Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Brain ; 147(4): 1231-1246, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-37812817

ABSTRACT

Dravet syndrome is an intractable developmental and epileptic encephalopathy caused by de novo variants in SCN1A resulting in haploinsufficiency of the voltage-gated sodium channel Nav1.1. We showed previously that administration of the antisense oligonucleotide STK-001, also called ASO-22, generated using targeted augmentation of nuclear gene output technology to prevent inclusion of the nonsense-mediated decay, or poison, exon 20N in human SCN1A, increased productive Scn1a transcript and Nav1.1 expression and reduced the incidence of electrographic seizures and sudden unexpected death in epilepsy in a mouse model of Dravet syndrome. Here, we investigated the mechanism of action of ASO-84, a surrogate for ASO-22 that also targets splicing of SCN1A exon 20N, in Scn1a+/- Dravet syndrome mouse brain. Scn1a +/- Dravet syndrome and wild-type mice received a single intracerebroventricular injection of antisense oligonucleotide or vehicle at postnatal Day 2. We examined the electrophysiological properties of cortical pyramidal neurons and parvalbumin-positive fast-spiking interneurons in brain slices at postnatal Days 21-25 and measured sodium currents in parvalbumin-positive interneurons acutely dissociated from postnatal Day 21-25 brain slices. We show that, in untreated Dravet syndrome mice, intrinsic cortical pyramidal neuron excitability was unchanged while cortical parvalbumin-positive interneurons showed biphasic excitability with initial hyperexcitability followed by hypoexcitability and depolarization block. Dravet syndrome parvalbumin-positive interneuron sodium current density was decreased compared to wild-type. GABAergic signalling to cortical pyramidal neurons was reduced in Dravet syndrome mice, suggesting decreased GABA release from interneurons. ASO-84 treatment restored action potential firing, sodium current density and GABAergic signalling in Dravet syndrome parvalbumin-positive interneurons. Our work suggests that interneuron excitability is selectively affected by ASO-84. This new work provides critical insights into the mechanism of action of this antisense oligonucleotide and supports the potential of antisense oligonucleotide-mediated upregulation of Nav1.1 as a successful strategy to treat Dravet syndrome.


Subject(s)
Epilepsies, Myoclonic , Oligonucleotides, Antisense , Mice , Animals , Humans , Oligonucleotides, Antisense/pharmacology , Parvalbumins/metabolism , Epilepsies, Myoclonic/genetics , NAV1.1 Voltage-Gated Sodium Channel/genetics , Interneurons/metabolism , gamma-Aminobutyric Acid , Disease Models, Animal
2.
Ann Clin Transl Neurol ; 7(11): 2137-2149, 2020 11.
Article in English | MEDLINE | ID: mdl-32979291

ABSTRACT

OBJECTIVE: Human variants in voltage-gated sodium channel (VGSC) α and ß subunit genes are linked to developmental and epileptic encephalopathies (DEEs). Inherited, biallelic, loss-of-function variants in SCN1B, encoding the ß1/ß1B subunits, are linked to early infantile DEE (EIEE52). De novo, monoallelic variants in SCN1A (Nav1.1), SCN2A (Nav1.2), SCN3A (Nav1.3), and SCN8A (Nav1.6) are also linked to DEEs. While these VGSC-linked DEEs have similar presentations, they have diverse mechanisms of altered neuronal excitability. Mouse models have suggested that Scn2a-, Scn3a-, and Scn8a-linked DEE variants are, in general, gain of function, resulting in increased persistent or resurgent sodium current (INa ) and pyramidal neuron hyperexcitability. In contrast, Scn1a-linked DEE variants, in general, are loss-of-function, resulting in decreased INa and hypoexcitability of fast-spiking interneurons. VGSC ß1 subunits associate with Nav1.1, Nav1.2, Nav1.3, and Nav1.6 and are expressed throughout the brain, raising the possibility that insults to both pyramidal and interneuron excitability may drive EIEE52 pathophysiology. METHODS: We investigated excitability defects in pyramidal and parvalbumin-positive (PV +) interneurons in the Scn1b-/- model of EIEE52. We also used Scn1bFL/FL mice to delete Scn1b in specific neuronal populations. RESULTS: Scn1b-/- cortical PV + interneurons were hypoexcitable, with reduced INa density. Scn1b-/- cortical pyramidal neurons had population-specific changes in excitability and impaired INa density. Scn1b deletion in PV + neurons resulted in 100% lethality, whereas deletion in Emx1 + or Camk2a + neurons did not affect survival. INTERPRETATION: This work suggests that SCN1B-linked DEE variants impact both excitatory and inhibitory neurons, leading to the increased severity of EIEE52 relative to other DEEs.


