP), in modulating synaptic transmission is less clear. What is the fidelity of a propagating AP waveform in the axon and what molecules shape it throughout the axonal arborization? Our work identifies several new features of AP propagation in unmyelinated axons: (1) branches of a single axonal arborization have variable AP waveforms independent of morphology, (2) Na+ channel ß2 subunits modulate AP-evoked Ca2+-influx, and (3) ß2 subunits maintain successful AP propagation across the axonal arbor. These findings are relevant to understanding the flow of excitation in the brain.


Subject(s)
Action Potentials , Axons/metabolism , Voltage-Gated Sodium Channel beta-2 Subunit/metabolism , Animals , Axons/physiology , CA1 Region, Hippocampal/cytology , Calcium Signaling , Cell Line , Cells, Cultured , Female , Male , Membrane Potentials , Rats , Rats, Sprague-Dawley , Synaptic Potentials
13.
Biol Cell ; 109(7): 273-291, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28597987

ABSTRACT

BACKGROUND INFORMATION: Cardiac channelopathies arise by mutations in genes encoding ion channel subunits. One example is Brugada Syndrome (BrS), which causes arrhythmias and sudden death. BrS is often associated with mutations in SCN5A, encoding Nav 1.5, the α subunit of the major cardiac voltage-gated sodium channel. This channel forms a protein complex including one or two associated ß subunits as well as other proteins. RESULTS: We analysed regulation of Nav 1.5 localisation and trafficking by ß2, specifically, Nav 1.5 arrival to the cell surface. We used polarised Madin-Darby canine kidney (MDCK) cells and mouse atria-derived HL-1 cells, which retain phenotypic features of adult cardiomyocytes. In both, Nav 1.5 was found essentially intracellular, mainly in the endoplasmic reticulum, whereas ß2 localised to the plasma membrane, and was restricted to the apical surface in MDCK cells. A fraction of ß2 interacted with Nav 1.5, despite their limited overlap. Importantly, ß2 promoted Nav 1.5 localisation to the cell surface. Both ß2 WT and the BrS-associated mutation D211G (substitution of Asp for Gly) effectively reached the plasma membrane. Strikingly, however, ß2 D211G was defective in promoting Nav 1.5 surface localisation. CONCLUSIONS: Our data sustain that ß2 promotes surface localisation of Nav 1.5, which can be affected due to ß2 mutations associated with channelopathies. SIGNIFICANCE: Our findings add to the understanding of ß2 role in Nav 1.5 trafficking and localisation, which must influence cell excitability and electrical coupling in the heart. This study will contribute to knowledge on development of arrhythmias.


Subject(s)
Brugada Syndrome/pathology , Cell Membrane/metabolism , Mutation , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Voltage-Gated Sodium Channel beta-2 Subunit/metabolism , Animals , Brugada Syndrome/genetics , Brugada Syndrome/metabolism , Cells, Cultured , Dogs , Humans , Madin Darby Canine Kidney Cells , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , Phenotype , Protein Subunits , Protein Transport , Voltage-Gated Sodium Channel beta-1 Subunit/genetics , Voltage-Gated Sodium Channel beta-1 Subunit/metabolism , Voltage-Gated Sodium Channel beta-2 Subunit/genetics
14.
Article in English | MEDLINE | ID: mdl-27932425

ABSTRACT

BACKGROUND: Mutations in SCN2B, encoding voltage-gated sodium channel ß2-subunits, are associated with human cardiac arrhythmias, including atrial fibrillation and Brugada syndrome. Because of this, we propose that ß2-subunits play critical roles in the establishment or maintenance of normal cardiac electric activity in vivo. METHODS AND RESULTS: To understand the pathophysiological roles of ß2 in the heart, we investigated the cardiac phenotype of Scn2b null mice. We observed reduced sodium and potassium current densities in ventricular myocytes, as well as conduction slowing in the right ventricular outflow tract region. Functional reentry, resulting from the interplay between slowed conduction, prolonged repolarization, and increased incidence of premature ventricular complexes, was found to underlie the mechanism of spontaneous polymorphic ventricular tachycardia. Scn5a transcript levels were similar in Scn2b null and wild-type ventricles, as were levels of Nav1.5 protein, suggesting that similar to the previous work in neurons, the major function of ß2-subunits in the ventricle is to chaperone voltage-gated sodium channel α-subunits to the plasma membrane. Interestingly, Scn2b deletion resulted in region-specific effects in the heart. Scn2b null atria had normal levels of sodium current density compared with wild type. Scn2b null hearts were more susceptible to atrial fibrillation, had increased levels of fibrosis, and higher repolarization dispersion than wild-type littermates. CONCLUSIONS: Genetic deletion of Scn2b in mice results in ventricular and atrial arrhythmias, consistent with reported SCN2B mutations in human patients.


