Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 149
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 165(2): 434-448, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26997484

ABSTRACT

Mutations in the Kv3.3 potassium channel (KCNC3) cause cerebellar neurodegeneration and impair auditory processing. The cytoplasmic C terminus of Kv3.3 contains a proline-rich domain conserved in proteins that activate actin nucleation through Arp2/3. We found that Kv3.3 recruits Arp2/3 to the plasma membrane, resulting in formation of a relatively stable cortical actin filament network resistant to cytochalasin D that inhibits fast barbed end actin assembly. These Kv3.3-associated actin structures are required to prevent very rapid N-type channel inactivation during short depolarizations of the plasma membrane. The effects of Kv3.3 on the actin cytoskeleton are mediated by the binding of the cytoplasmic C terminus of Kv3.3 to Hax-1, an anti-apoptotic protein that regulates actin nucleation through Arp2/3. A human Kv3.3 mutation within a conserved proline-rich domain produces channels that bind Hax-1 but are impaired in recruiting Arp2/3 to the plasma membrane, resulting in growth cones with deficient actin veils in stem cell-derived neurons.


Subject(s)
Actin Cytoskeleton/metabolism , Actin-Related Protein 2/metabolism , Actin-Related Protein 3/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Shaw Potassium Channels/metabolism , Spinocerebellar Ataxias/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Amino Acid Sequence , Cell Membrane/metabolism , Molecular Sequence Data , Mutation , Neurons/metabolism , Pluripotent Stem Cells/metabolism , Shaw Potassium Channels/chemistry , Shaw Potassium Channels/genetics , Signal Transduction , rac GTP-Binding Proteins/metabolism
2.
Proc Natl Acad Sci U S A ; 121(3): e2307776121, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38194456

ABSTRACT

De novo heterozygous variants in KCNC2 encoding the voltage-gated potassium (K+) channel subunit Kv3.2 are a recently described cause of developmental and epileptic encephalopathy (DEE). A de novo variant in KCNC2 c.374G > A (p.Cys125Tyr) was identified via exome sequencing in a patient with DEE. Relative to wild-type Kv3.2, Kv3.2-p.Cys125Tyr induces K+ currents exhibiting a large hyperpolarizing shift in the voltage dependence of activation, accelerated activation, and delayed deactivation consistent with a relative stabilization of the open conformation, along with increased current density. Leveraging the cryogenic electron microscopy (cryo-EM) structure of Kv3.1, molecular dynamic simulations suggest that a strong π-π stacking interaction between the variant Tyr125 and Tyr156 in the α-6 helix of the T1 domain promotes a relative stabilization of the open conformation of the channel, which underlies the observed gain of function. A multicompartment computational model of a Kv3-expressing parvalbumin-positive cerebral cortex fast-spiking γ-aminobutyric acidergic (GABAergic) interneuron (PV-IN) demonstrates how the Kv3.2-Cys125Tyr variant impairs neuronal excitability and dysregulates inhibition in cerebral cortex circuits to explain the resulting epilepsy.


Subject(s)
Epilepsy , Shaw Potassium Channels , Humans , Shaw Potassium Channels/genetics , Interneurons , Cerebral Cortex , Epilepsy/genetics , Mutation
3.
Proc Natl Acad Sci U S A ; 119(31): e2204901119, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35881790

ABSTRACT

Although a wide variety of genetic tools has been developed to study learning and memory, the molecular basis of memory encoding remains incompletely understood. Here, we undertook an unbiased approach to identify novel genes critical for memory encoding. From a large-scale, in vivo mutagenesis screen using contextual fear conditioning, we isolated in mice a mutant, named Clueless, with spatial learning deficits. A causative missense mutation (G434V) was found in the voltage-gated potassium channel, subfamily C member 3 (Kcnc3) gene in a region that encodes a transmembrane voltage sensor. Generation of a Kcnc3G434V CRISPR mutant mouse confirmed this mutation as the cause of the learning defects. While G434V had no effect on transcription, translation, or trafficking of the channel, electrophysiological analysis of the G434V mutant channel revealed a complete loss of voltage-gated conductance, a broadening of the action potential, and decreased neuronal firing. Together, our findings have revealed a role for Kcnc3 in learning and memory.


Subject(s)
Hippocampus , Learning Disabilities , Memory , Mutation, Missense , Shaw Potassium Channels , Action Potentials/physiology , Animals , Hippocampus/physiopathology , Learning Disabilities/genetics , Mice , Mice, Inbred C57BL , Shaw Potassium Channels/genetics , Shaw Potassium Channels/physiology
4.
BMC Neurol ; 24(1): 169, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38783211

ABSTRACT

BACKGROUND: Progressive Myoclonic Epilepsy (PME) is a group of rare diseases that are difficult to differentiate from one another based on phenotypical characteristics. CASE REPORT: We report a case of PME type 7 due to a pathogenic variant in KCNC1 with myoclonus improvement after epileptic seizures. DISCUSSION: Myoclonus improvement after seizures may be a clue to the diagnosis of Progressive Myoclonic Epilepsy type 7.