Subject(s)
Cerebral Cortex/physiopathology , Interneurons/physiology , Pyramidal Cells/physiology , Spasms, Infantile/genetics , Spasms, Infantile/physiopathology , Voltage-Gated Sodium Channel beta-1 Subunit/physiology , Animals , Cell Count , Disease Models, Animal , Humans , Infant, Newborn , Interneurons/cytology , Mice , Mice, Congenic , Mice, Inbred C57BL , Parvalbumins/metabolism , Pyramidal Cells/cytology , Voltage-Gated Sodium Channel beta-1 Subunit/genetics
3.
Ann Clin Transl Neurol ; 6(6): 1121-1126, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31211177

ABSTRACT

Pathogenic loss-of-function variants in SCN1B are linked to Dravet syndrome (DS). Previous work suggested that neuronal pathfinding defects underlie epileptogenesis and SUDEP in the Scn1b null mouse model of DS. We tested this hypothesis by inducing Scn1b deletion in adult mice that had developed normally. Epilepsy and SUDEP, which occur by postnatal day 21 in Scn1b null animals, were observed within 20 days of induced Scn1b deletion in adult mice, suggesting that epileptogenesis in SCN1B-DS does not result from defective brain development. Thus, the developmental brain defects observed previously in Scn1b null mice may model other co-morbidities of DS.


Subject(s)
Seizures/genetics , Seizures/physiopathology , Sudden Unexpected Death in Epilepsy , Voltage-Gated Sodium Channel beta-1 Subunit/deficiency , Animals , Brain/metabolism , Epilepsy/genetics , Epilepsy/physiopathology , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
Sci Rep ; 9(1): 6210, 2019 04 17.
Article in English | MEDLINE | ID: mdl-30996233

ABSTRACT

Dravet syndrome (DS) is a catastrophic developmental and epileptic encephalopathy characterized by severe, pharmacoresistant seizures and the highest risk of Sudden Unexpected Death in Epilepsy (SUDEP) of all epilepsy syndromes. Here, we investigated the time course of maturation of neuronal GABAergic signaling in the Scn1b-/- and Scn1a+/- mouse models of DS. We found that GABAergic signaling remains immature in both DS models, with a depolarized reversal potential for GABAA-evoked currents compared to wildtype in the third postnatal week. Treatment of Scn1b-/- mice with bumetanide resulted in a delay in SUDEP onset compared to controls in a subset of mice, without prevention of seizure activity or amelioration of failure to thrive. We propose that delayed maturation of GABAergic signaling may contribute to epileptogenesis in SCN1B- and SCN1A-linked DS. Thus, targeting the polarity of GABAergic signaling in brain may be an effective therapeutic strategy to reduce SUDEP risk in DS.


Subject(s)
Epilepsies, Myoclonic/etiology , NAV1.1 Voltage-Gated Sodium Channel/deficiency , Voltage-Gated Sodium Channel beta-1 Subunit/deficiency , gamma-Aminobutyric Acid/metabolism , Animals , Bumetanide/therapeutic use , Death, Sudden , Disease Models, Animal , Epilepsies, Myoclonic/drug therapy , Epilepsies, Myoclonic/metabolism , Epilepsies, Myoclonic/mortality , Epilepsy , Gene Knockdown Techniques , Mice , Seizures , Time Factors
5.
Proc Natl Acad Sci U S A ; 114(9): 2383-2388, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28193882

