Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 600
Filter
1.
Nature ; 622(7982): 410-417, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37758949

ABSTRACT

The Kv2.1 voltage-activated potassium (Kv) channel is a prominent delayed-rectifier Kv channel in the mammalian central nervous system, where its mechanisms of activation and inactivation are critical for regulating intrinsic neuronal excitability1,2. Here we present structures of the Kv2.1 channel in a lipid environment using cryo-electron microscopy to provide a framework for exploring its functional mechanisms and how mutations causing epileptic encephalopathies3-7 alter channel activity. By studying a series of disease-causing mutations, we identified one that illuminates a hydrophobic coupling nexus near the internal end of the pore that is critical for inactivation. Both functional and structural studies reveal that inactivation in Kv2.1 results from dynamic alterations in electromechanical coupling to reposition pore-lining S6 helices and close the internal pore. Consideration of these findings along with available structures for other Kv channels, as well as voltage-activated sodium and calcium channels, suggests that related mechanisms of inactivation are conserved in voltage-activated cation channels and likely to be engaged by widely used therapeutics to achieve state-dependent regulation of channel activity.


Subject(s)
Ion Channel Gating , Mutation , Shab Potassium Channels , Animals , Humans , Cryoelectron Microscopy , Hydrophobic and Hydrophilic Interactions , Ion Channel Gating/genetics , Shab Potassium Channels/genetics , Shab Potassium Channels/metabolism , Shab Potassium Channels/ultrastructure , Spasms, Infantile/genetics
2.
Am J Hum Genet ; 111(4): 761-777, 2024 04 04.
Article in English | MEDLINE | ID: mdl-38503299

ABSTRACT

Ion channels mediate voltage fluxes or action potentials that are central to the functioning of excitable cells such as neurons. The KCNB family of voltage-gated potassium channels (Kv) consists of two members (KCNB1 and KCNB2) encoded by KCNB1 and KCNB2, respectively. These channels are major contributors to delayed rectifier potassium currents arising from the neuronal soma which modulate overall excitability of neurons. In this study, we identified several mono-allelic pathogenic missense variants in KCNB2, in individuals with a neurodevelopmental syndrome with epilepsy and autism in some individuals. Recurrent dysmorphisms included a broad forehead, synophrys, and digital anomalies. Additionally, we selected three variants where genetic transmission has not been assessed, from two epilepsy studies, for inclusion in our experiments. We characterized channel properties of these variants by expressing them in oocytes of Xenopus laevis and conducting cut-open oocyte voltage clamp electrophysiology. Our datasets indicate no significant change in absolute conductance and conductance-voltage relationships of most disease variants as compared to wild type (WT), when expressed either alone or co-expressed with WT-KCNB2. However, variants c.1141A>G (p.Thr381Ala) and c.641C>T (p.Thr214Met) show complete abrogation of currents when expressed alone with the former exhibiting a left shift in activation midpoint when expressed alone or with WT-KCNB2. The variants we studied, nevertheless, show collective features of increased inactivation shifted to hyperpolarized potentials. We suggest that the effects of the variants on channel inactivation result in hyper-excitability of neurons, which contributes to disease manifestations.


Subject(s)
Epilepsy , Mutation, Missense , Neurodevelopmental Disorders , Shab Potassium Channels , Animals , Humans , Action Potentials , Epilepsy/genetics , Neurons , Oocytes , Xenopus laevis , Shab Potassium Channels/genetics , Shab Potassium Channels/metabolism , Neurodevelopmental Disorders/genetics
3.
Proc Natl Acad Sci U S A ; 119(30): e2117135119, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35862456

ABSTRACT

The endoplasmic reticulum (ER) forms a continuous and dynamic network throughout a neuron, extending from dendrites to axon terminals, and axonal ER dysfunction is implicated in several neurological disorders. In addition, tight junctions between the ER and plasma membrane (PM) are formed by several molecules including Kv2 channels, but the cellular functions of many ER-PM junctions remain unknown. Recently, dynamic Ca2+ uptake into the ER during electrical activity was shown to play an essential role in synaptic transmission. Our experiments demonstrate that Kv2.1 channels are necessary for enabling ER Ca2+ uptake during electrical activity, as knockdown (KD) of Kv2.1 rendered both the somatic and axonal ER unable to accumulate Ca2+ during electrical stimulation. Moreover, our experiments demonstrate that the loss of Kv2.1 in the axon impairs synaptic vesicle fusion during stimulation via a mechanism unrelated to voltage. Thus, our data demonstrate that a nonconducting role of Kv2.1 exists through its binding to the ER protein VAMP-associated protein (VAP), which couples ER Ca2+ uptake with electrical activity. Our results further suggest that Kv2.1 has a critical function in neuronal cell biology for Ca2+ handling independent of voltage and reveals a critical pathway for maintaining ER lumen Ca2+ levels and efficient neurotransmitter release. Taken together, these findings reveal an essential nonclassical role for both Kv2.1 and the ER-PM junctions in synaptic transmission.