Subject(s)
Myoclonic Epilepsies, Progressive , Seizures , Humans , Myoclonic Epilepsies, Progressive/complications , Myoclonic Epilepsies, Progressive/diagnosis , Seizures/diagnosis , Seizures/complications , Seizures/etiology , Seizures/drug therapy , Myoclonus/diagnosis , Myoclonus/etiology , Myoclonus/complications , Myoclonus/drug therapy , Male , Shaw Potassium Channels/genetics , Female , Electroencephalography/methods
5.
J Biol Chem ; 296: 100302, 2021.
Article in English | MEDLINE | ID: mdl-33465376

ABSTRACT

3,4-Diaminopyridine (3,4-DAP) increases transmitter release from neuromuscular junctions (NMJs), and low doses of 3,4-DAP (estimated to reach ∼1 µM in serum) are the Food and Drug Administration (FDA)-approved treatment for neuromuscular weakness caused by Lambert-Eaton myasthenic syndrome. Canonically, 3,4-DAP is thought to block voltage-gated potassium (Kv) channels, resulting in prolongation of the presynaptic action potential (AP). However, recent reports have shown that low millimolar concentrations of 3,4-DAP have an off-target agonist effect on the Cav1 subtype ("L-type") of voltage-gated calcium (Cav) channels and have speculated that this agonist effect might contribute to 3,4-DAP effects on transmitter release at the NMJ. To address 3,4-DAP's mechanism(s) of action, we first used the patch-clamp electrophysiology to characterize the concentration-dependent block of 3,4-DAP on the predominant presynaptic Kv channel subtypes found at the mammalian NMJ (Kv3.3 and Kv3.4). We identified a previously unreported high-affinity (1-10 µM) partial antagonist effect of 3,4-DAP in addition to the well-known low-affinity (0.1-1 mM) antagonist activity. We also showed that 1.5-µM DAP had no effects on Cav1.2 or Cav2.1 current. Next, we used voltage imaging to show that 1.5- or 100-µM 3,4-DAP broadened the AP waveform in a dose-dependent manner, independent of Cav1 calcium channels. Finally, we demonstrated that 1.5- or 100-µM 3,4-DAP augmented transmitter release in a dose-dependent manner and this effect was also independent of Cav1 channels. From these results, we conclude that low micromolar concentrations of 3,4-DAP act solely on Kv channels to mediate AP broadening and enhance transmitter release at the NMJ.


Subject(s)
Amifampridine/pharmacology , Neuromuscular Agents/pharmacology , Neuromuscular Junction/drug effects , Potassium Channel Blockers/pharmacology , Presynaptic Terminals/drug effects , Shaw Potassium Channels/metabolism , Acetylcholine/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Calcium Channels, N-Type/genetics , Calcium Channels, N-Type/metabolism , Dose-Response Relationship, Drug , Female , Gene Expression , Male , Mice , Microelectrodes , Neuromuscular Junction/metabolism , Presynaptic Terminals/metabolism , Rana pipiens , Shaw Potassium Channels/antagonists & inhibitors , Shaw Potassium Channels/genetics , Tissue Culture Techniques
6.
FASEB J ; 35(1): e21241, 2021 01.
Article in English | MEDLINE | ID: mdl-33368632

ABSTRACT

The voltage-gated potassium channel Kv3.4 is a crucial regulator of nociceptive signaling in the dorsal root ganglion (DRG) and the dorsal horn of the spinal cord. Moreover, Kv3.4 dysfunction has been linked to neuropathic pain. Although kinases and phosphatases can directly modulate Kv3.4 gating, the signaling mechanisms regulating the expression and stability of the Kv3.4 protein are generally unknown. We explored a potential role of PKCε and found an unexpected interaction that has a positive effect on Kv3.4 expression. Co-immunoprecipitation studies revealed a physical association between PKCε and Kv3.4 in both heterologous cells and rat DRG neurons. Furthermore, in contrast to the wild-type and constitutively active forms of PKCε, expression of a catalytically inactive form of the enzyme inhibits Kv3.4 expression and membrane localization through a dominant negative effect. Co-expression of Kv3.4 with the wild-type, constitutively active, or catalytically inactive forms of PKCε had no significant effects on Kv3.4 gating. These results suggest that a novel physical interaction of the Kv3.4 channel with functional PKCε primarily determines its stability and localization in DRG neurons. This interaction is akin to those of previously identified accessory ion channel proteins, which could be significant in neural tissues where Kv3.4 regulates electrical signaling.