ABSTRACT

Patients with early infantile epileptic encephalopathy (EIEE) experience severe seizures and cognitive impairment and are at increased risk for sudden unexpected death in epilepsy (SUDEP). EIEE13 [Online Mendelian Inheritance in Man (OMIM) # 614558] is caused by de novo missense mutations in the voltage-gated sodium channel gene SCN8A Here, we investigated the neuronal phenotype of a mouse model expressing the gain-of-function SCN8A patient mutation, p.Asn1768Asp (Nav1.6-N1768D). Our results revealed regional and neuronal subtype specificity in the effects of the N1768D mutation. Acutely dissociated hippocampal neurons from Scn8aN1768D/+ mice showed increases in persistent sodium current (INa) density in CA1 pyramidal but not bipolar neurons. In CA3, INa,P was increased in both bipolar and pyramidal neurons. Measurement of action potential (AP) firing in Scn8aN1768D/+ pyramidal neurons in brain slices revealed early afterdepolarization (EAD)-like AP waveforms in CA1 but not in CA3 hippocampal or layer II/III neocortical neurons. The maximum spike frequency evoked by depolarizing current injections in Scn8aN1768D/+ CA1, but not CA3 or neocortical, pyramidal cells was significantly reduced compared with WT. Spontaneous firing was observed in subsets of neurons in CA1 and CA3, but not in the neocortex. The EAD-like waveforms of Scn8aN1768D/+ CA1 hippocampal neurons were blocked by tetrodotoxin, riluzole, and SN-6, implicating elevated persistent INa and reverse mode Na/Ca exchange in the mechanism of hyperexcitability. Our results demonstrate that Scn8a plays a vital role in neuronal excitability and provide insight into the mechanism and future treatment of epileptogenesis in EIEE13.


Subject(s)
CA1 Region, Hippocampal/metabolism , Mutation , NAV1.6 Voltage-Gated Sodium Channel/genetics , Pyramidal Cells/metabolism , Spasms, Infantile/genetics , Action Potentials/drug effects , Amino Acid Substitution , Animals , Benzyl Compounds/pharmacology , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/pathology , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/pathology , Disease Models, Animal , Gene Expression , Humans , Mice , Mice, Transgenic , NAV1.6 Voltage-Gated Sodium Channel/metabolism , Neocortex/drug effects , Neocortex/metabolism , Neocortex/pathology , Organ Specificity , Pyramidal Cells/drug effects , Pyramidal Cells/pathology , Riluzole/pharmacology , Sodium Channel Blockers/pharmacology , Spasms, Infantile/metabolism , Spasms, Infantile/physiopathology , Tetrodotoxin/pharmacology , Thiazolidines/pharmacology
6.
J Neurosci ; 36(23): 6213-24, 2016 06 08.
Article in English | MEDLINE | ID: mdl-27277800

ABSTRACT

UNLABELLED: Voltage-gated sodium channel (VGSC) ß subunits signal through multiple pathways on multiple time scales. In addition to modulating sodium and potassium currents, ß subunits play nonconducting roles as cell adhesion molecules, which allow them to function in cell-cell communication, neuronal migration, neurite outgrowth, neuronal pathfinding, and axonal fasciculation. Mutations in SCN1B, encoding VGSC ß1 and ß1B, are associated with epilepsy. Autosomal-dominant SCN1B-C121W, the first epilepsy-associated VGSC mutation identified, results in genetic epilepsy with febrile seizures plus (GEFS+). This mutation has been shown to disrupt both the sodium-current-modulatory and cell-adhesive functions of ß1 subunits expressed in heterologous systems. The goal of this study was to compare mice heterozygous for Scn1b-C121W (Scn1b(+/W)) with mice heterozygous for the Scn1b-null allele (Scn1b(+/-)) to determine whether the C121W mutation results in loss-of-function in vivo We found that Scn1b(+/W) mice were more susceptible than Scn1b(+/-) and Scn1b(+/+) mice to hyperthermia-induced convulsions, a model of pediatric febrile seizures. ß1-C121W subunits are expressed at the neuronal cell surface in vivo However, despite this, ß1-C121W polypeptides are incompletely glycosylated and do not associate with VGSC α subunits in the brain. ß1-C121W subcellular localization is restricted to neuronal cell bodies and is not detected at axon initial segments in the cortex or cerebellum or at optic nerve nodes of Ranvier of Scn1b(W/W) mice. These data, together with our previous results showing that ß1-C121W cannot participate in trans-homophilic cell adhesion, lead to the hypothesis that SCN1B-C121W confers a deleterious gain-of-function in human GEFS+ patients. SIGNIFICANCE STATEMENT: The mechanisms underlying genetic epilepsy syndromes are poorly understood. Closing this gap in knowledge is essential to the development of new medicines to treat epilepsy. We have used mouse models to understand the mechanism of a mutation in the sodium channel gene SCN1B linked to genetic epilepsy with febrile seizures plus. We report that sodium channel ß1 subunit proteins encoded by this mutant gene are expressed at the surface of neuronal cell bodies; however, they do not associate with the ion channel complex nor are they transported to areas of the axon that are critical for proper neuronal firing. We conclude that this disease-causing mutation is not simply a loss-of-function, but instead results in a deleterious gain-of-function in the brain.