Subject(s)
Endoplasmic Reticulum , Shab Potassium Channels , Calcium/metabolism , Calcium Signaling , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Neurons/metabolism , Shab Potassium Channels/metabolism , Synaptic Transmission
4.
Pharmacogenomics J ; 24(4): 22, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38992025

ABSTRACT

Bevacizumab-induced hypertension poses a therapeutic challenge and identifying biomarkers for hypertension can enhance therapy safety. Lower plasma levels of VEGF-A, angiopoietin-2, and rs6770663 in KCNAB1 were previously associated with increased risk of bevacizumab-induced hypertension. This study investigated whether these factors independently contribute to grade 2-3 bevacizumab-induced hypertension risk in 277 cancer patients (CALGB/Alliance 90401). Multivariable analyses assessed the independent association of each factor and hypertension. Likelihood ratio test (LRT) evaluated the explanatory significance of combining protein levels and rs6770663 in predicting hypertension. Boostrap was employed to assess the mediation effect of protein levels on the rs6770663 association with hypertension. Lower protein levels and rs6770663 were independently associated with increased hypertension risk. Adding rs6770663 to protein levels improved the prediction of hypertension (LRT p = 0.0002), with no mediation effect observed. Protein levels of VEGF-A, angiopoietin-2 and rs6770663 in KCNAB1 are independent risk factors and, when combined, may improve prediction of bevacizumab-induced hypertension. ClinicalTrials.gov Identifier: NCT00110214.


Subject(s)
Angiopoietin-2 , Bevacizumab , Hypertension , Vascular Endothelial Growth Factor A , Adult , Aged , Female , Humans , Male , Middle Aged , Angiogenesis Inhibitors/adverse effects , Angiopoietin-2/blood , Angiopoietin-2/genetics , Bevacizumab/adverse effects , Bevacizumab/therapeutic use , Hypertension/genetics , Hypertension/chemically induced , Hypertension/blood , Neoplasms/drug therapy , Neoplasms/blood , Neoplasms/genetics , Polymorphism, Single Nucleotide/genetics , Risk Factors , Shab Potassium Channels/genetics , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/genetics
5.
Neurocase ; 30(2): 68-72, 2024 04.
Article in English | MEDLINE | ID: mdl-38769754

ABSTRACT

KCNB1-associated encephalopathy is characterized by intellectual disability (ID), autism spectrum disorder and epilepsy. Specific treatments are still lacking. We describe a 12-year-old boy with severe ID and treatment-resistant seizures due to a pathogenic KCNB1 variant. His EEG showed a CSWS pattern. Aged 11, he started treatment with highly purified cannabidiol (CBD) and has been seizure free for 18 months, with significant EEG and social skills improvements. This suggests CBD may benefit CSWS, likely due to its anti-inflammatory properties. Some preclinical studies also indicate CBDs interact with voltage-gated channels, leading us to speculate its possible role for treating KCNB1 related encephalopathy.


Subject(s)
Cannabidiol , Electroencephalography , Child , Humans , Male , Cannabidiol/pharmacology , Epilepsy/drug therapy , Epilepsy/physiopathology , Intellectual Disability/drug therapy , Intellectual Disability/complications , Shab Potassium Channels/genetics
6.
Proc Natl Acad Sci U S A ; 118(46)2021 11 16.
Article in English | MEDLINE | ID: mdl-34750263

ABSTRACT

In mammalian brain neurons, membrane depolarization leads to voltage-gated Ca2+ channel-mediated Ca2+ influx that triggers diverse cellular responses, including gene expression, in a process termed excitation-transcription coupling. Neuronal L-type Ca2+ channels, which have prominent populations on the soma and distal dendrites of hippocampal neurons, play a privileged role in excitation-transcription coupling. The voltage-gated K+ channel Kv2.1 organizes signaling complexes containing the L-type Ca2+ channel Cav1.2 at somatic endoplasmic reticulum-plasma membrane junctions. This leads to enhanced clustering of Cav1.2 channels, increasing their activity. However, the downstream consequences of the Kv2.1-mediated regulation of Cav1.2 localization and function on excitation-transcription coupling are not known. Here, we have identified a region between residues 478 to 486 of Kv2.1's C terminus that mediates the Kv2.1-dependent clustering of Cav1.2. By disrupting this Ca2+ channel association domain with either mutations or with a cell-penetrating interfering peptide, we blocked the Kv2.1-mediated clustering of Cav1.2 at endoplasmic reticulum-plasma membrane junctions and the subsequent enhancement of its channel activity and somatic Ca2+ signals without affecting the clustering of Kv2.1. These interventions abolished the depolarization-induced and L-type Ca2+ channel-dependent phosphorylation of the transcription factor CREB and the subsequent expression of c-Fos in hippocampal neurons. Our findings support a model whereby the Kv2.1-Ca2+ channel association domain-mediated clustering of Cav1.2 channels imparts a mechanism to control somatic Ca2+ signals that couple neuronal excitation to gene expression.