Subject(s)
Ganglia, Spinal/metabolism , Gene Expression Regulation , Neurons/metabolism , Protein Kinase C-epsilon/metabolism , Shaw Potassium Channels/biosynthesis , Animals , CHO Cells , Cricetulus , HEK293 Cells , Humans , Protein Kinase C-epsilon/genetics , Rats , Shaw Potassium Channels/genetics
7.
FASEB J ; 35(12): e22053, 2021 12.
Article in English | MEDLINE | ID: mdl-34820911

ABSTRACT

Mutations in KCNC3, the gene that encodes the Kv3.3 voltage dependent potassium channel, cause Spinocerebellar Ataxia type 13 (SCA13), a disease associated with disrupted motor behaviors, progressive cerebellar degeneration, and abnormal auditory processing. The Kv3.3 channel directly binds Hax-1, a cell survival protein. A disease-causing mutation, Kv3.3-G592R, causes overstimulation of Tank Binding Kinase 1 (Tbk1) in the cerebellum, resulting in the degradation of Hax-1 by promoting its trafficking into multivesicular bodies and then to lysosomes. We have now tested the effects of antisense oligonucleotides (ASOs) directed against the Kv3.3 channel on both wild type mice and those bearing the Kv3.3-G592R-encoding mutation. Intracerebroventricular infusion of the Kcnc3-specific ASO suppressed both mRNA and protein levels of the Kv3.3 channel. In wild-type animals, this produced no change in levels of activated Tbk1, Hax-1 or Cd63, a tetraspanin marker for late endosomes/multivesicular bodies. In contrast, in mice homozygous for the Kv3.3-G592R-encoding mutation, the same ASO reduced Tbk1 activation and levels of Cd63, while restoring the expression of Hax-1 in the cerebellum. The motor behavior of the mice was tested using a rotarod assay. Surprisingly, the active ASO had no effects on the motor behavior of wild type mice but restored the behavior of the mutant mice to those of age-matched wild type animals. Our findings indicate that, in mature intact animals, suppression of Kv3.3 expression can reverse the deleterious effects of a SCA13 mutation while having little effect on wild type animals. Thus, targeting Kv3.3 expression may prove a viable therapeutic approach for SCA13.


Subject(s)
Motor Disorders/prevention & control , Mutation , Oligonucleotides, Antisense/administration & dosage , Protein Serine-Threonine Kinases/metabolism , Shaw Potassium Channels/antagonists & inhibitors , Spinocerebellar Ataxias/complications , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Disorders/etiology , Motor Disorders/metabolism , Motor Disorders/pathology , Protein Serine-Threonine Kinases/genetics , Shaw Potassium Channels/genetics , Shaw Potassium Channels/metabolism
8.
Am J Med Genet A ; 188(7): 2071-2081, 2022 07.
Article in English | MEDLINE | ID: mdl-35366058

ABSTRACT

Currently, protein-coding de novo variants and large copy number variants have been identified as important for ~30% of individuals with autism. One approach to identify relevant variation in individuals who lack these types of events is by utilizing newer genomic technologies. In this study, highly accurate PacBio HiFi long-read sequencing was applied to a family with autism, epileptic encephalopathy, cognitive impairment, and mild dysmorphic features (two affected female siblings, unaffected parents, and one unaffected male sibling) with no known clinical variant. From our long-read sequencing data, a de novo missense variant in the KCNC2 gene (encodes Kv3.2) was identified in both affected children. This variant was phased to the paternal chromosome of origin and is likely a germline mosaic. In silico assessment revealed the variant was not in controls, highly conserved, and predicted damaging. This specific missense variant (Val473Ala) has been shown in both an ortholog and paralog of Kv3.2 to accelerate current decay, shift the voltage dependence of activation, and prevent the channel from entering a long-lasting open state. Seven additional missense variants have been identified in other individuals with neurodevelopmental disorders (p = 1.03 × 10-5 ). KCNC2 is most highly expressed in the brain; in particular, in the thalamus and is enriched in GABAergic neurons. Long-read sequencing was useful in discovering the relevant variant in this family with autism that had remained a mystery for several years and will potentially have great benefits in the clinic once it is widely available.


Subject(s)
Autistic Disorder , Epilepsy , Shaw Potassium Channels , Autistic Disorder/genetics , Child , Epilepsy/genetics , Female , Germ Cells , Humans , Male , Mosaicism , Mutation, Missense , Shaw Potassium Channels/genetics
9.
Acta Neurol Scand ; 146(6): 699-707, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36225112

ABSTRACT

With the development and application of next-generation sequencing technology, the aetiological diagnosis of genetic epilepsy is rapidly becoming easier and less expensive. Additionally, there is a growing body of research into precision therapy based on genetic diagnosis. The numerous genes in the potassium ion channel family constitute the largest family of ion channels: this family is divided into different subtypes. Potassium ion channels play a crucial role in the electrical activity of neurons and are directly involved in the mechanism of epileptic seizures. In China, scientific research on genetic diagnosis and studies of precision therapy for genetic epilepsy are progressing rapidly. Many cases of epilepsy caused by mutation of potassium channel genes have been identified, and several potassium channel gene targets and drug candidates have been discovered. The purpose of this review is to briefly summarize the progress of research on the precise diagnosis and treatment of potassium ion channel-related genetic epilepsy, especially the research conducted in China. Here in, we review several large cohort studies on the genetic diagnosis of epilepsy in China in recent years, summarized the proportion of potassium channel genes. We focus on the progress of precison therapy on some hot epilepsy related potassium channel genes: KCNA1, KCNA2, KCNB1, KCNC1, KCND2, KCNQ2, KCNQ3, KCNMA1, and KCNT1.