Subject(s)
Atrial Fibrillation/genetics , Heart Conduction System/physiopathology , NAV1.5 Voltage-Gated Sodium Channel/genetics , Potassium Channels/genetics , Tachycardia, Ventricular/genetics , Voltage-Gated Sodium Channel beta-2 Subunit/genetics , Action Potentials , Animals , Atrial Fibrillation/physiopathology , Blotting, Western , Cells, Cultured , Gene Deletion , Genetic Predisposition to Disease , Mice , Monocytes , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Tachycardia, Ventricular/physiopathology
15.
Cell Mol Neurobiol ; 36(8): 1399-1408, 2016 Nov.
Article in English | MEDLINE | ID: mdl-26852328

ABSTRACT

The beta-2 subunit of the mammalian brain voltage-gated sodium channel (SCN2B) was examined in the rat trigeminal ganglion (TG) and trigeminal sensory nuclei. In the TG, 42.6 % of sensory neurons were immunoreactive (IR) for SCN2B. These neurons had various cell body sizes. In facial skins and oral mucosae, corpuscular nerve endings contained SCN2B-immunoreactivity. SCN2B-IR nerve fibers formed nerve plexuses beneath taste buds in the tongue and incisive papilla. However, SCN2B-IR free nerve endings were rare in cutaneous and mucosal epithelia. Tooth pulps, muscle spindles and major salivary glands were also innervated by SCN2B-IR nerve fibers. A double immunofluorescence method revealed that about 40 % of SCN2B-IR neurons exhibited calcitonin gene-related peptide (CGRP)-immunoreactivity. However, distributions of SCN2B- and CGRP-IR nerve fibers were mostly different in facial, oral and cranial structures. By retrograde tracing method, 60.4 and 85.3 % of TG neurons innervating the facial skin and tooth pulp, respectively, showed SCN2B-immunoreactivity. CGRP-immunoreactivity was co-localized by about 40 % of SCN2B-IR cutaneous and tooth pulp TG neurons. In trigeminal sensory nuclei of the brainstem, SCN2B-IR neuronal cell bodies were common in deep laminae of the subnucleus caudalis, and the subnuclei interpolaris and oralis. In the mesencephalic trigeminal tract nucleus, primary sensory neurons also exhibited SCN2B-immunoreactivity. In other regions of trigeminal sensory nuclei, SCN2B-IR cells were very infrequent. SCN2B-IR neuropil was detected in deep laminae of the subnucleus caudalis as well as in the subnuclei interpolaris, oralis and principalis. These findings suggest that SCN2B is expressed by various types of sensory neurons in the TG. There appears to be SCN2B-containing pathway in the TG and trigeminal sensory nuclei.


Subject(s)
Trigeminal Ganglion/metabolism , Trigeminal Nuclei/metabolism , Voltage-Gated Sodium Channel beta-2 Subunit/biosynthesis , Animals , Calcitonin Gene-Related Peptide/metabolism , Face/innervation , Male , Mouth/innervation , Mouth/metabolism , Rats , Rats, Wistar , Sensory Receptor Cells/metabolism , Skull/innervation , Skull/metabolism
16.
Mol Neurobiol ; 53(2): 955-967, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25575679