Subject(s)
Calcium Channels, L-Type/genetics , Cell Membrane/genetics , Endoplasmic Reticulum/genetics , Neurons/physiology , Shab Potassium Channels/genetics , Transcription, Genetic/genetics , Animals , Cells, Cultured , Dendrites/genetics , Female , HEK293 Cells , Hippocampus/physiology , Humans , Male , Mice , Phosphorylation/genetics , Rats
7.
Int J Mol Sci ; 25(16)2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39201645

ABSTRACT

Migraines are a common type of headache affecting around 15% of the population. The signalling pathways leading to migraines have not been fully understood, but neuronal voltage-gated ion channels, such as KCNG4, have been linked to this pathology. KCNG4 (Kv6.4) is a silent member of the superfamily of voltage-gated potassium (Kv) channels, which expresses in heterotetramers with members of the KCNB (Kv2) family. The genetic variant Kv6.4-L360P has previously been linked to migraines, but their mode of action remains unknown. Here, we characterized the molecular characteristics of Kv6.4-L360P when co-expressed with Kv2.1. We found that Kv6.4-L360P almost completely abolishes Kv2 currents, and we propose that this mechanism in the trigeminal system, linked to the initiation of migraine, leads to the pathology.


Subject(s)
Migraine Disorders , Potassium Channels, Voltage-Gated , Shab Potassium Channels , Animals , Humans , Genetic Variation , HEK293 Cells , Migraine Disorders/genetics , Migraine Disorders/metabolism , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Shab Potassium Channels/genetics , Shab Potassium Channels/metabolism
8.
Am J Hum Genet ; 107(4): 683-697, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32853554

ABSTRACT

More than 100 genetic etiologies have been identified in developmental and epileptic encephalopathies (DEEs), but correlating genetic findings with clinical features at scale has remained a hurdle because of a lack of frameworks for analyzing heterogenous clinical data. Here, we analyzed 31,742 Human Phenotype Ontology (HPO) terms in 846 individuals with existing whole-exome trio data and assessed associated clinical features and phenotypic relatedness by using HPO-based semantic similarity analysis for individuals with de novo variants in the same gene. Gene-specific phenotypic signatures included associations of SCN1A with "complex febrile seizures" (HP: 0011172; p = 2.1 × 10-5) and "focal clonic seizures" (HP: 0002266; p = 8.9 × 10-6), STXBP1 with "absent speech" (HP: 0001344; p = 1.3 × 10-11), and SLC6A1 with "EEG with generalized slow activity" (HP: 0010845; p = 0.018). Of 41 genes with de novo variants in two or more individuals, 11 genes showed significant phenotypic similarity, including SCN1A (n = 16, p < 0.0001), STXBP1 (n = 14, p = 0.0021), and KCNB1 (n = 6, p = 0.011). Including genetic and phenotypic data of control subjects increased phenotypic similarity for all genetic etiologies, whereas the probability of observing de novo variants decreased, emphasizing the conceptual differences between semantic similarity analysis and approaches based on the expected number of de novo events. We demonstrate that HPO-based phenotype analysis captures unique profiles for distinct genetic etiologies, reflecting the breadth of the phenotypic spectrum in genetic epilepsies. Semantic similarity can be used to generate statistical evidence for disease causation analogous to the traditional approach of primarily defining disease entities through similar clinical features.