Subject(s)
Epilepsy , Potassium Channels , Humans , Potassium Channels/genetics , KCNQ3 Potassium Channel/genetics , KCNQ2 Potassium Channel/genetics , Epilepsy/diagnosis , Epilepsy/genetics , Mutation/genetics , Shaw Potassium Channels/genetics , Potassium Channels, Sodium-Activated/genetics , Nerve Tissue Proteins/genetics
10.
J Neurophysiol ; 126(2): 532-539, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34232791

ABSTRACT

Channelopathies caused by mutations in genes encoding ion channels generally produce a clear change in channel function. Accordingly, mutations in KCNC1, which encodes the voltage-dependent Kv3.1 potassium channel, result in progressive myoclonus epilepsy as well as other developmental and epileptic encephalopathies, and these have been shown to reduce or fully abolish current amplitude. One exception to this is the mutation A513V Kv3.1b, located in the cytoplasmic C-terminal domain of the channel protein. This de novo variant was detected in a patient with epilepsy of infancy with focal migrating seizures (EIFMS), but no difference could be detected between A513V Kv3.1 current and that of wild-type Kv3.1. Using both biochemical and electrophysiological approaches, we have now confirmed that this variant produces functional channels but find that the A513V mutation renders the channel completely insensitive to regulation by phosphorylation at S503, a nearby regulatory site in the C-terminus. In this respect, the mutation resembles those in another channel, KCNT1, which are the major cause of EIFMS. Because the amplitude of Kv3.1 current is constantly adjusted by phosphorylation in vivo, our findings suggest that loss of such regulation contributes to EIFMS phenotype and emphasize the role of channel modulation for normal neuronal function.NEW & NOTEWORTHY Ion channel mutations that cause serious human diseases generally alter the biophysical properties or expression of the channel. We describe a de novo mutation in the Kv3.1 potassium channel that causes severe intellectual disability with early-onset epilepsy. The properties of this channel appear identical to those of wild-type channels, but the mutation prevents phosphorylation of the channel by protein kinase C. Our findings emphasize the role of channel modulation in normal brain function.


Subject(s)
Epilepsy/genetics , Mutation , Shaw Potassium Channels/metabolism , Sialyltransferases/deficiency , Animals , CHO Cells , Cricetinae , Cricetulus , Epilepsy/metabolism , Phosphorylation , Protein Kinase C/metabolism , Shaw Potassium Channels/chemistry , Shaw Potassium Channels/genetics , Sialyltransferases/genetics , Sialyltransferases/metabolism
11.
Clin Genet ; 99(3): 418-424, 2021 03.
Article in English | MEDLINE | ID: mdl-33349918

ABSTRACT

This study aimed to evaluate the clinical utility of whole-exome sequencing in a group of infantile-onset epilepsy patients who tested negative for epilepsy using a gene panel test. Whole-exome sequencing was performed on 59 patients who tested negative on customized epilepsy gene panel testing. We identified eight pathogenic or likely pathogenic sequence variants in eight different genes (FARS2, YWHAG, KCNC1, DYRK1A, SMC1A, PIGA, OGT, and FGF12), one pathogenic structural variant (8.6 Mb-sized deletion on chromosome X [140 994 419-149 630 805]), and three putative low-frequency mosaic variants from three different genes (GABBR2, MTOR, and CUX1). Subsequent whole-exome sequencing revealed an additional 8% of diagnostic yield with genetic confirmation of epilepsy in 55.4% (62/112) of our cohort. Three genes (YWHAG, KCNC1, and FGF12) were identified as epilepsy-causing genes after the original gene panel was designed. The others were initially linked with mitochondrial encephalopathy or different neurodevelopmental disorders, although an epilepsy phenotype was listed as one of the clinical features. Application of whole-exome sequencing following epilepsy gene panel testing provided 8% of additional diagnostic yield in an infantile-onset epilepsy cohort. Whole-exome sequencing could provide an opportunity to reanalyze newly recognized epilepsy-linked genes without updating the gene panel design.


