Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
Nat Rev Cardiol ; 14(9): 521-535, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28470179

ABSTRACT

Cardiac arrhythmias confer a considerable burden of morbidity and mortality in industrialized countries. Although coronary artery disease and heart failure are the prevalent causes of cardiac arrest, in 5-15% of patients, structural abnormalities at autopsy are absent. In a proportion of these patients, mutations in genes encoding cardiac ion channels are documented (inherited channelopathies), but, to date, the molecular autopsy is negative in nearly 70% of patients. Emerging evidence indicates that autoimmunity is involved in the pathogenesis of cardiac arrhythmias. In particular, several arrhythmogenic autoantibodies targeting specific calcium, potassium, or sodium channels in the heart have been identified. Experimental and clinical studies demonstrate that these autoantibodies can promote conduction disturbances and life-threatening tachyarrhythmias by inducing substantial electrophysiological changes. In this Review, we propose the term 'autoimmune cardiac channelopathies' to define this novel pathogenic mechanism of cardiac arrhythmias, which could be more frequent and clinically relevant than previously appreciated. Indeed, pathogenic autoantibodies against ion channels are detectable not only in patients with manifest autoimmune disease, but also in apparently healthy individuals, which suggests a causal role in some cases of unexplained arrhythmias and cardiac arrest. Considering this possibility and performing specific testing in patients with 'idiopathic' rhythm disturbances could create novel treatment opportunities.


Subject(s)
Arrhythmias, Cardiac/etiology , Autoimmune Diseases/complications , Channelopathies/complications , Adult , Antibodies, Anti-Idiotypic/immunology , Arrhythmias, Cardiac/immunology , Arrhythmias, Cardiac/therapy , Autoantibodies/metabolism , Autoimmune Diseases/diagnosis , Autoimmune Diseases/therapy , Calcium Channels/immunology , Channelopathies/diagnosis , Channelopathies/therapy , Child , Humans , Ion Channels/immunology , Myocytes, Cardiac/immunology , Potassium Channels/immunology , Sodium Channels/immunology
2.
Channels (Austin) ; 10(4): 297-312, 2016 Jul 03.
Article in English | MEDLINE | ID: mdl-26889602

ABSTRACT

Intracellular Fibroblast Growth Factor 14 (iFGF14) and the other intracellular FGFs (iFGF11-13) regulate the properties and densities of voltage-gated neuronal and cardiac Na(+) (Nav) channels. Recent studies have demonstrated that the iFGFs can also regulate native voltage-gated Ca(2+) (Cav) channels. In the present study, a mass spectrometry (MS)-based proteomic approach was used to identify the components of native cerebellar iFGF14 complexes. Using an anti-iFGF14 antibody, native iFGF14 complexes were immunoprecipitated from wild type adult mouse cerebellum. Parallel control experiments were performed on cerebellar proteins isolated from mice (Fgf14(-/-)) harboring a targeted disruption of the Fgf14 locus. MS analyses of immunoprecipitated proteins demonstrated that the vast majority of proteins identified in native cerebellar iFGF14 complexes are Nav channel pore-forming (α) subunits or proteins previously reported to interact with Nav α subunits. In contrast, no Cav channel α or accessory subunits were revealed in cerebellar iFGF14 immunoprecipitates. Additional experiments were completed using an anti-PanNav antibody to immunoprecipitate Nav channel complexes from wild type and Fgf14(-/-) mouse cerebellum. Western blot and MS analyses revealed that the loss of iFGF14 does not measurably affect the protein composition or the relative abundance of Nav channel interacting proteins in native adult mouse cerebellar Nav channel complexes.


Subject(s)
Cerebellum/chemistry , Fibroblast Growth Factors/analysis , Proteomics/methods , Sodium Channels/analysis , Animals , Antibodies, Monoclonal/immunology , Cerebellum/cytology , Cerebellum/metabolism , Chromatography, High Pressure Liquid , Electrophoresis, Gel, Two-Dimensional , Female , Fibroblast Growth Factors/immunology , Immunoprecipitation , Male , Mass Spectrometry , Mice , Sodium Channels/immunology , Sodium Channels/metabolism
4.
Nat Immunol ; 13(9): 880-7, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22842345