Subject(s)
GABA Plasma Membrane Transport Proteins/genetics , Munc18 Proteins/genetics , NAV1.1 Voltage-Gated Sodium Channel/genetics , Seizures/genetics , Spasms, Infantile/genetics , Speech Disorders/genetics , Child, Preschool , Cohort Studies , Female , Gene Expression , Gene Ontology , Humans , Male , Mutation , Phenotype , Seizures/classification , Seizures/diagnosis , Seizures/physiopathology , Semantics , Shab Potassium Channels/genetics , Spasms, Infantile/classification , Spasms, Infantile/diagnosis , Spasms, Infantile/physiopathology , Speech Disorders/classification , Speech Disorders/diagnosis , Speech Disorders/physiopathology , Terminology as Topic , Exome Sequencing
9.
J Cell Sci ; 134(12)2021 06 15.
Article in English | MEDLINE | ID: mdl-34137443

ABSTRACT

The Kv2 channels encode delayed rectifier currents that regulate membrane potential in many tissues. They also have a non-conducting function to form stable junctions between the endoplasmic reticulum and plasma membranes, creating membrane contact sites that mediate functions distinct from membrane excitability. Therefore, proteins that interact with Kv2.1 and Kv2.2 channels can alter conducting and/or non-conducting channel properties. One member of the AMIGO family of proteins is an auxiliary ß-subunit for Kv2 channels and modulates Kv2.1 electrical activity. However, the AMIGO family has two additional members of ∼50% similarity that have not yet been characterized as Kv2 ß-subunits. In this work, we show that the surface trafficking and localization of all three AMIGOs are controlled by their assembly with both Kv2 channels. Additionally, assembly of each AMIGO with either Kv2.1 or Kv2.2 hyperpolarizes the channel activation midpoint by -10 mV. However, only AMIGO2 significantly slows inactivation and deactivation, leading to a prolonged open state of Kv2 channels. The co-regulatory effects of Kv2s and AMIGOs likely fine-tune both the electrical and non-electrical properties of the cells in which they are expressed.


Subject(s)
Neurons , Shab Potassium Channels , Cell Adhesion Molecules , HEK293 Cells , Hippocampus/metabolism , Humans , Membrane Glycoproteins , Nerve Tissue Proteins , Neurons/metabolism , Shab Potassium Channels/genetics , Shab Potassium Channels/metabolism
10.
J Hum Genet ; 68(1): 25-31, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36257979

ABSTRACT

KCNB1 encodes the α-subunit of Kv2.1, the main contributor to neuronal delayed rectifier potassium currents. The subunit consists of six transmembrane α helices (S1-S6), comprising the voltage-sensing domain (S1-S4) and the pore domain (S5-P-S6). Heterozygous KCNB1 pathogenic variants are associated with developmental and epileptic encephalopathy. Here we report an individual who shows the milder phenotype compared to the previously reported cases, including delayed language development, mild intellectual disability, attention deficit hyperactivity disorder, late-onset epilepsy responsive to an antiepileptic drug, elevation of serum creatine kinase, and peripheral axonal neuropathy. On the other hand, his brain MRI showed characteristic findings including periventricular heterotopia, polymicrogyria, and abnormal corpus callosum. Exome sequencing identified a novel de novo KCNB1 variant c.574G>A, p.(Ala192Thr) located in the S1 segment of the voltage-sensing domain. Functional analysis using the whole-cell patch-clamp technique in Neuro2a cells showed that the Ala192Thr mutant reduces both activation and inactivation of the channel at membrane voltages in the range of -50 to -30 mV. Our case could expand the phenotypic spectrum of patients with KCNB1 variants, and suggested that variants located in the S1 segment might be associated with a milder outcome of seizures.


Subject(s)
Periventricular Nodular Heterotopia , Shab Potassium Channels , Humans , Corpus Callosum/diagnostic imaging , Corpus Callosum/pathology , Epilepsy/etiology , Epilepsy/genetics , Periventricular Nodular Heterotopia/genetics , Phenotype , Seizures/etiology , Seizures/genetics , Shab Potassium Channels/genetics
11.
Cereb Cortex ; 32(5): 1055-1076, 2022 02 19.
Article in English | MEDLINE | ID: mdl-34435615

ABSTRACT

Coincidence detection and cortical rhythmicity are both greatly influenced by neurons' propensity to fire bursts of action potentials. In the neocortex, repetitive burst firing can also initiate abnormal neocortical rhythmicity (including epilepsy). Bursts are generated by inward currents that underlie a fast afterdepolarization (fADP) but less is known about outward currents that regulate bursting. We tested whether Kv2 channels regulate the fADP and burst firing in labeled layer 5 PNs from motor cortex of the Thy1-h mouse. Kv2 block with guangxitoxin-1E (GTx) converted single spike responses evoked by dendritic stimulation into multispike bursts riding on an enhanced fADP. Immunohistochemistry revealed that Thy1-h PNs expressed Kv2.1 (not Kv2.2) channels perisomatically (not in the dendrites). In somatic macropatches, GTx-sensitive current was the largest component of outward current with biophysical properties well-suited for regulating bursting. GTx drove ~40% of Thy1 PNs stimulated with noisy somatic current steps to repetitive burst firing and shifted the maximal frequency-dependent gain. A network model showed that reduction of Kv2-like conductance in a small subset of neurons resulted in repetitive bursting and entrainment of the circuit to seizure-like rhythmic activity. Kv2 channels play a dominant role in regulating onset bursts and preventing repetitive bursting in Thy1 PNs.