Subject(s)
14-3-3 Proteins/genetics , Epilepsy/diagnosis , Epilepsy/genetics , Fibroblast Growth Factors/genetics , Genetic Variation , Molecular Diagnostic Techniques/methods , Shaw Potassium Channels/genetics , Age of Onset , DNA Copy Number Variations , Female , Genetic Predisposition to Disease , Genetic Testing/methods , Humans , Infant , Infant, Newborn , Male , Mitochondrial Encephalomyopathies/genetics , Mutation Rate , Neurodevelopmental Disorders/genetics , Sequence Analysis, DNA , Exome Sequencing/methods
12.
Cerebellum ; 20(1): 41-53, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32789747

ABSTRACT

Spinocerebellar ataxia type 3 (SCA3) is the second-most common CAG repeat disease, caused by a glutamine-encoding expansion in the ATXN3 protein. SCA3 is characterized by spinocerebellar degeneration leading to progressive motor incoordination and early death. Previous studies suggest that potassium channel dysfunction underlies early abnormalities in cerebellar cortical Purkinje neuron firing in SCA3. However, cerebellar cortical degeneration is often modest both in the human disease and mouse models of SCA3, raising uncertainty about the role of cerebellar dysfunction in SCA3. Here, we address this question by investigating Purkinje neuron excitability in SCA3. In early-stage SCA3 mice, we confirm a previously identified increase in excitability of cerebellar Purkinje neurons and associate this excitability with reduced transcripts of two voltage-gated potassium (KV) channels, Kcna6 and Kcnc3, as well as motor impairment. Intracerebroventricular delivery of antisense oligonucleotides (ASO) to reduce mutant ATXN3 restores normal excitability to SCA3 Purkinje neurons and rescues transcript levels of Kcna6 and Kcnc3. Interestingly, while an even broader range of KV channel transcripts shows reduced levels in late-stage SCA3 mice, cerebellar Purkinje neuron physiology was not further altered despite continued worsening of motor impairment. These results suggest the progressive motor phenotype observed in SCA3 may not reflect ongoing changes in the cerebellar cortex but instead dysfunction of other neuronal structures within and beyond the cerebellum. Nevertheless, the early rescue of both KV channel expression and neuronal excitability by ASO treatment suggests that cerebellar cortical dysfunction contributes meaningfully to motor dysfunction in SCA3.


Subject(s)
Ataxin-3/genetics , Machado-Joseph Disease/drug therapy , Machado-Joseph Disease/genetics , Oligonucleotides, Antisense/therapeutic use , Purkinje Cells/pathology , Repressor Proteins/genetics , Animals , Behavior, Animal , Humans , Injections, Intraventricular , Kv1.6 Potassium Channel/drug effects , Kv1.6 Potassium Channel/genetics , Machado-Joseph Disease/psychology , Mice , Mice, Transgenic , Patch-Clamp Techniques , Phenotype , Potassium Channels, Voltage-Gated/drug effects , Shaw Potassium Channels/drug effects , Shaw Potassium Channels/genetics , Treatment Outcome
13.
Am J Med Genet A ; 185(11): 3384-3389, 2021 11.
Article in English | MEDLINE | ID: mdl-34448338

ABSTRACT

Developmental and epileptic encephalopathies (DEE) are a heterogenous group of conditions characterized by the co-occurrence of epilepsy and intellectual/developmental disability. Despite several known DEE-related genes, including these encoding ion channels, still many cases remain without molecular diagnosis. Here, we present a 2-year-old girl with severe DEE in whom whole exome sequencing revealed de novo p.(Val471Leu) variant in the KCNC2 encoding Kv3.2, a voltage-gated potassium channel. To the best of our knowledge, this is the third DEE case due to KCNC2 mutation. Our clinical and molecular findings, particularly the recurrence of p.(Val471Leu) in patient with similar clinical phenotype, further support KCNC2 as a novel DEE-associated gene.


Subject(s)
Brain Diseases/genetics , Developmental Disabilities/genetics , Intellectual Disability/genetics , Shaw Potassium Channels/genetics , Brain Diseases/physiopathology , Child, Preschool , Developmental Disabilities/physiopathology , Epilepsy , Female , Genetic Predisposition to Disease , Humans , Intellectual Disability/physiopathology , Mutation, Missense/genetics , Phenotype , Exome Sequencing
14.
Epilepsia ; 62(5): 1256-1267, 2021 05.
Article in English | MEDLINE | ID: mdl-33735526