Subject(s)
Epilepsy/genetics , Neurons/physiology , Polymorphism, Single Nucleotide/genetics , Voltage-Gated Sodium Channel beta-1 Subunit/genetics , Voltage-Gated Sodium Channel beta-1 Subunit/metabolism , Animals , Animals, Newborn , Biotinylation , Cells, Cultured , Cerebral Cortex/cytology , Cysteine/genetics , Disease Models, Animal , Epilepsy/etiology , Epilepsy/pathology , Fever/complications , Gene Expression Regulation, Developmental/genetics , Immunoprecipitation , Mice , Mice, Transgenic , Statistics, Nonparametric , Tryptophan/genetics
7.
Annu Rev Physiol ; 77: 481-504, 2015.
Article in English | MEDLINE | ID: mdl-25668026

ABSTRACT

Voltage-gated sodium channels (VGSCs) are responsible for the initiation and propagation of action potentials in excitable cells. VGSCs in mammalian brain are heterotrimeric complexes of α and ß subunits. Although ß subunits were originally termed auxiliary, we now know that they are multifunctional signaling molecules that play roles in both excitable and nonexcitable cell types and with or without the pore-forming α subunit present. ß subunits function in VGSC and potassium channel modulation, cell adhesion, and gene regulation, with particularly important roles in brain development. Mutations in the genes encoding ß subunits are linked to a number of diseases, including epilepsy, sudden death syndromes like SUDEP and SIDS, and cardiac arrhythmia. Although VGSC ß subunit-specific drugs have not yet been developed, this protein family is an emerging therapeutic target.


Subject(s)
Channelopathies/genetics , Channelopathies/physiopathology , Mutation/genetics , Voltage-Gated Sodium Channel beta Subunits/genetics , Voltage-Gated Sodium Channel beta Subunits/physiology , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Autistic Disorder/genetics , Autistic Disorder/physiopathology , Cell Adhesion/genetics , Cell Adhesion/physiology , Epilepsy/genetics , Epilepsy/physiopathology , Gene Expression Profiling , Humans , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/physiopathology , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/physiology
8.
Ann Neurol ; 74(1): 128-39, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23821540

ABSTRACT

OBJECTIVE: Neuronal channelopathies cause brain disorders, including epilepsy, migraine, and ataxia. Despite the development of mouse models, pathophysiological mechanisms for these disorders remain uncertain. One particularly devastating channelopathy is Dravet syndrome (DS), a severe childhood epilepsy typically caused by de novo dominant mutations in the SCN1A gene encoding the voltage-gated sodium channel Na(v) 1.1. Heterologous expression of mutant channels suggests loss of function, raising the quandary of how loss of sodium channels underlying action potentials produces hyperexcitability. Mouse model studies suggest that decreased Na(v) 1.1 function in interneurons causes disinhibition. We aim to determine how mutant SCN1A affects human neurons using the induced pluripotent stem cell (iPSC) method to generate patient-specific neurons. METHODS: Here we derive forebrain-like pyramidal- and bipolar-shaped neurons from 2 DS subjects and 3 human controls by iPSC reprogramming of fibroblasts. DS and control iPSC-derived neurons are compared using whole-cell patch clamp recordings. Sodium current density and intrinsic neuronal excitability are examined. RESULTS: Neural progenitors from DS and human control iPSCs display a forebrain identity and differentiate into bipolar- and pyramidal-shaped neurons. DS patient-derived neurons show increased sodium currents in both bipolar- and pyramidal-shaped neurons. Consistent with increased sodium currents, both types of patient-derived neurons show spontaneous bursting and other evidence of hyperexcitability. Sodium channel transcripts are not elevated, consistent with a post-translational mechanism. INTERPRETATION: These data demonstrate that epilepsy patient-specific iPSC-derived neurons are useful for modeling epileptic-like hyperactivity. Our findings reveal a previously unrecognized cell-autonomous epilepsy mechanism potentially underlying DS, and offer a platform for screening new antiepileptic therapies.


Subject(s)
Epilepsies, Myoclonic/genetics , Epilepsies, Myoclonic/pathology , Mutation/genetics , NAV1.1 Voltage-Gated Sodium Channel/genetics , Neurons/physiology , Cell Differentiation , Cells, Cultured , Child , Female , Fibroblasts/physiology , Humans , Inhibitory Postsynaptic Potentials/genetics , Male , Membrane Potentials , Patch-Clamp Techniques
9.
Proc Natl Acad Sci U S A ; 110(3): 1089-94, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23277545

ABSTRACT

Voltage-gated Na(+) channel (VGSC) ß1 subunits, encoded by SCN1B, are multifunctional channel modulators and cell adhesion molecules (CAMs). Mutations in SCN1B are associated with the genetic epilepsy with febrile seizures plus (GEFS+) spectrum disorders in humans, and Scn1b-null mice display severe spontaneous seizures and ataxia from postnatal day (P)10. The goal of this study was to determine changes in neuronal pathfinding during early postnatal brain development of Scn1b-null mice to test the hypothesis that these CAM-mediated roles of Scn1b may contribute to the development of hyperexcitability. c-Fos, a protein induced in response to seizure activity, was up-regulated in the Scn1b-null brain at P16 but not at P5. Consistent with this, epileptiform activity was observed in hippocampal and cortical slices prepared from the P16 but not from the P5-P7 Scn1b-null brain. On the basis of these results, we investigated neuronal pathfinding at P5. We observed disrupted fasciculation of parallel fibers in the P5 null cerebellum. Further, P5 null mice showed reduced neuron density in the dentate gyrus granule cell layer, increased proliferation of granule cell precursors in the hilus, and defective axonal extension and misorientation of somata and processes of inhibitory neurons in the dentate gyrus and CA1. Thus, Scn1b is critical for neuronal proliferation, migration, and pathfinding during the critical postnatal period of brain development. We propose that defective neuronal proliferation, migration, and pathfinding in response to Scn1b deletion may contribute to the development of hyperexcitability.


Subject(s)
Brain/growth & development , Brain/metabolism , Voltage-Gated Sodium Channel beta-1 Subunit/deficiency , Age Factors , Animals , Ataxia/etiology , Ataxia/metabolism , Ataxia/pathology , Body Patterning/genetics , Body Patterning/physiology , Brain/cytology , Cell Movement , Cell Proliferation , Electrophysiological Phenomena , Hippocampus/growth & development , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Neurons/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Seizures/etiology , Seizures/metabolism , Seizures/pathology , Voltage-Gated Sodium Channel beta-1 Subunit/genetics , Voltage-Gated Sodium Channel beta-1 Subunit/metabolism
10.
J Neurosci ; 31(41): 14577-91, 2011 Oct 12.
Article in English | MEDLINE | ID: mdl-21994374