Subject(s)
Neocortex , Shab Potassium Channels , Action Potentials/physiology , Animals , Mice , Neocortex/metabolism , Neurons/physiology , Pyramidal Cells/physiology , Shab Potassium Channels/metabolism
12.
Proc Natl Acad Sci U S A ; 117(17): 9365-9376, 2020 04 28.
Article in English | MEDLINE | ID: mdl-32284408

ABSTRACT

The electrically silent (KvS) members of the voltage-gated potassium (Kv) subfamilies Kv5, Kv6, Kv8, and Kv9 selectively modulate Kv2 subunits by forming heterotetrameric Kv2/KvS channels. Based on the reported 3:1 stoichiometry of Kv2.1/Kv9.3 channels, we tested the hypothesis that Kv2.1/Kv6.4 channels express, in contrast to the assumed 3:1, in a 2:2 stoichiometry. We investigate the Kv2.1/Kv6.4 stoichiometry using single subunit counting and functional characterization of tetrameric concatemers. For selecting the most probable stoichiometry, we introduce a model-selection method that is applicable for any multimeric complex by investigating the stoichiometry of Kv2.1/Kv6.4 channels. Weighted likelihood calculations bring rigor to a powerful technique. Using the weighted-likelihood model-selection method and analysis of electrophysiological data, we show that Kv2.1/Kv6.4 channels express, in contrast to the assumed 3:1, in a 2:2 stoichiometry. Within this stoichiometry, the Kv6.4 subunits have to be positioned alternating with Kv2.1 to express functional channels. The variability in Kv2/KvS assembly increases the diversity of heterotetrameric configurations and extends the regulatory possibilities of KvS by allowing the presence of more than one silent subunit.


Subject(s)
Potassium Channels, Voltage-Gated/metabolism , Potassium/metabolism , Shab Potassium Channels/metabolism , Animals , Antibodies , Cell Line , Fibroblasts , Gene Expression Regulation , HEK293 Cells , Humans , Membrane Potentials , Mice , Oocytes/metabolism , Photobleaching , Potassium Channels, Voltage-Gated/genetics , Receptor Protein-Tyrosine Kinases/genetics , Recombinant Proteins , Shab Potassium Channels/genetics , Shab Potassium Channels/immunology , Xenopus
13.
Proc Natl Acad Sci U S A ; 117(7): 3858-3866, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32015129

ABSTRACT

The accepted role of the protein Kv2.1 in arterial smooth muscle cells is to form K+ channels in the sarcolemma. Opening of Kv2.1 channels causes membrane hyperpolarization, which decreases the activity of L-type CaV1.2 channels, lowering intracellular Ca2+ ([Ca2+]i) and causing smooth muscle relaxation. A limitation of this model is that it is based exclusively on data from male arterial myocytes. Here, we used a combination of electrophysiology as well as imaging approaches to investigate the role of Kv2.1 channels in male and female arterial myocytes. We confirmed that Kv2.1 plays a canonical conductive role but found it also has a structural role in arterial myocytes to enhance clustering of CaV1.2 channels. Less than 1% of Kv2.1 channels are conductive and induce membrane hyperpolarization. Paradoxically, by enhancing the structural clustering and probability of CaV1.2-CaV1.2 interactions within these clusters, Kv2.1 increases Ca2+ influx. These functional impacts of Kv2.1 depend on its level of expression, which varies with sex. In female myocytes, where expression of Kv2.1 protein is higher than in male myocytes, Kv2.1 has conductive and structural roles. Female myocytes have larger CaV1.2 clusters, larger [Ca2+]i, and larger myogenic tone than male myocytes. In contrast, in male myocytes, Kv2.1 channels regulate membrane potential but not CaV1.2 channel clustering. We propose a model in which Kv2.1 function varies with sex: in males, Kv2.1 channels control membrane potential but, in female myocytes, Kv2.1 plays dual electrical and CaV1.2 clustering roles. This contributes to sex-specific regulation of excitability, [Ca2+]i, and myogenic tone in arterial myocytes.