ABSTRACT

OBJECTIVE: Mutations in KCNC1 can cause severe neurological dysfunction, including intellectual disability, epilepsy, and ataxia. The Arg320His variant, which occurs in the voltage-sensing domain of the channel, causes a highly penetrant and specific form of progressive myoclonus epilepsy with severe ataxia, designated myoclonus epilepsy and ataxia due to potassium channel mutation (MEAK). KCNC1 encodes the voltage-gated potassium channel KV 3.1, a channel that is important for enabling high-frequency firing in interneurons, raising the possibility that MEAK is associated with reduced interneuronal function. METHODS: To determine how this variant triggers MEAK, we expressed KV 3.1bR320H in cortical interneurons in vitro and investigated the effects on neuronal function and morphology. We also performed electrophysiological recordings of oocytes expressing KV 3.1b to determine whether the mutation introduces gating pore currents. RESULTS: Expression of the KV 3.1bR320H variant profoundly reduced excitability of mature cortical interneurons, and cells expressing these channels were unable to support high-frequency firing. The mutant channel also had an unexpected effect on morphology, severely impairing neurite development and interneuron viability, an effect that could not be rescued by blocking KV 3 channels. Oocyte recordings confirmed that in the adult KV 3.1b isoform, R320H confers a dominant negative loss-of-function effect by slowing channel activation, but does not introduce potentially toxic gating pore currents. SIGNIFICANCE: Overall, our data suggest that, in addition to the regulation of high-frequency firing, KV 3.1 channels play a hitherto unrecognized role in neuronal development. MEAK may be described as a developmental dendritopathy.


Subject(s)
Dendrites/pathology , Myoclonic Epilepsies, Progressive/physiopathology , Neurogenesis/genetics , Shaw Potassium Channels/genetics , Animals , Humans , Interneurons/pathology , Mice , Mice, Inbred C57BL , Mutation , Myoclonic Epilepsies, Progressive/genetics
15.
J Neurosci ; 39(20): 3948-3969, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30862666

ABSTRACT

Purkinje cells (PCs) are primarily affected in neurodegenerative spinocerebellar ataxias (SCAs). For generating animal models for SCAs, genetic regulatory elements specifically targeting PCs are required, thereby linking pathological molecular effects with impaired function and organismic behavior. Because cerebellar anatomy and function are evolutionary conserved, zebrafish represent an excellent model to study SCAs in vivo We have isolated a 258 bp cross-species PC-specific enhancer element that can be used in a bidirectional manner for bioimaging of transgene-expressing PCs in zebrafish (both sexes) with variable copy numbers for tuning expression strength. Emerging ectopic expression at high copy numbers can be further eliminated by repurposing microRNA-mediated posttranslational mRNA regulation.Subsequently, we generated a transgenic SCA type 13 (SCA13) model, using a zebrafish-variant mimicking a human pathological SCA13R420H mutation, resulting in cell-autonomous progressive PC degeneration linked to cerebellum-driven eye-movement deficits as observed in SCA patients. This underscores that investigating PC-specific cerebellar neuropathologies in zebrafish allows for interconnecting bioimaging of disease mechanisms with behavioral analysis suitable for therapeutic compound testing.SIGNIFICANCE STATEMENT SCA13 patients carrying a KCNC3R420H allele have been shown to display mid-onset progressive cerebellar atrophy, but genetic modeling of SCA13 by expressing this pathogenic mutant in different animal models has not resulted in neuronal degeneration so far; likely because the transgene was expressed in heterologous cell types. We developed a genetic system for tunable PC-specific coexpression of several transgenes to manipulate and simultaneously monitor cerebellar PCs. We modeled a SCA13 zebrafish accessible for bioimaging to investigate disease progression, revealing robust PC degeneration, resulting in impaired eye movement. Our transgenic zebrafish mimicking both neuropathological and behavioral changes manifested in SCA-affected patients will be suitable for investigating causes of cerebellar diseases in vivo from the molecular to the behavioral level.


Subject(s)
Cerebellum/metabolism , Disease Models, Animal , Purkinje Cells/metabolism , Spinocerebellar Ataxias/congenital , Animals , Animals, Genetically Modified , Cerebellum/growth & development , Cerebellum/physiopathology , Female , Gene Expression Regulation , Male , RNA, Messenger/metabolism , Regulatory Elements, Transcriptional , Shaw Potassium Channels/genetics , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/metabolism , Zebrafish , Zebrafish Proteins/genetics
16.
Neuropediatrics ; 51(5): 368-372, 2020 10.
Article in English | MEDLINE | ID: mdl-32392612

ABSTRACT

Patients with neurofibromatosis type 1 (NF1) have an increased risk for West syndrome (WS), but the underlying mechanisms linking NF1 and WS are unknown. In contrast to other neurocutaneous syndromes, intracerebral abnormalities explaining the course of infantile spasms (IS) are often absent and the seizure outcome is usually favorable. Several studies have investigated a potential genotype-phenotype correlation between NF1 and seizure susceptibility, but an association was not identified. Therefore, we identified three patients with NF1-related WS (NF1-WS) in a cohort of 51 NF1 patients and performed whole-exome sequencing (WES) to identify genetic modifiers. In two NF1 patients with WS and good seizure outcome, we did not identify variants in epilepsy-related genes. However, in a single patient with NF1-WS and transition to drug-resistant epilepsy, we identified a de novo variant in KCNC2 (c.G499T, p.D167Y) coding for Kv3.2 as a previously undescribed potassium channel to be correlated to epilepsy. Electrophysiological studies of the identified KCNC2 variant demonstrated both a strong loss-of-function effect for the current amplitude and a gain-of-function effect for the channel activation recommending a complex network effect. These results suggest that systematic genetic analysis for potentially secondary genetic etiologies in NF1 patients and severe epilepsy presentations should be done.