ABSTRACT

The role of sodium channel voltage-gated beta 2 (SCN2B) in brain aging is largely unknown. The present study was therefore designed to determine the role of SCN2B in brain aging by using the senescence-accelerated mice prone 8 (SAMP8), a brain senescence-accelerated animal model, together with the SCN2B transgenic mice. The results showed that SAMP8 exhibited impaired learning and memory functions, assessed by the Morris water maze test, as early as 8 months of age. The messenger RNA (mRNA) and protein expressions of SCN2B were also upregulated in the prefrontal cortex at this age. Treatment with traditional Chinese anti-aging medicine Xueshuangtong (Panax notoginseng saponins, PNS) significantly reversed the SCN2B expressions in the prefrontal cortex, resulting in improved learning and memory. Moreover, SCN2B knockdown transgenic mice were generated and bred to determine the roles of SCN2B in brain senescence. A reduction in the SCN2B level by 60.68% resulted in improvement in the hippocampus-dependent spatial recognition memory and long-term potential (LTP) slope of field excitatory postsynaptic potential (fEPSP), followed by an upregulation of COX5A mRNA levels and downregulation of fibroblast growth factor-2 (FGF-2) mRNA expression. Together, the present findings indicated that SCN2B could play an important role in the aging-related cognitive deterioration, which is associated with the regulations of COX5A and FGF-2. These findings could provide the potential strategy of candidate target to develop antisenescence drugs for the treatment of brain aging.


Subject(s)
Aging/metabolism , Brain/metabolism , Electron Transport Complex IV/metabolism , Fibroblast Growth Factor 2/metabolism , Neuronal Plasticity , Voltage-Gated Sodium Channel beta-2 Subunit/metabolism , Animals , Gene Expression Regulation , Gene Knockdown Techniques , Male , Maze Learning , Memory , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
17.
Mol Pain ; 11: 60, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26408173

ABSTRACT

BACKGROUND: Increased electrical activity in peripheral sensory neurons including dorsal root ganglia (DRG) and trigeminal ganglia neurons is an important mechanism underlying pain. Voltage gated sodium channels (VGSC) contribute to the excitability of sensory neurons and are essential for the upstroke of action potentials. A unique type of VGSC current, resurgent current (INaR), generates an inward current at repolarizing voltages through an alternate mechanism of inactivation referred to as open-channel block. INaRs are proposed to enable high frequency firing and increased INaRs in sensory neurons are associated with pain pathologies. While Nav1.6 has been identified as the main carrier of fast INaR, our understanding of the mechanisms that contribute to INaR generation is limited. Specifically, the open-channel blocker in sensory neurons has not been identified. Previous studies suggest Navß4 subunit mediates INaR in central nervous system neurons. The goal of this study was to determine whether Navß4 regulates INaR in DRG sensory neurons. RESULTS: Our immunocytochemistry studies show that Navß4 expression is highly correlated with Nav1.6 expression predominantly in medium-large diameter rat DRG neurons. Navß4 knockdown decreased endogenous fast INaR in medium-large diameter neurons as measured with whole-cell voltage clamp. Using a reduced expression system in DRG neurons, we isolated recombinant human Nav1.6 sodium currents in rat DRG neurons and found that overexpression of Navß4 enhanced Nav1.6 INaR generation. By contrast neither overexpression of Navß2 nor overexpression of a Navß4-mutant, predicted to be an inactive form of Navß4, enhanced Nav1.6 INaR generation. DRG neurons transfected with wild-type Navß4 exhibited increased excitability with increases in both spontaneous activity and evoked activity. Thus, Navß4 overexpression enhanced INaR and excitability, whereas knockdown or expression of mutant Navß4 decreased INaR generation. CONCLUSION: INaRs are associated with inherited and acquired pain disorders. However, our ability to selectively target and study this current has been hindered due to limited understanding of how it is generated in sensory neurons. This study identified Navß4 as an important regulator of INaR and excitability in sensory neurons. As such, Navß4 is a potential target for the manipulation of pain sensations.


Subject(s)
Ion Channel Gating , Sensory Receptor Cells/metabolism , Voltage-Gated Sodium Channel beta-4 Subunit/metabolism , Amino Acid Sequence , Animals , Ganglia, Spinal/metabolism , Gene Knockdown Techniques , Humans , Male , Models, Biological , Molecular Sequence Data , NAV1.6 Voltage-Gated Sodium Channel/metabolism , Rats, Sprague-Dawley , Voltage-Gated Sodium Channel beta-2 Subunit , Voltage-Gated Sodium Channel beta-4 Subunit/chemistry
18.
Mol Neurodegener ; 10: 36, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26259688

ABSTRACT

BACKGROUND: Previous studies have demonstrated that the trafficking defects of Nav1.1/Nav1.2 are involved in the dementia pathophysiology. However, the detailed mechanisms are not fully understood. Moreover, whether the impaired miRNAs regulation linked to dementia is a key player in sodium channel trafficking disturbance remains unclear. The cognitive impairment induced by chronic cerebral ischemia through chronic brain hypoperfusion (CBH) is likely reason to precede dementia. Therefore, our goal in the present study was to examine the role of microRNA-9 (miR-9) in regulating Nav1.1/Nav1.2 trafficking under CBH generated by bilateral common carotid artery occlusion (2VO). RESULTS: The impairment of Nav1.1/Nav1.2 trafficking and decreased expression of Navß2 were found in the hippocampi and cortices of rats following CBH generated by bilateral 2VO. MiR-9 was increased in both the hippocampi and cortices of rats following CBH by qRT-PCR. Intriguingly, miR-9 suppressed, while AMO-miR-9 enhanced, the trafficking of Nav1.1/Nav1.2 from cytoplasm to cell membrane. Further study showed that overexpression of miR-9 inhibited the Navß2 expression by targeting on its coding sequence (CDS) domain by dual luciferase assay. However, binding-site mutation or miR-masks failed to influence Navß2 expression as well as Nav1.1/Nav1.2 trafficking process, indicating that Navß2 is a potential target for miR-9. Lentivirus-mediated miR-9 overexpression also inhibited Navß2 expression and elicited translocation deficits to cell membrane of Nav1.1/Nav1.2 in rats, whereas injection of lentivirus-mediated miR-9 knockdown could reverse the impaired trafficking of Nav1.1/Nav1.2 triggered by 2VO. CONCLUSIONS: We conclude that miR-9 may play a key role in regulating the process of Nav1.1/Nav1.2 trafficking via targeting on Navß2 protein in 2VO rats at post-transcriptional level, and inhibition of miR-9 may be a potentially valuable approach to prevent Nav1.1/Nav1.2 trafficking disturbance induced by CBH.


Subject(s)
Brain Ischemia/metabolism , MicroRNAs/pharmacology , NAV1.1 Voltage-Gated Sodium Channel/metabolism , NAV1.2 Voltage-Gated Sodium Channel/metabolism , Nerve Tissue Proteins/metabolism , Protein Transport/genetics , Voltage-Gated Sodium Channel Blockers , Animals , Brain Ischemia/genetics , Carotid Artery, Common , Cerebral Cortex/metabolism , Chronic Disease , Gene Expression Regulation , Gene Knockdown Techniques , Genetic Vectors/pharmacology , Hippocampus/metabolism , Lentivirus/genetics , Ligation , Male , MicroRNAs/genetics , NAV1.1 Voltage-Gated Sodium Channel/genetics , NAV1.2 Voltage-Gated Sodium Channel/genetics , Nerve Tissue Proteins/genetics , Oligonucleotides, Antisense/pharmacology , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Sodium/metabolism , Voltage-Gated Sodium Channel beta-2 Subunit/biosynthesis , Voltage-Gated Sodium Channel beta-2 Subunit/genetics
19.
PLoS One ; 10(7): e0132888, 2015.
Article in English | MEDLINE | ID: mdl-26173111

ABSTRACT

BACKGROUND: Brugada syndrome (BrS) is a rare genetic cardiac arrhythmia that can lead to sudden cardiac death in patients with a structurally normal heart. Genetic variations in SCN5A can be identified in approximately 20-25% of BrS cases. The aim of our work was to determine the spectrum and prevalence of genetic variations in a Spanish cohort diagnosed with BrS. METHODOLOGY/PRINCIPAL FINDINGS: We directly sequenced fourteen genes reported to be associated with BrS in 55 unrelated patients clinically diagnosed. Our genetic screening allowed the identification of 61 genetic variants. Of them, 20 potentially pathogenic variations were found in 18 of the 55 patients (32.7% of the patients, 83.3% males). Nineteen of them were located in SCN5A, and had either been previously reported as pathogenic variations or had a potentially pathogenic effect. Regarding the sequencing of the minority genes, we discovered a potentially pathogenic variation in SCN2B that was described to alter sodium current, and one nonsense variant of unknown significance in RANGRF. In addition, we also identified 40 single nucleotide variations which were either synonymous variants (four of them had not been reported yet) or common genetic variants. We next performed MLPA analysis of SCN5A for the 37 patients without an identified genetic variation, and no major rearrangements were detected. Additionally, we show that being at the 30-50 years range or exhibiting symptoms are factors for an increased potentially pathogenic variation discovery yield. CONCLUSIONS: In summary, the present study is the first comprehensive genetic evaluation of 14 BrS-susceptibility genes and MLPA of SCN5A in a Spanish BrS cohort. The mean pathogenic variation discovery yield is higher than that described for other European BrS cohorts (32.7% vs 20-25%, respectively), and is even higher for patients in the 30-50 years age range.


Subject(s)
Brugada Syndrome/genetics , Genetic Predisposition to Disease/genetics , Hispanic or Latino/genetics , Polymorphism, Single Nucleotide/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Gene Rearrangement/genetics , Genetic Testing/methods , Humans , Male , Middle Aged , NAV1.5 Voltage-Gated Sodium Channel/genetics , Voltage-Gated Sodium Channel beta-2 Subunit/genetics , Young Adult
20.
PLoS One ; 9(6): e98408, 2014.
Article in English | MEDLINE | ID: mdl-24892658

ABSTRACT

Prostate cancer (PCa) is believed to metastasize through the blood/lymphatics systems; however, PCa may utilize the extensive innervation of the prostate for glandular egress. The interaction of PCa and its nerve fibers is observed in 80% of PCa and is termed perineural invasion (PNI). PCa cells have been observed traveling through the endoneurium of nerves, although the underlying mechanisms have not been elucidated. Voltage sensitive sodium channels (VSSC) are multimeric transmembrane protein complexes comprised of a pore-forming α subunit and one or two auxiliary beta (ß) subunits with inherent cell adhesion molecule (CAM) functions. The beta-2 isoform (gene SCN2B) interacts with several neural CAMs, while interacting putatively with other prominent neural CAMs. Furthermore, beta-2 exhibits elevated mRNA and protein levels in highly metastatic and castrate-resistant PCa. When overexpressed in weakly aggressive LNCaP cells (2BECFP), beta-2 alters LNCaP cell morphology and enhances LNCaP cell metastasis associated behavior in vitro. We hypothesize that PCa cells use beta-2 as a CAM during PNI and subsequent PCa metastasis. The objective of this study was to determine the effect of beta-2 expression on PCa cell neurotropic metastasis associated behavior. We overexpressed beta-2 as a fusion protein with enhanced cyan fluorescence protein (ECFP) in weakly aggressive LNCaP cells and observed neurotropic effects utilizing our novel ex vivo organotypic spinal cord co-culture model, and performed functional assays with neural matrices and atomic force microscopy. With increased beta-2 expression, PCa cells display a trend of enhanced association with nerve axons. On laminin, a neural CAM, overexpression of beta-2 enhances PCa cell migration, invasion, and growth. 2BECFP cells exhibit marked binding affinity to laminin relative to LNECFP controls, and recombinant beta-2 ectodomain elicits more binding events to laminin than BSA control. Functional overexpression of VSSC beta subunits in PCa may mediate PCa metastatic behavior through association with neural matrices.


Subject(s)
Biophysical Phenomena , Neurons/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Voltage-Gated Sodium Channel beta-2 Subunit/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Axons/drug effects , Axons/metabolism , Axons/pathology , Bacterial Proteins/metabolism , Biophysical Phenomena/drug effects , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Elastic Modulus/drug effects , Humans , Laminin/pharmacology , Luminescent Proteins/metabolism , Male , Mice, Transgenic , Microscopy, Atomic Force , Molecular Sequence Data , Neoplasm Invasiveness , Neurons/drug effects , Protein Binding/drug effects , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Spinal Cord/pathology , Voltage-Gated Sodium Channel beta-2 Subunit/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...