Subject(s)
Arteries/metabolism , Calcium Channels, L-Type/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Shab Potassium Channels/metabolism , Animals , Calcium/metabolism , Calcium Channels, L-Type/genetics , Cells, Cultured , Female , Male , Membrane Potentials , Mice, Inbred C57BL , Mice, Knockout , Shab Potassium Channels/genetics
14.
Biophys J ; 121(8): 1395-1416, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35314141

ABSTRACT

Kv2 voltage-gated potassium channels are modulated by amphoterin-induced gene and open reading frame (AMIGO) neuronal adhesion proteins. Here, we identify steps in the conductance activation pathway of Kv2.1 channels that are modulated by AMIGO1 using voltage-clamp recordings and spectroscopy of heterologously expressed Kv2.1 and AMIGO1 in mammalian cell lines. AMIGO1 speeds early voltage-sensor movements and shifts the gating charge-voltage relationship to more negative voltages. The gating charge-voltage relationship indicates that AMIGO1 exerts a larger energetic effect on voltage-sensor movement than is apparent from the midpoint of the conductance-voltage relationship. When voltage sensors are detained at rest by voltage-sensor toxins, AMIGO1 has a greater impact on the conductance-voltage relationship. Fluorescence measurements from voltage-sensor toxins bound to Kv2.1 indicate that with AMIGO1, the voltage sensors enter their earliest resting conformation, yet this conformation is less stable upon voltage stimulation. We conclude that AMIGO1 modulates the Kv2.1 conductance activation pathway by destabilizing the earliest resting state of the voltage sensors.


Subject(s)
Ion Channel Gating , Shab Potassium Channels , Animals , Cell Line , Mammals/metabolism , Neurons/metabolism , Shab Potassium Channels/metabolism
15.
Biophys J ; 121(5): 755-768, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35101417

ABSTRACT

Ion channels are well known for their ability to regulate the cell membrane potential. However, many ion channels also have functions that do not involve ion conductance. Kv2 channels are one family of ion channels whose non-conducting functions are central to mammalian cell physiology. Kv2.1 and Kv2.2 channels form stable contact sites between the endoplasmic reticulum and plasma membrane via an interaction with endoplasmic reticulum resident proteins. To perform this structural role, Kv2 channels are expressed at extremely high densities on the plasma membranes of many cell types, including central pyramidal neurons, α-motoneurons, and smooth muscle cells. Research from our lab and others has shown that the majority of these plasma membrane Kv2.1 channels do not conduct potassium in response to depolarization. The mechanism of this channel silencing is unknown but is thought to be dependent on channel density in the membrane. Furthermore, the prevalence of a non-conducting population of Kv2.2 channels has not been directly tested. In this work we make improved measurements of the numbers of conducting and non-conducting Kv2.1 channels expressed in HEK293 cells and expand the investigation of non-conducting channels to three additional Kv α-subunits: Kv2.2, Kv1.4, and Kv1.5. By comparing the numbers of gating and conducting channels in individual HEK293 cells, we found that on average, only 50% of both Kv2.1 and Kv2.2 channels conducted potassium and, as previously suggested, that fraction decreased with increased channel density in the plasma membrane. At the highest spatial densities tested, which are comparable with those found at Kv2 clusters in situ, only 20% of Kv2.1 and Kv2.2 channels conducted potassium. We also show for the first time that Kv1.4 and Kv1.5 exhibit density-dependent silencing, suggesting that this phenomenon has an underlying mechanism that is shared by Kv channels from multiple families.


Subject(s)
Myocytes, Smooth Muscle , Shab Potassium Channels , Animals , Cell Membrane/metabolism , HEK293 Cells , Humans , Mammals/metabolism , Potassium/metabolism , Shab Potassium Channels/metabolism
16.
Epilepsy Behav ; 126: 108471, 2022 01.
Article in English | MEDLINE | ID: mdl-34915430

ABSTRACT

AIM: KCNB1 encephalopathy encompasses a broad phenotypic spectrum associating intellectual disability, behavioral disturbances, and epilepsies of various severity. Using standardized parental questionnaires, we aimed to capture the heterogeneity of the adaptive and behavioral features in a series of patients with KCNB1 pathogenic variants. METHODS: We included 25 patients with a KCNB1 encephalopathy, aged from 3.2 to 34.1 years (median = 10 years). Adaptive functioning was assessed in all patients using the French version of the Vineland Adaptive Behavior Scales, Second Edition (VABS-II) questionnaire. We screened global behavior with the Childhood Behavioral Check-List (CBCL, Achenbach) and autism spectrum disorder (ASD) with the Social Communication Questionnaire (SCQ). We used a cluster analysis to identify subgroups of adaptive profiles. RESULTS: VABS-II questionnaire showed pathological adaptive behavior in all participants with a severity of adaptive deficiency ranging from mild in 8/20 to severe in 7/20. Eight out of 16 were at risk of Attention Problems at the CBCL and 13/18 were at risk of autism spectrum disorder (ASD). The adaptive behavior composite score significantly decreased with age (Spearman's Rho=-0.72, p<0.001) but not the equivalent ages, suggesting stagnation and slowing but no regression over time. The clustering analysis identified two subgroups of patients, one showing more severe adaptive behavior. The severity of the epilepsy phenotype predicted the severity of the behavioral profile with a sensitivity of 70% and a specificity of 90.9%. CONCLUSION: This study confirms the deleterious consequences of early-onset epilepsy in addition to the impact of the gene dysfunction in patients with KCNB1 encephalopathy. ASD and attention disorders are frequent. Parental questionnaires should be considered as useful tools for early screening and care adaptation.


Subject(s)
Autism Spectrum Disorder , Brain Diseases , Epilepsy , Intellectual Disability , Adaptation, Psychological , Adolescent , Adult , Autism Spectrum Disorder/complications , Autism Spectrum Disorder/epidemiology , Autism Spectrum Disorder/genetics , Brain Diseases/complications , Brain Diseases/epidemiology , Brain Diseases/genetics , Child , Child, Preschool , Epilepsy/genetics , Humans , Intellectual Disability/epidemiology , Intellectual Disability/genetics , Intellectual Disability/psychology , Shab Potassium Channels/genetics , Young Adult
17.
Proc Natl Acad Sci U S A ; 116(31): 15696-15705, 2019 07 30.
Article in English | MEDLINE | ID: mdl-31308225

ABSTRACT

The neuronal cell death-promoting loss of cytoplasmic K+ following injury is mediated by an increase in Kv2.1 potassium channels in the plasma membrane. This phenomenon relies on Kv2.1 binding to syntaxin 1A via 9 amino acids within the channel intrinsically disordered C terminus. Preventing this interaction with a cell and blood-brain barrier-permeant peptide is neuroprotective in an in vivo stroke model. Here a rational approach was applied to define the key molecular interactions between syntaxin and Kv2.1, some of which are shared with mammalian uncoordinated-18 (munc18). Armed with this information, we found a small molecule Kv2.1-syntaxin-binding inhibitor (cpd5) that improves cortical neuron survival by suppressing SNARE-dependent enhancement of Kv2.1-mediated currents following excitotoxic injury. We validated that cpd5 selectively displaces Kv2.1-syntaxin-binding peptides from syntaxin and, at higher concentrations, munc18, but without affecting either synaptic or neuronal intrinsic properties in brain tissue slices at neuroprotective concentrations. Collectively, our findings provide insight into the role of syntaxin in neuronal cell death and validate an important target for neuroprotection.


Subject(s)
Brain/metabolism , Neuroprotective Agents , Shab Potassium Channels/metabolism , Syntaxin 1/metabolism , Animals , Munc18 Proteins/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Rats , SNARE Proteins/metabolism
18.
J Neurosci ; 40(45): 8698-8714, 2020 11 04.
Article in English | MEDLINE | ID: mdl-33046554

ABSTRACT

The formation of memory for a novel experience is a critical cognitive capacity. The ability to form novel memories is sensitive to age-related pathologies and disease, to which prolonged metabolic stress is a major contributing factor. Presently, we describe a dopamine-dependent redox modulation pathway within the hippocampus of male mice that promotes memory consolidation. Namely, following novel information acquisition, quinone reductase 2 (QR2) is suppressed by miRNA-182 (miR-182) in the CA1 region of the hippocampus via dopamine D1 receptor (D1R) activation, a process largely facilitated by locus coeruleus activity. This pathway activation reduces ROS generated by QR2 enzymatic activity, a process that alters the intrinsic properties of CA1 interneurons 3 h following learning, in a form of oxidative eustress. Interestingly, novel experience decreases QR2 expression predominately in inhibitory interneurons. Additionally, we find that in aged animals this newly described QR2 pathway is chronically under activated, resulting in miR-182 underexpression and QR2 overexpression. This leads to accumulative oxidative stress, which can be seen in CA1 via increased levels of oxidized, inactivated potassium channel Kv2.1, which undergoes disulfide bridge oligomerization. This newly described interneuron-specific molecular pathway lies alongside the known mRNA translation-dependent processes necessary for long-term memory formation, entrained by dopamine in CA1. It is a process crucial for the distinguishing features of novel memory, and points to a promising new target for memory enhancement in aging and age-dependent diseases.SIGNIFICANCE STATEMENT One way in which evolution dictates which sensory information will stabilize as an internal representation, relies on information novelty. Dopamine is a central neuromodulator involved in this process in the mammalian hippocampus. Here, we describe for the first time a dopamine D1 receptor-dependent quinone reductase 2 pathway in interneurons. This is a targeted redox event necessary to delineate a novel experience to a robust long-term internal representation. Activation of this pathway alone can explain the effect novelty has on "flashbulb" memories, and it can become dysfunctional with age and diseases, such as Alzheimer's disease.


Subject(s)
CA1 Region, Hippocampal/physiology , Dopamine/physiology , Interneurons/physiology , Memory/physiology , Quinone Reductases/physiology , Signal Transduction/physiology , Aging/physiology , Aging/psychology , Animals , CA1 Region, Hippocampal/growth & development , Dopamine Antagonists/pharmacology , Fear/psychology , Male , Memory Consolidation/physiology , Memory, Long-Term , Mice , Mice, Inbred C57BL , MicroRNAs/biosynthesis , MicroRNAs/genetics , Oxidative Stress , Reactive Oxygen Species/metabolism , Recognition, Psychology , Shab Potassium Channels/metabolism
19.
J Physiol ; 599(5): 1391-1420, 2021 03.
Article in English | MEDLINE | ID: mdl-33449375

ABSTRACT

The spatial and temporal balance of spinal α-motoneuron (αMN) intrinsic membrane conductances underlies the neural output of the final common pathway for motor commands. Although the complete set and precise localization of αMN K+ channels and their respective outward conductances remain unsettled, important K+ channel subtypes have now been documented, including Kv1, Kv2, Kv7, TASK, HCN and SK isoforms. Unique kinetics and gating parameters allow these channels to differentially shape and/or modify αMN firing properties, and recent immunohistochemical localization of K+ -channel complexes reveals a framework in which their spatial distribution and/or focal clustering within different surface membrane compartments is highly tuned to their physiological function. Moreover, highly evolved regulatory mechanisms enable specific channels to operate over variable levels of αMN activity and contribute to either state-dependent enhancement or diminution of firing. While recent data suggest an additional, non-conducting role for clustered Kv2.1 channels in the formation of endoplasmic reticulum-plasma membrane junctions postsynaptic to C-bouton synapses, electrophysiological evidence demonstrates that conducting Kv2.1 channels effectively regulate αMN firing, especially during periods of high activity in which the cholinergic C-boutons are engaged. Intense αMN activity or cell injury rapidly disrupts the clustered organization of Kv2.1 channels in αMNs and further impacts their physiological role. Thus, αMN K+ channels play a critical regulatory role in motor processing and are potential therapeutic targets for diseases affecting αMN excitability and motor output, including amyotrophic lateral sclerosis.


Subject(s)
Motor Neurons , Shab Potassium Channels , Animals , Electrophysiological Phenomena , Mammals , Synapses
20.
J Biol Chem ; 295(50): 17337-17348, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33060203

ABSTRACT

Transient receptor potential vanilloid 1 (TRPV1) channel is a multimodal receptor that is responsible for nociceptive, thermal, and mechanical sensations. However, which biomolecular partners specifically interact with TRPV1 remains to be elucidated. Here, we used cDNA library screening of genes from mouse dorsal root ganglia combined with patch-clamp electrophysiology to identify the voltage-gated potassium channel auxiliary subunit Kvß1 physically interacting with TRPV1 channel and regulating its function. The interaction was validated in situ using endogenous dorsal root ganglia neurons, as well as a recombinant expression model in HEK 293T cells. The presence of Kvß1 enhanced the expression stability of TRPV1 channels on the plasma membrane and the nociceptive current density. Surprisingly, Kvß1 interaction also shifted the temperature threshold for TRPV1 thermal activation. Using site-specific mapping, we further revealed that Kvß1 interacted with the membrane-distal domain and membrane-proximal domain of TRPV1 to regulate its membrane expression and temperature-activation threshold, respectively. Our data therefore suggest that Kvß1 is a key element in the TRPV1 signaling complex and exerts dual regulatory effects in a site-specific manner.


Subject(s)
Ganglia, Spinal/metabolism , Neurons/metabolism , Shab Potassium Channels/metabolism , Signal Transduction , TRPV Cation Channels/metabolism , Animals , Cell Membrane/genetics , Cell Membrane/metabolism , HEK293 Cells , Humans , Mice , Protein Subunits/genetics , Protein Subunits/metabolism , Rats , Shab Potassium Channels/genetics , TRPV Cation Channels/genetics
SELECTION OF CITATIONS
SEARCH DETAIL