Subject(s)
Neurofibromatosis 1/genetics , Shaw Potassium Channels/genetics , Spasms, Infantile/genetics , Comorbidity , Humans , Infant , Exome Sequencing
17.
Int J Mol Sci ; 21(19)2020 Sep 29.
Article in English | MEDLINE | ID: mdl-33003279

ABSTRACT

Kv3.1 channel is abundantly expressed in neurons and its dysfunction causes sleep loss, neurodegenerative diseases and depression. Fluoxetine, a serotonin selective reuptake inhibitor commonly used to treat depression, acts also on Kv3.1. To define the relationship between Kv3.1 and serotonin receptors (SR) pharmacological modulation, we showed that 1C11, a serotonergic cell line, expresses different voltage gated potassium (VGK) channels subtypes in the presence (differentiated cells (1C11D)) or absence (not differentiated cells (1C11ND)) of induction. Only Kv1.2 and Kv3.1 transcripts increase even if the level of Kv3.1b transcripts is highest in 1C11D and, after fluoxetine, in 1C11ND but decreases in 1C11D. The Kv3.1 channel protein is expressed in 1C11ND and 1C11D but is enhanced by fluoxetine only in 1C11D. Whole cell measurements confirm that 1C11 cells express (VGK) currents, increasing sequentially as a function of cell development. Moreover, SR 5HT1b is highly expressed in 1C11D but fluoxetine increases the level of transcript in 1C11ND and significantly decreases it in 1C11D. Serotonin dosage shows that fluoxetine at 10 nM blocks serotonin reuptake in 1C11ND but slows down its release when cells are differentiated through a decrease of 5HT1b receptors density. We provide the first experimental evidence that 1C11 expresses Kv3.1b, which confirms its major role during differentiation. Cells respond to the fluoxetine effect by upregulating Kv3.1b expression. On the other hand, the possible relationship between the fluoxetine effect on the kinetics of 5HT1b differentiation and Kv3.1bexpression, would suggest the Kv3.1b channel as a target of an antidepressant drug as well as it was suggested for 5HT1b.


Subject(s)
Fluoxetine/pharmacology , Serotonergic Neurons/drug effects , Serotonin Plasma Membrane Transport Proteins/genetics , Shaw Potassium Channels/genetics , Animals , CHO Cells , Cell Differentiation/drug effects , Cell Line, Tumor , Cricetulus , Depression/drug therapy , Depression/genetics , Gene Expression Regulation, Developmental/drug effects , Humans , Kv1.2 Potassium Channel/genetics , Serotonergic Neurons/metabolism , Serotonin/genetics , Serotonin/metabolism , Serotonin Antagonists/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology
18.
Am J Physiol Heart Circ Physiol ; 317(3): H496-H504, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31274353

ABSTRACT

Accumulating evidence supports that the brain renin-angiotensin system (RAS), including prorenin (PR) and its receptor (PRR), two newly discovered RAS players, contribute to sympathoexcitation in salt-sensitive hypertension. Still, whether PR also contributed to elevated circulating levels of neurohormones such as vasopressin (VP) during salt-sensitive hypertension, and if so, what are the precise underlying mechanisms, remains to be determined. To address these questions, we obtained patch-clamp recordings from hypothalamic magnocellular neurosecretory neurons (MNNs) that synthesize the neurohormones oxytocin and VP in acute hypothalamic slices obtained from sham and deoxycorticosterone acetate (DOCA)-salt-treated hypertensive rats. We found that focal application of PR markedly increased membrane excitability and firing responses in MNNs of DOCA-salt, compared with sham rats. This effect included a shorter latency to spike initiation and increased numbers of spikes in response to depolarizing stimuli and was mediated by a more robust inhibition of A-type K+ channels in DOCA-salt compared with sham rats. On the other hand, the afterhyperpolarizing potential mediated by the activation of Ca2+-dependent K+ channel was not affected by PR. mRNA expression of PRR, VP, and the Kv4.3 K+ channel subunit in the supraoptic nucleus of DOCA-salt hypertensive rats was increased compared with sham rats. Finally, we report a significant decrease of plasma VP levels in neuron-selective PRR knockdown mice treated with DOCA-salt, compared with wild-type DOCA-salt-treated mice. Together, these results support that activation of PRR contributes to increased excitability and firing discharge of MNNs and increased plasma levels of VP in DOCA-salt hypertension.NEW & NOTEWORTHY Our studies support that prorenin (PR) and its receptor (PRR) within the hypothalamus contribute to elevated plasma vasopressin levels in deoxycorticosterone acetate-salt hypertension, in part because of an exacerbated effect of PR on magnocellular neurosecretory neuron excitability; Moreover, our study implicates A-type K+ channels as key underlying molecular targets mediating these effects. Thus, PR/PRR stands as a novel therapeutic target for the treatment of neurohumoral activation in salt-sensitive hypertension.