ABSTRACT

Scn1b-null mice have a severe neurological and cardiac phenotype. Human mutations in SCN1B result in epilepsy and cardiac arrhythmia. SCN1B is expressed as two developmentally regulated splice variants, ß1 and ß1B, that are each expressed in brain and heart in rodents and humans. Here, we studied the structure and function of ß1B and investigated a novel human SCN1B epilepsy-related mutation (p.G257R) unique to ß1B. We show that wild-type ß1B is not a transmembrane protein, but a soluble protein expressed predominantly during embryonic development that promotes neurite outgrowth. Association of ß1B with voltage-gated Na+ channels Na(v)1.1 or Na(v)1.3 is not detectable by immunoprecipitation and ß1B does not affect Na(v)1.3 cell surface expression as measured by [(3)H]saxitoxin binding. However, ß1B coexpression results in subtle alteration of Na(v)1.3 currents in transfected cells, suggesting that ß1B may modulate Na+ current in brain. Similar to the previously characterized p.R125C mutation, p.G257R results in intracellular retention of ß1B, generating a functional null allele. In contrast, two other SCN1B mutations associated with epilepsy, p.C121W and p.R85H, are expressed at the cell surface. We propose that ß1B p.G257R may contribute to epilepsy through a mechanism that includes intracellular retention resulting in aberrant neuronal pathfinding.


Subject(s)
Epilepsy/genetics , Mutation/genetics , Sodium Channels/genetics , Sodium Channels/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Arginine/genetics , Biotinylation/methods , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cells, Cultured , Cerebellum , Cricetinae , Cricetulus , Female , Gene Expression Regulation, Developmental/genetics , Genotype , Glycine/genetics , Humans , Immunoprecipitation/methods , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NAV1.3 Voltage-Gated Sodium Channel , Neurites/metabolism , Neurons/cytology , Neurons/physiology , Patch-Clamp Techniques , Protein Isoforms/genetics , RNA, Messenger/metabolism , Saxitoxin/pharmacokinetics , Sodium Channels/deficiency , Statistics, Nonparametric , Transfection/methods , Tritium/pharmacokinetics , Voltage-Gated Sodium Channel beta-1 Subunit
11.
Neurosci Lett ; 480(1): 40-3, 2010 Aug 09.
Article in English | MEDLINE | ID: mdl-20641161

ABSTRACT

The dopamine transporter (DAT) is a critical regulator of dopaminergic neurotransmission. Research in both rat striatum and heterologous cells suggests that protein kinase C beta (PKCbeta) is important for proper trafficking of DAT. However, a critical gap that is missing from the literature is the localization of PKCbeta to mesencephalic dopaminergic neurons. In this study we examined the co-localization of DAT, which serves to identify dopaminergic neurons, and PKCbeta in mesencephalic dopaminergic cells. Using immunofluorescence and confocal microscopy, we demonstrated co-localization of DAT and PKCbeta in primary cultures of mesencephalic neurons and in dopamine neurons in rat substantia nigra and ventral tegmental area. PKCbetawas not specific for dopamine neurons in the two brain regions. This is the first demonstration of co-localization of PKCbeta and DAT in mesencephalic neurons. The co-localization of PKCbeta with DAT in mesencephalic neurons corroborates our previous studies demonstrating a role for PKCbeta in DAT function.


Subject(s)
Dopamine Plasma Membrane Transport Proteins/metabolism , Mesencephalon/metabolism , Neurons/metabolism , Protein Kinase C/metabolism , Age Factors , Animals , Animals, Newborn , Cells, Cultured , Dopamine/metabolism , Female , Fluorescent Antibody Technique , Mesencephalon/cytology , Microscopy, Confocal , Protein Kinase C beta , Rats , Rats, Sprague-Dawley , Substantia Nigra/metabolism , Ventral Tegmental Area/metabolism
12.
J Neurosci ; 30(2): 600-8, 2010 Jan 13.
Article in English | MEDLINE | ID: mdl-20071523

ABSTRACT

Patients with PMP22 deficiency present with focal sensory and motor deficits when peripheral nerves are stressed by mechanical force. It has been hypothesized that these focal deficits are due to mechanically induced conduction block (CB). To test this hypothesis, we induced 60-70% CB (defined by electrophysiological criteria) by nerve compression in an authentic mouse model of hereditary neuropathy with liability to pressure palsies (HNPP) with an inactivation of one of the two pmp22 alleles (pmp22(+/-)). Induction time for the CB was significantly shorter in pmp22(+/-) mice than that in pmp22(+/+) mice. This shortened induction was also found in myelin-associated glycoprotein knock-out mice, but not in the mice with deficiency of myelin protein zero, a major structural protein of compact myelin. Pmp22(+/-) nerves showed intact tomacula with no segmental demyelination in both noncompressed and compressed conditions, normal molecular architecture, and normal concentration of voltage-gated sodium channels by [(3)H]-saxitoxin binding assay. However, focal constrictions were observed in the axonal segments enclosed by tomacula, a pathological hallmark of HNPP. The constricted axons increase axial resistance to action potential propagation, which may hasten the induction of CB in Pmp22 deficiency. Together, these results demonstrate that a function of Pmp22 is to protect the nerve from mechanical injury.


Subject(s)
Muscle, Skeletal/physiopathology , Myelin Proteins/deficiency , Neural Conduction/physiology , Peripheral Nerves/physiopathology , Action Potentials/physiology , Age Factors , Animals , Biophysics , Disease Models, Animal , Electric Stimulation/methods , Gene Expression Regulation/genetics , Kv1.2 Potassium Channel/metabolism , Luminescent Proteins/genetics , Mice , Mice, Knockout , Microscopy, Electron, Transmission/methods , Myelin Basic Protein/metabolism , Myelin-Associated Glycoprotein/metabolism , Nerve Block/methods , Nerve Fibers/metabolism , Nerve Fibers/pathology , Neural Conduction/genetics , Peripheral Nerves/pathology , Peripheral Nerves/ultrastructure , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/pathology , Peripheral Nervous System Diseases/physiopathology , Protein Binding/genetics , Reaction Time/genetics , Saxitoxin/pharmacokinetics , Sodium Channels/metabolism , Tritium/pharmacokinetics
13.
J Neurosci ; 29(34): 10764-78, 2009 Aug 26.
Article in English | MEDLINE | ID: mdl-19710327

ABSTRACT

Dravet syndrome (also called severe myoclonic epilepsy of infancy) is one of the most severe forms of childhood epilepsy. Most patients have heterozygous mutations in SCN1A, encoding voltage-gated sodium channel Na(v)1.1 alpha subunits. Sodium channels are modulated by beta1 subunits, encoded by SCN1B, a gene also linked to epilepsy. Here we report the first patient with Dravet syndrome associated with a recessive mutation in SCN1B (p.R125C). Biochemical characterization of p.R125C in a heterologous system demonstrated little to no cell surface expression despite normal total cellular expression. This occurred regardless of coexpression of Na(v)1.1 alpha subunits. Because the patient was homozygous for the mutation, these data suggest a functional SCN1B null phenotype. To understand the consequences of the lack of beta1 cell surface expression in vivo, hippocampal slice recordings were performed in Scn1b(-/-) versus Scn1b(+/+) mice. Scn1b(-/-) CA3 neurons fired evoked action potentials with a significantly higher peak voltage and significantly greater amplitude compared with wild type. However, in contrast to the Scn1a(+/-) model of Dravet syndrome, we found no measurable differences in sodium current density in acutely dissociated CA3 hippocampal neurons. Whereas Scn1b(-/-) mice seize spontaneously, the seizure susceptibility of Scn1b(+/-) mice was similar to wild type, suggesting that, like the parents of this patient, one functional SCN1B allele is sufficient for normal control of electrical excitability. We conclude that SCN1B p.R125C is an autosomal recessive cause of Dravet syndrome through functional gene inactivation.


Subject(s)
Epilepsies, Myoclonic/genetics , Epilepsies, Myoclonic/physiopathology , Polymorphism, Single Nucleotide/genetics , Sodium Channels/genetics , Animals , Arginine/genetics , Biophysics , Cell Line, Transformed , Cysteine/genetics , DNA Mutational Analysis , Disease Models, Animal , Electric Stimulation , Epilepsies, Myoclonic/mortality , Female , Green Fluorescent Proteins/genetics , Hippocampus/pathology , Humans , In Vitro Techniques , Infant , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , NAV1.1 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/deficiency , Oocytes , Sodium Channels/deficiency , Temperature , Transfection , Twins , Voltage-Gated Sodium Channel beta-1 Subunit , Xenopus laevis
14.
Mol Cell Neurosci ; 40(2): 143-55, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19013247

ABSTRACT

Multiple sclerosis (MS) is a CNS disease that includes demyelination and axonal degeneration. Voltage-gated Na+ channels are abnormally expressed and distributed in MS and its animal model, Experimental Allergic Encephalomyelitis (EAE). Up-regulation of Na+ channels along demyelinated axons is proposed to lead to axonal loss in MS/EAE. We hypothesized that Na+ channel beta2 subunits (encoded by Scn2b) are involved in MS/EAE pathogenesis, as beta2 is responsible for regulating levels of channel cell surface expression in neurons. We induced non-relapsing EAE in Scn2b(+/+) and Scn2b(-/-) mice on the C57BL/6 background. Scn2b(-/-) mice display a dramatic reduction in EAE symptom severity and lethality as compared to wildtype, with significant decreases in axonal degeneration and axonal loss. Scn2b(-/-) mice show normal peripheral immune cell populations, T cell proliferation, cytokine release, and immune cell infiltration into the CNS in response to EAE, suggesting that Scn2b inactivation does not compromise immune function. Our data suggest that loss of beta2 is neuroprotective in EAE by prevention of Na+ channel up-regulation in response to demyelination.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/physiopathology , Multiple Sclerosis/metabolism , Protein Subunits/metabolism , Sodium Channels/metabolism , Animals , Axons/metabolism , Axons/pathology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/pathology , Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/pathology , Myelin-Oligodendrocyte Glycoprotein , Optic Nerve/cytology , Optic Nerve/metabolism , Optic Nerve/pathology , Peptide Fragments/immunology , Protein Subunits/genetics , Sodium Channels/genetics , Spinal Cord/cytology , Spinal Cord/metabolism , Spinal Cord/pathology , Spleen/cytology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Voltage-Gated Sodium Channel beta-2 Subunit
15.
J Neurosci ; 24(16): 4030-42, 2004 Apr 21.
Article in English | MEDLINE | ID: mdl-15102918

ABSTRACT

Sodium channel beta1 subunits modulate alpha subunit gating and cell surface expression and participate in cell adhesive interactions in vitro. beta1-/- mice appear ataxic and display spontaneous generalized seizures. In the optic nerve, the fastest components of the compound action potential are slowed and the number of mature nodes of Ranvier is reduced, but Na(v)1.6, contactin, caspr 1, and K(v)1 channels are all localized normally at nodes. At the ultrastructural level, the paranodal septate-like junctions immediately adjacent to the node are missing in a subset of axons, suggesting that beta1 may participate in axo-glial communication at the periphery of the nodal gap. Sodium currents in dissociated hippocampal neurons are normal, but Na(v)1.1 expression is reduced and Na(v)1.3 expression is increased in a subset of pyramidal neurons in the CA2/CA3 region, suggesting a basis for the epileptic phenotype. Our results show that beta1 subunits play important roles in the regulation of sodium channel density and localization, are involved in axo-glial communication at nodes of Ranvier, and are required for normal action potential conduction and control of excitability in vivo.


Subject(s)
Neurons/metabolism , Ranvier's Nodes/ultrastructure , Sodium Channels/metabolism , Action Potentials/physiology , Animals , Ataxia/complications , Ataxia/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cells, Cultured , Contactins , Dwarfism/complications , Dwarfism/genetics , Epilepsy/complications , Epilepsy/genetics , Mice , Mice, Knockout , NAV1.1 Voltage-Gated Sodium Channel , NAV1.6 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/metabolism , Neurons/ultrastructure , Patch-Clamp Techniques , Phenotype , Potassium Channels/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Pyramidal Cells/metabolism , Sodium/metabolism , Sodium Channels/genetics , Stem Cells/metabolism , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...