ABSTRACT

The sustained entry of Ca(2+) into CD4(+)CD8(+) double-positive thymocytes is required for positive selection. Here we identified a voltage-gated Na(+) channel (VGSC) that was essential for positive selection of CD4(+) T cells. Pharmacological inhibition of VGSC activity inhibited the sustained Ca(2+) influx induced by positively selecting ligands and the in vitro positive selection of CD4(+) but not CD8(+) T cells. In vivo short hairpin RNA (shRNA)-mediated knockdown of the gene encoding a regulatory ß-subunit of a VGSC specifically inhibited the positive selection of CD4(+) T cells. Ectopic expression of VGSC in peripheral AND CD4(+) T cells bestowed the ability to respond to a positively selecting ligand, which directly demonstrated that VGSC expression was responsible for the enhanced sensitivity. Thus, active VGSCs in thymocytes provide a mechanism by which a weak positive selection signal can induce the sustained Ca(2+) signals required for CD4(+) T cell development.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Sodium Channels/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Flow Cytometry , Humans , Ion Channel Gating , Mice , Mice, Transgenic , NAV1.5 Voltage-Gated Sodium Channel , Reverse Transcriptase Polymerase Chain Reaction , Voltage-Gated Sodium Channel beta-4 Subunit
6.
Channels (Austin) ; 6(2): 103-10, 2012.
Article in English | MEDLINE | ID: mdl-22622154

ABSTRACT

Loss-of-function mutations in the pore-forming α subunit of the voltage-gated sodium channel 1.7 (Nav 1.7) cause congenital indifference to pain and anosmia. We used immunohistochemical techniques to study Nav 1.7 localization in the rat olfactory system in order to better understand its role in olfaction. We confirm that Nav 1.7 is expressed on olfactory sensory axons and report its presence on vomeronasal axons, indicating an important role for Nav 1.7 in transmission of pheromonal cues. Following neuroepithelial injury, Nav 1.7 was transiently expressed by cells of monocytic lineage. These findings support an emerging role for Nav 1.7 in immune function. This sodium channel may provide an important pharmacological target for treatment of inflammatory injury and inflammatory pain syndromes.


Subject(s)
Olfactory Mucosa/metabolism , Sodium Channels/metabolism , Animals , Axons/metabolism , CHO Cells , Cricetinae , Cricetulus , Humans , Immunohistochemistry , Male , Monocytes/immunology , Monocytes/metabolism , NAV1.7 Voltage-Gated Sodium Channel , Olfactory Bulb/cytology , Olfactory Mucosa/drug effects , Olfactory Mucosa/innervation , Rats , Rats, Sprague-Dawley , Smell/physiology , Sodium Channels/immunology , Sodium Channels/physiology , Vomeronasal Organ/cytology , Vomeronasal Organ/physiology
7.
Neuron ; 66(4): 508-22, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20510856

ABSTRACT

Na(x) is the sodium-level sensor of body fluids in the brain involved in sodium homeostasis. Na(x)-knockout mice do not stop ingesting salt even when dehydrated. Here we report a case with clinical features of essential hypernatremia without demonstrable hypothalamic structural lesions, who was diagnosed as a paraneoplastic neurologic disorder. The patient had autoantibodies directed against Na(x), along with a ganglioneuroma composed of Schwann-like cells robustly expressing Na(x). The removal of the tumor did not reduce the autoantibody levels or relieve the symptoms. Intravenous injection of the immunoglobulin fraction of the patient's serum into mice induced abnormalities in water/salt intake and diuresis, which led to hypernatremia. In the brains of these mice, cell death was observed along with focal deposits of complement C3 and inflammatory infiltrates in circumventricular organs where Na(x) is specifically expressed. Our findings thus provide new insights into the pathogenesis of hypernatremia relevant to the sodium-level-sensing mechanism in humans.


Subject(s)
Autoantibodies/physiology , Autoimmune Diseases/immunology , Brain/immunology , Hypernatremia/immunology , Sodium Channels/immunology , Animals , Autoantibodies/biosynthesis , Autoantibodies/blood , Autoimmune Diseases/metabolism , Brain/pathology , Brain/surgery , Child , Drinking/immunology , Female , Humans , Hypernatremia/surgery , Mice , Voltage-Gated Sodium Channels
8.
J Neurol Sci ; 292(1-2): 63-71, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20219211

ABSTRACT

Intravenous immunoglobulin (IVIg) infusions may provide clinical benefits in multifocal motor neuropathy (MMN) and chronic inflammatory demyelinating polyneuropathy (CIDP). The short delay in the clinical response to IVIg therapy is not consistent with a process of remyelination or axonal regeneration. We assessed whether or not the efficacy of IVIg infusions in MMN and CIDP could reflect changes in axonal membrane properties and nerve excitability. Ulnar motor nerve excitability was studied before and after three to five consecutive days of IVIg infusions (0.4 g/kg/day) in 10 patients with MMN, 10 patients with CIDP, and 10 neurological controls (CTRLs). Excitability recovery cycle, stimulus-response and strength-duration properties were investigated. The recovery cycle parameters (absolute and relative refractory period durations, refractoriness and supernormality) were similar in all groups and did not change after IVIg infusions. At baseline, patients with CIDP, but not with MMN, showed a reduced strength-duration time constant (chronaxie) and increased rheobase when compared to CTRLs. After IVIg infusions, strength-duration time constant remained stable in CTRLs, but decreased in patients with MMN or CIDP. Rheobase increased in the three groups after treatment. The decreased strength-duration time constant after IVIg infusions in patients with MMN or CIDP could reflect a reduction of persistent Na(+) current, able to limit intraaxonal Na(+) accumulation and then to produce neuroprotective effects. However, this could also reflect compensatory mechanisms that did not directly underlie the therapeutic effect. Whatever the underlying process, this result revealed that IVIgs were able to produce early nerve excitability changes.


Subject(s)
Demyelinating Diseases/therapy , Immunoglobulins, Intravenous/therapeutic use , Neural Conduction/immunology , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/therapy , Ulnar Nerve/immunology , Adult , Aged , Aged, 80 and over , Demyelinating Diseases/immunology , Demyelinating Diseases/physiopathology , Electromyography , Female , Hand Strength , Humans , Male , Middle Aged , Neurons, Afferent/immunology , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/immunology , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/physiopathology , Sodium Channels/immunology , Treatment Outcome , Ulnar Nerve/physiopathology
9.
Neuroscience ; 159(3): 1126-34, 2009 Mar 31.
Article in English | MEDLINE | ID: mdl-19356693

ABSTRACT

Acid-sensing ion channels (ASICs), the members of the epithelial sodium channel/degenerin (ENaC/DEG) superfamily, are proton-gated voltage-insensitive cation channels. Six ASIC subunits have been identified and characterized in the mammalian nervous system so far. Of these subunits, ASIC3 has been shown to be predominantly expressed in the peripheral nervous system of rodents and implicated in mechnosensation, chemosensation and pain perception. Little is known on ASIC3 in the brain. We thus employed reverse transcription-polymerase chain reaction (RT-PCR) and Western blot to examine the expression of ASIC3 in various rat brain regions, including hippocampus, amygdala, caudate putamen, prefrontal cortex, and hypothalamus. Specific attention was paid to the distribution of ASIC3 in the hypothalamus of rats by using immunohistochemistry. ASIC3 immunoreactivity showed a widespread pattern throughout the hypothalamus, with the highest density in paraventricular nucleus, supraoptic nucleus, suprachiasmatic nucleus, arcuate nucleus, dorsomedial nucleus, median preoptic nucleus, ventromedial preoptic nucleus, and dorsal tuberomammillary nucleus. This study may contribute to the understanding of ASIC3 functions in the CNS.


Subject(s)
Brain/metabolism , Hypothalamus/metabolism , Nerve Tissue Proteins/metabolism , Sodium Channels/metabolism , Acid Sensing Ion Channels , Animals , Antibodies , Blotting, Western , Ganglia, Spinal/metabolism , Immunohistochemistry , Male , Nerve Tissue Proteins/immunology , RNA, Messenger , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sodium Channels/immunology , Tubulin/metabolism
10.
J Clin Neurosci ; 16(6): 733-41, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19356935

ABSTRACT

Guillain-Barré syndrome (GBS) is an acute polyneuropathy consisting of different subtypes. Acute inflammatory demyelinating polyradiculoneuropathy, the classic demyelinating form of GBS, accounts for 90% of all GBS cases in the Western world. Acute motor axonal neuropathy (AMAN) and acute motor and sensory axonal neuropathy (AMSAN) are axonal forms of GBS that are more prevalent in Asia, South and Central America, often preceded by infection by Campylobacter jejuni. AMAN and AMSAN may be mediated by specific anti-ganglioside antibodies that inhibit transient sodium ion (Na+) channels. The efficacy of plasmapheresis and intravenous immunoglobulin has been established in large international randomised trials, with corticosteroids proven ineffective. Although axonal demyelination is an established pathophysiological process in GBS, the rapid improvement of clinical deficits with treatment is consistent with Na+ channel blockade by antibodies or other circulating factors, such as cytokines. This review provides an update on the epidemiology, clinical features, diagnosis, pathogenesis and treatment of GBS.


Subject(s)
Autoantibodies/immunology , Guillain-Barre Syndrome/physiopathology , Infections/complications , Nerve Fibers, Myelinated/immunology , Peripheral Nervous System/physiopathology , Sodium Channels/immunology , Guillain-Barre Syndrome/immunology , Guillain-Barre Syndrome/therapy , Humans , Immunotherapy/methods , Immunotherapy/trends , Infections/immunology , Nerve Fibers, Myelinated/pathology , Neural Conduction/immunology , Peripheral Nervous System/immunology , Ranvier's Nodes/immunology
11.
J Neurosci ; 28(47): 12510-22, 2008 Nov 19.
Article in English | MEDLINE | ID: mdl-19020043

ABSTRACT

Voltage-gated Na(+) channels initiate and propagate action potentials in excitable cells. Mammalian Na(+) channels are composed of one pore-forming alpha-subunit and two beta-subunits. SCN1B encodes the Na(+) channel beta1-subunit that modulates channel gating and voltage dependence, regulates channel cell surface expression, and functions as a cell adhesion molecule (CAM). We recently identified scn1ba, a zebrafish ortholog of SCN1B. Here we report that zebrafish express a second beta1-like paralog, scn1bb. In contrast to the restricted expression of scn1ba mRNA in excitable cells, we detected scn1bb transcripts and protein in several ectodermal derivatives including neurons, glia, the lateral line, peripheral sensory structures, and tissues derived from other germ layers such as the pronephros. As expected for beta1-subunits, elimination of Scn1bb protein in vivo by morpholino knock-down reduced Na(+) current amplitudes in Rohon-Beard neurons of zebrafish embryos, consistent with effects observed in heterologous systems. Further, after Scn1bb knock-down, zebrafish embryos displayed defects in Rohon-Beard mediated touch sensitivity, demonstrating the significance of Scn1bb modulation of Na(+) current to organismal behavior. In addition to effects associated with Na(+) current modulation, Scn1bb knockdown produced phenotypes consistent with CAM functions. In particular, morpholino knock-down led to abnormal development of ventrally projecting spinal neuron axons, defasciculation of the olfactory nerve, and increased hair cell number in the inner ear. We propose that, in addition to modulation of electrical excitability, Scn1bb plays critical developmental roles by functioning as a CAM in the zebrafish embryonic nervous system.


Subject(s)
Axons/physiology , Motor Neurons/cytology , Motor Neurons/physiology , Neuroglia/physiology , Sodium Channels/metabolism , Touch/physiology , Zebrafish Proteins/metabolism , Analysis of Variance , Animals , Animals, Genetically Modified , Antibodies, Monoclonal/pharmacology , Axons/drug effects , Body Patterning/genetics , Body Patterning/physiology , Central Nervous System/cytology , Central Nervous System/metabolism , Embryo, Nonmammalian , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Kidney/cytology , Kidney/embryology , Kidney/metabolism , Motor Neurons/classification , Motor Neurons/drug effects , Sequence Alignment/methods , Sodium Channels/immunology , Spinal Cord/cytology , Tubulin/metabolism , Voltage-Gated Sodium Channel beta-1 Subunit , Zebrafish , Zebrafish Proteins/immunology
12.
Pain ; 140(2): 292-304, 2008 Nov 30.
Article in English | MEDLINE | ID: mdl-18834667

ABSTRACT

The roles of ion channels in sensory neurons were examined in experimental models of muscle pain in the rat. Rats were injected with 50 microl of 4% carrageenan or subjected to an eccentric exercise (ECC) of the gastrocnemius muscle (GM). The Randall-Selitto and von Frey tests were performed on the calves to evaluate mechanical hyperalgesia of the muscle. The changes in expression of four genes and proteins of ion channels in dorsal root ganglia were examined using quantitative PCR and immunohistochemistry, respectively. Effects of antagonists to transient receptor potential (TRP) channels and acid sensing ion channels (ASICs) on the mechanical hyperalgesia induced by carrageenan injection or ECC were evaluated. The mechanical hyperalgesia was observed 6-24h after carrageenan injection and 1-3 days after ECC in the Randall-Selitto test. Infiltrations of the inflammatory cells in the GM were seen in carrageenan-injected animals but not in those subjected to ECC. Expressions of genes and proteins in sensory neurons showed no changes. Intramuscular injection of antagonists to TRPV1 showed an almost complete suppressive effect on ECC-induced muscle hyperalgesia but not a carrageenan-induced one. Antagonists to TRP channels and ASICs showed suppressive effects for both carrageenan- and ECC-induced muscle hyperalgesia. The carrageenan injection and ECC models are useful models of acute inflammatory pain and delayed onset muscle soreness (DOMS), respectively, and the time course and underlying etiology might be different. TRP channels and ASICs are closely related to the development of muscle mechanical hyperalgesia, and TRPV1 is involved in ECC-induced DOMS.


Subject(s)
Disease Models, Animal , Fibromyalgia/immunology , Ganglia, Spinal/immunology , Hyperalgesia/immunology , Myositis/immunology , Nerve Tissue Proteins/immunology , Sodium Channels/immunology , TRPC Cation Channels/immunology , Acid Sensing Ion Channels , Animals , Male , Rats , Rats, Sprague-Dawley , Touch
13.
J Neurosci Res ; 86(15): 3359-74, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18627035

ABSTRACT

An infecting strain VLA2/18 of Campylobacter jejuni was obtained from an individual with campylobacteriosis and used to prepare chicken sera by experimental infection to investigate the role of serum anti-ganglioside antibodies in Guillain-Barré syndrome. Both sera of the patient and chicken contained anti-ganglioside antibodies and anti-Lipid A (anti-Kdo2-Lipid A) antibodies directed against the lipid A portion of the bacterial lipooligosaccharide. The anti-Kdo2-Lipid A activities inhibited voltage-gated Na (Nav) channel of NSC-34 cells in culture. We hypothesized that anti-Kdo2-Lipid A antibody acts on the functional inhibition of Nav1.4. To test this possibility, a rabbit peptide antibody (anti-Nav1.4 pAb) against a 19-mer peptide (KELKDNHILNHVGLTDGPR) on the alpha subunit of Nav1.4 was produced. Anti-Nav1.4 pAb was cross-reactive to Kdo2-Lipid A. Anti-Kdo2-lipid A antibody activity in the chicken serum was tested for the Na(+) current inhibition in NSC-34 cells in combination with mu-Conotoxin and tetrodotoxin. Contrary to our expectations, the anti-Kdo2-Lipid A antibody activity was extended to Nav channels other than Nav1.4. By overlapping structural analysis, it was found that there might be multiple peptide epitopes containing certain dipeptides showing a structural similarity with v-Lipid A. Thus, our study suggests the possibility that there are multiple epitopic peptides on the extracellular domains of Nav1.1 to 1.9, and some of them may represent target sites for anti-Kdo2-Lipid A antibody, to induce neurophysiological changes in GBS by disrupting the normal function of the Nav channels.


Subject(s)
Autoantibodies/immunology , Autoantigens/immunology , Campylobacter Infections/immunology , Campylobacter jejuni/immunology , Lipid A/immunology , Sodium Channels/immunology , Amino Acid Sequence , Animals , Antibody Specificity , Blotting, Western , Chickens , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Epitopes, B-Lymphocyte/immunology , Gangliosides/immunology , Humans , Immunohistochemistry , Mice , Molecular Sequence Data , Patch-Clamp Techniques , Peptides/immunology , Protein Isoforms/immunology , Sodium Channels/chemistry
14.
Eur J Neurosci ; 27(12): 3095-108, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18598256

ABSTRACT

Principal cells of the medial nucleus of the trapezoid body (MNTB) are simple round neurons that receive a large excitatory synapse (the calyx of Held) and many small inhibitory synapses on the soma. Strangely, these neurons also possess one or two short tufted dendrites, whose function is unknown. Here we assess the role of these MNTB cell dendrites using patch-clamp recordings, imaging and immunohistochemistry techniques. Using outside-out patches and immunohistochemistry, we demonstrate the presence of dendritic Na+ channels. Current-clamp recordings show that tetrodotoxin applied onto dendrites impairs action potential (AP) firing. Using Na+ imaging, we show that the dendrite may serve to maintain AP amplitudes during high-frequency firing, as Na+ clearance indendritic compartments is faster than axonal compartments. Prolonged high-frequency firing can diminish Na+ gradients in the axon while the dendritic gradient remains closer to resting conditions; therefore, the dendrite can provide additional inward current during prolonged firing. Using electron microscopy, we demonstrate that there are small excitatory synaptic boutons on dendrites. Multi-compartment MNTB cell simulations show that, with an active dendrite, dendritic excitatory postsynaptic currents (EPSCs) elicit delayed APs compared with calyceal EPSCs. Together with high- and low-threshold voltage-gated K+ currents, we suggest that the function of the MNTB dendrite is to improve high-fidelity firing, and our modelling results indicate that an active dendrite could contribute to a 'dual' firing mode for MNTB cells (an instantaneous response to calyceal inputs and a delayed response to non-calyceal dendritic excitatory postsynaptic potentials).


Subject(s)
Action Potentials/physiology , Dendrites/physiology , Neurons/physiology , Olivary Nucleus/cytology , Olivary Nucleus/physiology , Action Potentials/drug effects , Animals , Antibodies , Axons/physiology , Calcium/metabolism , Dendrites/ultrastructure , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Microscopy, Electron , Models, Neurological , Neurons/ultrastructure , Patch-Clamp Techniques , Rats , Rats, Wistar , Sodium/metabolism , Sodium Channel Blockers/pharmacology , Sodium Channels/immunology , Sodium Channels/physiology , Sodium-Potassium-Exchanging ATPase/immunology , Sodium-Potassium-Exchanging ATPase/physiology , Tetrodotoxin/pharmacology
15.
BMC Genomics ; 8: 226, 2007 Jul 10.
Article in English | MEDLINE | ID: mdl-17623064

ABSTRACT

BACKGROUND: Voltage-gated Na+ channel beta1 (Scn1b) subunits are multi-functional proteins that play roles in current modulation, channel cell surface expression, cell adhesion, cell migration, and neurite outgrowth. We have shown previously that beta1 modulates electrical excitability in vivo using a mouse model. Scn1b null mice exhibit spontaneous seizures and ataxia, slowed action potential conduction, decreased numbers of nodes of Ranvier in myelinated axons, alterations in nodal architecture, and differences in Na+ channel alpha subunit localization. The early death of these mice at postnatal day 19, however, make them a challenging model system to study. As a first step toward development of an alternative model to investigate the physiological roles of beta1 subunits in vivo we cloned two beta1-like subunit cDNAs from D. rerio. RESULTS: Two beta1-like subunit mRNAs from zebrafish, scn1ba_tv1 and scn1ba_tv2, arise from alternative splicing of scn1ba. The deduced amino acid sequences of Scn1ba_tv1 and Scn1ba_tv2 are identical except for their C-terminal domains. The C-terminus of Scn1ba_tv1 contains a tyrosine residue similar to that found to be critical for ankyrin association and Na+ channel modulation in mammalian beta1. In contrast, Scn1ba_tv2 contains a unique, species-specific C-terminal domain that does not contain a tyrosine. Immunohistochemical analysis shows that, while the expression patterns of Scn1ba_tv1 and Scn1ba_tv2 overlap in some areas of the brain, retina, spinal cord, and skeletal muscle, only Scn1ba_tv1 is expressed in optic nerve where its staining pattern suggests nodal expression. Both scn1ba splice forms modulate Na+ currents expressed by zebrafish scn8aa, resulting in shifts in channel gating mode, increased current amplitude, negative shifts in the voltage dependence of current activation and inactivation, and increases in the rate of recovery from inactivation, similar to the function of mammalian beta1 subunits. In contrast to mammalian beta1, however, neither zebrafish subunit produces a complete shift to the fast gating mode and neither subunit produces complete channel inactivation or recovery from inactivation. CONCLUSION: These data add to our understanding of structure-function relationships in Na+ channel beta1 subunits and establish zebrafish as an ideal system in which to determine the contribution of scn1ba to electrical excitability in vivo.


Subject(s)
Gene Expression Regulation, Developmental , Sodium Channels/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Alternative Splicing , Amino Acid Sequence , Animals , Antibodies/chemistry , Antibodies/pharmacology , Base Sequence , Cells, Cultured , Cloning, Molecular , Cricetinae , Cricetulus , Electrophysiology , Embryo, Nonmammalian , Models, Molecular , Molecular Sequence Data , Protein Isoforms/genetics , Rats , Sequence Homology, Amino Acid , Sodium Channels/immunology , Sodium Channels/metabolism , Sodium Channels/physiology , Species Specificity , Tissue Distribution , Voltage-Gated Sodium Channel beta-1 Subunit , Xenopus laevis , Zebrafish/embryology , Zebrafish Proteins/immunology , Zebrafish Proteins/metabolism
16.
Brain Res ; 1107(1): 1-12, 2006 Aug 30.
Article in English | MEDLINE | ID: mdl-16815341

ABSTRACT

High titer polyclonal antibodies were produced in rabbit against a peptide unique to NaV1.2 sodium channels. NaV1.2 antibodies displayed 500,000-fold greater affinity for the NaV1.2 peptide compared with NaV1.1 or NaV1.3 peptides from the same region. These antibodies, when coupled to Sepharose beads, retained saxitoxin binding sites from solubilized rat brain membranes. Eluted protein from this antibody-affinity column was recognized by antibodies directed against neuronal voltage-gated sodium channels. Rabbit antibodies, which had been partially purified, were used in immunocytochemical localization of the NaV1.2 channel in 50 microm rat brain slices at dilutions of 1:1000 or 1:2000. NaV1.2 channels were predominately localized in unmyelinated fibers in the cortex, hippocampus, spinal cord and hypothalamus. Varicosities were seen in fiber staining which may reflect true varicosities in the fiber or simply varying densities of sodium channels along the fiber. Cell body staining with the NaV1.2 antibody was primarily observed in the hypothalamus. Antibody staining in the cerebellum was complex, with staining observed primarily in posterior lobes and considerably lower amounts of staining observed in anterior lobes. Specific staining was limited to fibers located in the granule and molecular layer, in an orientation consistent with granule cell unmyelinated axon labeling.


Subject(s)
Brain/metabolism , Nerve Tissue Proteins/metabolism , Sodium Channels/metabolism , Spinal Cord/metabolism , Animals , Antibodies/metabolism , Antibody Specificity/physiology , Blotting, Western/methods , Brain/anatomy & histology , Cats , Enzyme-Linked Immunosorbent Assay/methods , Immunohistochemistry/methods , NAV1.1 Voltage-Gated Sodium Channel , NAV1.2 Voltage-Gated Sodium Channel , NAV1.3 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/immunology , Protein Subunits/immunology , Protein Subunits/metabolism , Rats , Sodium Channels/immunology
17.
J Neurosci Methods ; 147(2): 88-98, 2005 Sep 30.
Article in English | MEDLINE | ID: mdl-16111763

ABSTRACT

Voltage-gated Na(+) channel (VGSC) diversity is achieved through a number of mechanisms: multiple subunits, multiple genes encoding the pore-forming VGSC alpha-subunit and multiple gene isoforms generated by alternative splicing. A major type of VGSCalpha alternative splicing is in D1:S3, which has been proposed to be developmentally regulated. We recently reported a D1:S3 spliced form of Na(v)1.5 in human metastatic breast cancer cells. This novel 'neonatal' isoform differs from the counterpart 'adult' form at seven amino acids (in the extracellular loop between S3-S4 of D1). Here, we generated an anti-peptide polyclonal antibody, named NESOpAb, which specifically recognised 'neonatal' but not 'adult' Na(v)1.5 when tested on cells specifically over-expressing one or other of these Na(v)1.5 spliced forms. The antibody was used to investigate developmental expression of 'neonatal' Na(v)1.5 (nNa(v)1.5) in a range of mouse tissues by immunohistochemistry. Overall, the results were consistent with nNa(v)1.5 protein being more abundantly expressed in selected tissues (particularly heart and brain) from neonate as compared to adult animals. Importantly, NESOpAb blocked functional nNa(v)1.5 ion conductance when applied extracellularly at concentrations as low as 0.05 ng/ml. Possible biological and clinical applications of NESOpAb are discussed.


Subject(s)
Alternative Splicing/physiology , Antibodies/immunology , Antibody Specificity , Muscle Proteins/immunology , Sodium Channels/immunology , Animals , Animals, Newborn , Blotting, Western/methods , Cell Line , Cloning, Molecular , Electric Stimulation/methods , Humans , Immunohistochemistry/methods , Membrane Potentials/drug effects , Membrane Potentials/genetics , Membrane Potentials/physiology , Mice , Muscle Proteins/chemistry , Muscle Proteins/genetics , Mutagenesis/physiology , NAV1.5 Voltage-Gated Sodium Channel , Patch-Clamp Techniques/methods , Protein Isoforms/chemistry , Protein Isoforms/immunology , RNA, Messenger/biosynthesis , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction/methods , Sodium Channels/chemistry , Sodium Channels/genetics , Transfection/methods
18.
Am J Physiol Cell Physiol ; 288(1): C141-7, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15355850

ABSTRACT

The purpose of this study was to determine whether there is a correlation between phosphorylation and activity of the epithelial sodium channel (ENaC). The three subunits that form the channel were immunoprecipitated from A6 cells by using specific polyclonal antibodies after labeling cells with (35)S or (32)P. When immune complexes were resolved on SDS-PAGE, the alpha-subunit migrated at 85 and 65 kDa, the beta-subunit at 115 and 100 kDa, and the gamma-subunit at 90 kDa. In the resting state all three subunits were phosphorylated. The alpha-subunit was phosphorylated only in the 65-kDa band, suggesting that the posttranslational modification that gives rise to the rapidly migrating form of alpha is a requirement for phosphorylation. Stimulation with 100 nM insulin for 30 min increased phosphorylation of alpha-, beta-, and gamma-subunits approximately twofold. Exposure to 1 microM aldosterone for 16 h increased protein abundance and phosphorylation proportionately in the three subunits. When insulin was applied to cells pretreated with aldosterone, phosphorylation was also increased approximately twofold, but the total amount of phosphorylated substrate was larger than in control conditions because of the action of aldosterone. This result might explain the synergistic increase in sodium transport under the same conditions. The protein kinase C inhibitor chelerythrine abolished insulin effects and decreased sodium transport and subunit phosphorylation. Together, our findings suggest that ENaC activity is controlled by subunit phosphorylation in cells that endogenously express the channel and the machinery for hormonal stimulation of sodium transport.


Subject(s)
Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Sodium Channels/metabolism , Sodium/metabolism , Aldosterone/pharmacology , Alkaloids , Animals , Antibody Specificity , Benzophenanthridines , Biological Transport/drug effects , Biological Transport/physiology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Epithelial Sodium Channels , Kidney/cytology , Phenanthridines/pharmacology , Phosphorylation/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Subunits , Sodium Channels/chemistry , Sodium Channels/immunology , Xenopus laevis
19.
Neurosci Behav Physiol ; 34(6): 579-85, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15368904

ABSTRACT

The effects of anti-ganglioside antibodies on the functional states of two types of influx Na+ current channels were studied. Experiments used 20% anti-ganglioside antiserum prepared by standard methods by immunizing rabbits with total bovine brain gangliosides. These experiments showed that incubation of neurons in physiological saline containing antiserum induced discordance in the operation of the two types of influx current Na+ channels responsible for spike generation. This reaction was found to be associated with the slowed rate of activation of TTX-sensitive Na+ channels. Synaptic stimulation of cells in the presence of antiserum induced blockade of TTX-insensitive influx Na+ current channels. High-frequency synaptic activation of cells (10 Hz) showed that apart from blockade of TTX-insensitive Na+ channels, anti-ganglioside antibodies prevented plastic rearrangements in the gate system of TTX-sensitive Na+ channels. This resulted in impairment of the development of the acclimation process - the response of the neuron to high-frequency stimulation seen in normal conditions.


Subject(s)
Gangliosides/antagonists & inhibitors , Gangliosides/immunology , Membrane Potentials/drug effects , Neurons/physiology , Sodium Channels/physiology , Synapses/physiology , Animals , Antibodies/pharmacology , Electric Stimulation , Female , Ganglia, Invertebrate/cytology , Ganglia, Invertebrate/immunology , Habituation, Psychophysiologic/drug effects , Habituation, Psychophysiologic/physiology , Immune Sera/immunology , Leeches , Membrane Potentials/immunology , Neurons/drug effects , Neurons/immunology , Rabbits , Sodium Channels/drug effects , Sodium Channels/immunology
20.
Neurology ; 63(1): 43-50, 2004 Jul 13.
Article in English | MEDLINE | ID: mdl-15249609

ABSTRACT

OBJECTIVE: To determine the prevalence of serum antibodies to the ionotropic glutamate receptor 3 (GluR3) in patients with Rasmussen encephalitis (RE), a severe epileptic disorder, and to compare with serum from control subjects and patients with intractable epilepsy (IE). METHODS: The authors looked for serum immunoglobulin (Ig) G antibodies to GluR3 in 30 patients with RE, including two patients who had plasma exchange and 12 who had been treated with IV Igs with varying results, and 49 patients with IE and 23 healthy individuals, using ELISA with GluR3B peptide, Western blot analysis of recombinant full-length GluR3, immunoprecipitation of [35S]- and [125I]-labeled GluR3 extracellular domains, immunohistochemistry on rat brain sections, and electrophysiology of GluR3 expressed in Xenopus oocytes. RESULTS: Low levels of antibodies to the GluR3B peptide were detected using ELISA in only 4 of the 79 patients with epilepsy (2 with RE and 2 with IE); binding to GluR3B in other sera was shown to be nonspecific. One other patient with IE had antibodies to recombinant GluR3 on Western blot analysis. However, none of the sera tested precipitated either the [35S]- or the [125I]-labeled GluR3 domains; none bound to rat brain sections in a manner similar to rabbit antibodies to GluR3; and none of the nine sera tested affected the electrophysiologic function of GluR3. CONCLUSIONS: GluR3 antibodies were only infrequently found in Rasmussen encephalitis or intractable epilepsy.


Subject(s)
Autoantibodies/blood , Encephalitis/immunology , Receptors, AMPA/immunology , Adolescent , Adult , Aged , Amino Acid Sequence , Animals , Antibodies, Anticardiolipin/blood , Antibody Specificity , Autoantigens/immunology , Brain/immunology , Cell Line , Child , Child, Preschool , Encephalitis/blood , Encephalitis/therapy , Epilepsy/blood , Epilepsy/immunology , Epitopes/chemistry , Epitopes/immunology , Female , G(M1) Ganglioside/immunology , Glutamate Decarboxylase/immunology , Humans , Infant , Middle Aged , Models, Molecular , Molecular Sequence Data , Protein Conformation , Rabbits , Rats , Receptors, AMPA/chemistry , Receptors, AMPA/genetics , Receptors, Nicotinic/immunology , Recombinant Proteins/immunology , Sodium Channels/immunology , alpha7 Nicotinic Acetylcholine Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...