Subject(s)
Blood Pressure , Hypertension/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Renin-Angiotensin System , Renin/metabolism , Vasopressins/blood , Animals , Desoxycorticosterone Acetate , Disease Models, Animal , Hypertension/blood , Hypertension/chemically induced , Hypertension/physiopathology , Hypothalamus/physiopathology , Male , Membrane Potentials , Mice, Knockout , Proton-Translocating ATPases/genetics , Proton-Translocating ATPases/metabolism , Rats, Wistar , Reaction Time , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Shaw Potassium Channels/genetics , Shaw Potassium Channels/metabolism , Sodium Chloride, Dietary , Time Factors , Up-Regulation
19.
Int J Mol Sci ; 20(3)2019 Feb 02.
Article in English | MEDLINE | ID: mdl-30717422

ABSTRACT

Bisoprolol (BIS) is a selective antagonist of ß1 adrenergic receptors. We examined the effects of BIS on M-type K⁺ currents (IK(M)) or erg-mediated K⁺ currents (IK(erg)) in pituitary GH3, R1220 cells, and hippocampal mHippoE-14 cells. As GH3 cells were exposed to BIS, amplitude of IK(M) was suppressed with an IC50 value of 1.21 µM. The BIS-induced suppression of IK(M) amplitude was not affected by addition of isoproterenol or ractopamine, but attenuated by flupirtine or ivabradine. In cell-attached current, BIS decreased the open probability of M-type K⁺ (KM) channels, along with decreased mean opening time of the channel. BIS decreased IK(erg) amplitude with an IC50 value of 6.42 µM. Further addition of PD-118057 attenuated BIS-mediated inhibition of IK(erg). Under current-clamp conditions, BIS depolarization increased the firing of spontaneous action potentials in GH3 cells; addition of flupirtine, but not ractopamine, reversed BIS-induced firing rate. In R1220 cells, BIS suppressed IK(M); subsequent application of ML-213(Kv7.2 channel activator) reversed BIS-induced suppression of the current. In hippocampal mHippoE-14 neurons, BIS inhibited IK(M) to a greater extent compared to its depressant effect on IK(erg). This demonstrated that in pituitary cells and hippocampal neurons the presence of BIS is capable of directly and differentially suppressing IK(M) and IK(erg), despite its antagonism of ß1-adrenergic receptors.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/pharmacology , Bisoprolol/pharmacology , Electrophysiological Phenomena/drug effects , Pituitary Gland/cytology , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Action Potentials/drug effects , Animals , KCNQ2 Potassium Channel/genetics , KCNQ2 Potassium Channel/metabolism , KCNQ3 Potassium Channel/genetics , KCNQ3 Potassium Channel/metabolism , Mice , Rats , Shaw Potassium Channels/genetics , Shaw Potassium Channels/metabolism
20.
J Cell Physiol ; 233(1): 549-558, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28322444

ABSTRACT

The potassium ion channel Kv3.1b is a member of a family of voltage-gated ion channels that are glycosylated in their mature form. In the present study, we demonstrate the impact of N-glycosylation at specific asparagine residues on the trafficking of the Kv3.1b protein. Large quantities of asparagine 229 (N229)-glycosylated Kv3.1b reached the plasma membrane, whereas N220-glycosylated and unglycosylated Kv3.1b were mainly retained in the endoplasmic reticulum (ER). These ER-retained Kv3.1b proteins were susceptible to degradation, when co-expressed with calnexin, whereas Kv3.1b pools located at the plasma membrane were resistant. Mass spectrometry analysis revealed a complex type Hex3 HexNAc4 Fuc1 glycan as the major glycan component of the N229-glycosylated Kv3.1b protein, as opposed to a high-mannose type Man8 GlcNAc2 glycan for N220-glycosylated Kv3.1b. Taken together, these results suggest that trafficking-dependent roles of the Kv3.1b potassium channel are dependent on N229 site-specific glycosylation and N-glycan structure, and operate through a mechanism whereby specific N-glycan structures regulate cell surface expression.


Subject(s)
Nerve Tissue Proteins/metabolism , Protein Processing, Post-Translational , Shaw Potassium Channels/metabolism , Animals , Asparagine , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Endoplasmic Reticulum/metabolism , Glycosylation , Mutation , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Protein Conformation , Protein Transport , Rats , Shaw Potassium Channels/chemistry , Shaw Potassium Channels/genetics , Structure-Activity Relationship , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL