Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 4087, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35840580

ABSTRACT

Kv3 channels have distinctive gating kinetics tailored for rapid repolarization in fast-spiking neurons. Malfunction of this process due to genetic variants in the KCNC1 gene causes severe epileptic disorders, yet the structural determinants for the unusual gating properties remain elusive. Here, we present cryo-electron microscopy structures of the human Kv3.1a channel, revealing a unique arrangement of the cytoplasmic tetramerization domain T1 which facilitates interactions with C-terminal axonal targeting motif and key components of the gating machinery. Additional interactions between S1/S2 linker and turret domain strengthen the interface between voltage sensor and pore domain. Supported by molecular dynamics simulations, electrophysiological and mutational analyses, we identify several residues in the S4/S5 linker which influence the gating kinetics and an electrostatic interaction between acidic residues in α6 of T1 and R449 in the pore-flanking S6T helices. These findings provide insights into gating control and disease mechanisms and may guide strategies for the design of pharmaceutical drugs targeting Kv3 channels.


Subject(s)
Ion Channel Gating , Shaw Potassium Channels , Cryoelectron Microscopy , Humans , Molecular Dynamics Simulation , Protein Structure, Secondary , Shaw Potassium Channels/chemistry , Shaw Potassium Channels/genetics , Shaw Potassium Channels/metabolism , Static Electricity
2.
Int J Mol Sci ; 22(22)2021 Nov 13.
Article in English | MEDLINE | ID: mdl-34830172

ABSTRACT

The voltage-gated K+ channels Kv3.1 display fast activation and deactivation kinetics and are known to have a crucial contribution to the fast-spiking phenotype of certain neurons. AahG50, as a natural product extracted from Androctonus australis hector venom, inhibits selectively Kv3.1 channels. In the present study, we focused on the biochemical and pharmacological characterization of the component in AahG50 scorpion venom that potently and selectively blocks the Kv3.1 channels. We used a combined optimization through advanced biochemical purification and patch-clamp screening steps to characterize the peptide in AahG50 active on Kv3.1 channels. We described the inhibitory effect of a toxin on Kv3.1 unitary current in black lipid bilayers. In silico, docking experiments are used to study the molecular details of the binding. We identified the first scorpion venom peptide inhibiting Kv3.1 current at 170 nM. This toxin is the alpha-KTx 15.1, which occludes the Kv3.1 channel pore by means of the lysine 27 lateral chain. This study highlights, for the first time, the modulation of the Kv3.1 by alpha-KTx 15.1, which could be an interesting starting compound for developing therapeutic biomolecules against Kv3.1-associated diseases.


Subject(s)
Molecular Docking Simulation , Potassium Channel Blockers/chemistry , Scorpion Venoms/chemistry , Shaw Potassium Channels , Animals , Humans , Scorpions/chemistry , Shaw Potassium Channels/antagonists & inhibitors , Shaw Potassium Channels/chemistry , Xenopus laevis
3.
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
4.
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
5.
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
6.
PLoS One ; 8(9): e75013, 2013.
Article in English | MEDLINE | ID: mdl-24040379

ABSTRACT

Glycoconjugates at the cell surface are crucial for cells to communicate with each other and the extracellular microenvironment. While it is generally accepted that glycans are vectorial biopolymers, their information content is unclear. This report provides evidence that distinct N-glycan structures influence the spatial arrangement of two integral membrane glycoproteins, Kv3.1 and E-cadherin, at the adherent membrane which in turn alter cellular properties. Distinct N-glycan structures were generated by heterologous expression of these glycoproteins in parental and glycosylation mutant Chinese hamster ovary cell lines. Unlike the N-linked glycans, the O-linked glycans of the mutant cell lines are similar to those of the parental cell line. Western and lectin blots of total membranes and GFP immunopurified samples, combined with glycosidase digestion reactions, were employed to verify the glycoproteins had predominantly complex, oligomannose, and bisecting type N-glycans from Pro(-)5, Lec1, and Lec10B cell lines, respectively. Based on total internal reflection fluorescence and differential interference contrast microscopy techniques, and cellular assays of live parental and glycosylation mutant CHO cells, we propose that glycoproteins with complex, oligomannose or bisecting type N-glycans relay information for localization of glycoproteins to various regions of the plasma membrane in both a glycan-specific and protein-specific manner, and furthermore cell-cell interactions are required for deciphering much of this information. These distinct spatial arrangements also impact cell adhesion and migration. Our findings provide direct evidence that N-glycan structures of glycoproteins contribute significantly to the information content of cells.


Subject(s)
Cell Membrane/chemistry , Glycoproteins/chemistry , Polysaccharides/chemistry , Animals , CHO Cells , Cadherins/chemistry , Cell Adhesion , Cell Movement , Cricetinae , Cricetulus , Genetic Vectors , Glycosylation , Green Fluorescent Proteins/chemistry , Lectins/chemistry , Microscopy, Fluorescence , Mutation , Polymers/chemistry , Shaw Potassium Channels/chemistry
7.
Biochim Biophys Acta ; 1828(2): 595-601, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23031574

ABSTRACT

The modulation of the Drosophila Shaw2 Kv channel by 1-alkanols and inhaled anesthetics is correlated with the involvement of the S4-S5 linker and C-terminus of S6, and consistent with stabilization of the channel's closed state. Structural analysis of peptides from S4-S5 (L45) and S6 (S6c), by nuclear magnetic resonance and circular dichroism spectroscopy supports that an α-helical conformation was adopted by L45, while S6c was only in an unstable/dynamic partially folded α-helix in dodecylphosphocholine micelles. Solvent accessibility and paramagnetic probing of L45 revealed that L45 lies parallel to the surface of micelles with charged and polar residues pointing towards the solution while hydrophobic residues are buried inside the micelles. Chemical shift perturbation introduced by 1-butanol on residues Gln320, Thr321, Phe322 and Arg323 of L45, as well as Thr423 and Gln424 of S6c indicates possible anesthetic binding sites on these two important components in the channel activation apparatus. Diffusion measurements confirmed the association of L45, S6c and 1-butanol with micelles which suggests the capability of 1-butanol to influence a possible interaction of L45 and S6c in the micelle environment.


Subject(s)
Anesthetics, General/chemistry , Drosophila Proteins/chemistry , Magnetic Resonance Spectroscopy/methods , Micelles , Peptides/chemistry , Shaw Potassium Channels/chemistry , 1-Butanol/chemistry , Animals , Binding Sites , Cell Membrane/metabolism , Circular Dichroism/methods , Diffusion , Drosophila melanogaster , Imaging, Three-Dimensional/methods , Kinetics , Models, Statistical , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/chemistry , Protein Binding , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Solvents/chemistry
8.
PLoS One ; 7(11): e49070, 2012.
Article in English | MEDLINE | ID: mdl-23145073

ABSTRACT

Recent studies have explored the utility of Fourier transform infrared spectroscopy (FTIR) in dynamic monitoring of soluble protein-protein interactions. Here, we investigated the applicability of FTIR to detect interaction between synthetic soluble and phospholipid-embedded peptides corresponding to, respectively, a voltage-gated potassium (Kv) channel inactivation domain (ID) and S4-S6 of the Shaker Kv channel (KV1; including the S4-S5 linker "pre-inactivation" ID binding site). KV1 was predominantly α-helical at 30°C when incorporated into dimyristoyl-l-α-phosphatidylcholine (DMPC) bilayers. Cooling to induce a shift in DMPC from liquid crystalline to gel phase reversibly decreased KV1 helicity, and was previously shown to partially extrude a synthetic S4 peptide. While no interaction was detected in liquid crystalline DMPC, upon cooling to induce the DMPC gel phase a reversible amide I peak (1633 cm(-1)) consistent with novel hydrogen bond formation was detected. This spectral shift was not observed for KV1 in the absence of ID (or vice versa), nor when the non-inactivating mutant V7E ID was applied to KV1 under similar conditions. Alteration of salt or redox conditions affected KV1-ID hydrogen bonding in a manner suggesting electrostatic KV1-ID interaction favored by a hairpin conformation for the ID and requiring extrusion of one or more KV1 domains from DMPC, consistent with ID binding to S4-S5. These findings support the utility of FTIR in detecting reversible interactions between soluble and membrane-embedded proteins, with lipid state-sensitivity of the conformation of the latter facilitating control of the interaction.


Subject(s)
Membrane Proteins , Peptides , Shaker Superfamily of Potassium Channels , Shaw Potassium Channels , Dimyristoylphosphatidylcholine/chemistry , Hydrogen Bonding , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Peptides/chemical synthesis , Peptides/chemistry , Peptides/metabolism , Protein Interaction Maps , Protein Structure, Tertiary , Shaker Superfamily of Potassium Channels/chemistry , Shaker Superfamily of Potassium Channels/metabolism , Shaw Potassium Channels/chemistry , Shaw Potassium Channels/metabolism , Solubility , Spectroscopy, Fourier Transform Infrared
9.
Biophys J ; 103(4): 669-76, 2012 Aug 22.
Article in English | MEDLINE | ID: mdl-22947928

ABSTRACT

Membrane proteins that respond to changes in transmembrane voltage are critical in regulating the function of living cells. The voltage-sensing domains (VSDs) of voltage-gated ion channels are extensively studied to elucidate voltage-sensing mechanisms, and yet many aspects of their structure-function relationship remain elusive. Here, we transplanted homologous amino acid motifs from the tetrameric voltage-activated potassium channel Kv3.1 to the monomeric VSD of Ciona intestinalis voltage-sensitive phosphatase (Ci-VSP) to explore which portions of Kv3.1 subunits depend on the tetrameric structure of Kv channels and which properties of Kv3.1 can be transferred to the monomeric Ci-VSP scaffold. By attaching fluorescent proteins to these chimeric VSDs, we obtained an optical readout to establish membrane trafficking and kinetics of voltage-dependent structural rearrangements. We found that motifs extending from 10 to roughly 100 amino acids can be readily transplanted from Kv3.1 into Ci-VSP to form engineered VSDs that efficiently incorporate into the plasma membrane and sense voltage. Some of the functional features of these engineered VSDs are reminiscent of Kv3.1 channels, indicating that these properties do not require interactions between Kv subunits or between the voltage sensing and the pore domains of Kv channels.


Subject(s)
Ciona intestinalis/enzymology , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Protein Engineering/methods , Shaw Potassium Channels/genetics , Shaw Potassium Channels/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Molecular Sequence Data , PC12 Cells , Phosphoric Monoester Hydrolases/chemistry , Porosity , Protein Structure, Tertiary , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Shaw Potassium Channels/chemistry
10.
Am J Physiol Lung Cell Mol Physiol ; 303(7): L598-607, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22865553

ABSTRACT

Pulmonary neuroepithelial bodies (NEBs), composed of clusters of amine [serotonin (5-HT)] and peptide-producing cells, are widely distributed within the airway mucosa of human and animal lungs. NEBs are thought to function as airway O(2)-sensors, since they are extensively innervated and release 5-HT upon hypoxia exposure. The small cell lung carcinoma cell line (H146) provides a useful model for native NEBs, since they contain (and secrete) 5-HT and share the expression of a membrane-delimited O(2) sensor [classical NADPH oxidase (NOX2) coupled to an O(2)-sensitive K(+) channel]. In addition, both native NEBs and H146 cells express different NADPH oxidase homologs (NOX1, NOX4) and its subunits together with a variety of O(2)-sensitive voltage-dependent K(+) channel proteins (K(v)) and tandem pore acid-sensing K(+) channels (TASK). Here we used H146 cells to investigate the role and interactions of various NADPH oxidase components in O(2)-sensing using a combination of coimmunoprecipitation, Western blot analysis (quantum dot labeling), and electrophysiology (patchclamp, amperometry) methods. Coimmunoprecipitation studies demonstrated formation of molecular complexes between NOX2 and K(v)3.3 and K(v)4.3 ion channels but not with TASK1 ion channels, while NOX4 associated with TASK1 but not with K(v) channel proteins. Downregulation of mRNA for NOX2, but not for NOX4, suppressed hypoxia-sensitive outward current and significantly reduced hypoxia -induced 5-HT release. Collectively, our studies suggest that NOX2/K(v) complexes are the predominant O(2) sensor in H146 cells and, by inference, in native NEBs. Present findings favor a NEB cell-specific plasma membrane model of O(2)-sensing and suggest that unique NOX/K(+) channel combinations may serve diverse physiological functions.


Subject(s)
Chemoreceptor Cells/physiology , Membrane Glycoproteins/physiology , NADPH Oxidases/physiology , Neuroepithelial Bodies/physiology , Oxygen/physiology , Cell Line, Tumor , Down-Regulation , Humans , Membrane Glycoproteins/chemistry , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/chemistry , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/physiology , Potassium Channels, Tandem Pore Domain/chemistry , Potassium Channels, Tandem Pore Domain/physiology , Serotonin/metabolism , Shal Potassium Channels/chemistry , Shal Potassium Channels/physiology , Shaw Potassium Channels/chemistry , Shaw Potassium Channels/physiology
11.
Biophys J ; 101(7): 1613-22, 2011 Oct 05.
Article in English | MEDLINE | ID: mdl-21961587

ABSTRACT

Several voltage-gated ion channels are modulated by clinically relevant doses of general anesthetics. However, the structural basis of this modulation is not well understood. Previous work suggested that n-alcohols and inhaled anesthetics stabilize the closed state of the Shaw2 voltage-gated (Kv) channel (K-Shaw2) by directly interacting with a discrete channel site. We hypothesize that the inhibition of K-Shaw2 channels by general anesthetics is governed by interactions between binding and effector sites involving components of the channel's activation gate. To investigate this hypothesis, we applied Ala/Val scanning mutagenesis to the S4-S5 linker and the post-PVP S6 segment, and conducted electrophysiological analysis to evaluate the energetic impact of the mutations on the inhibition of the K-Shaw2 channel by 1-butanol and halothane. These analyses identified residues that determine an apparent binding cooperativity and residue pairs that act in concert to modulate gating upon anesthetic binding. In some instances, due to their critical location, key residues also influence channel gating. Complementing these results, molecular dynamics simulations and in silico docking experiments helped us visualize possible anesthetic sites and interactions. We conclude that the inhibition of K-Shaw2 by general anesthetics results from allosteric interactions between distinct but contiguous binding and effector sites involving inter- and intrasubunit interfaces.


Subject(s)
Anesthetics, General/pharmacology , Molecular Dynamics Simulation , Potassium Channel Blockers/pharmacology , Shaw Potassium Channels/antagonists & inhibitors , Shaw Potassium Channels/metabolism , 1-Butanol/pharmacology , Amino Acid Sequence , Computational Biology , Dose-Response Relationship, Drug , Electrophysiological Phenomena/drug effects , Halothane/pharmacology , Ion Channel Gating/drug effects , Molecular Sequence Data , Mutagenesis , Mutation , Protein Structure, Secondary , Shaw Potassium Channels/chemistry , Shaw Potassium Channels/genetics
12.
Biophys J ; 101(6): 1354-63, 2011 Sep 21.
Article in English | MEDLINE | ID: mdl-21943416

ABSTRACT

Potassium currents generated by voltage-gated potassium (Kv) channels comprising α-subunits from the Kv1, 2, and 3 subfamilies facilitate high-frequency firing of mammalian neurons. Within these subfamilies, only three α-subunits (Kv1.4, Kv3.3, and Kv3.4) generate currents that decay rapidly in the open state because an N-terminal ball domain blocks the channel pore after activation-a process termed N-type inactivation. Despite its importance to shaping cellular excitability, little is known of the processes regulating surface expression of N-type α-subunits, versus their slowly inactivating (delayed rectifier) counterparts. Here we found that currents generated by homomeric Kv1.4, Kv3.3, and Kv3.4 channels are all strongly suppressed by the single transmembrane domain ancillary (ß) subunits KCNE1 and KCNE2. A combination of electrophysiological, biochemical, and immunofluorescence analyses revealed this suppression is due to KCNE1 and KCNE2 retaining Kv1.4 and Kv3.4 intracellularly, early in the secretory pathway. The retention is specific, requires α-ß coassembly, and does not involve the dynamin-dependent endocytosis pathway. However, the small fraction of Kv3.4 that escapes KCNE-dependent retention is regulated by dynamin-dependent endocytosis. The findings illustrate two contrasting mechanisms controlling surface expression of N-type Kv α-subunits and therefore, potentially, cellular excitability and refractory periods.


Subject(s)
Electric Conductivity , Potassium Channels, Voltage-Gated/metabolism , Shaw Potassium Channels/antagonists & inhibitors , Shaw Potassium Channels/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Endocytosis , Gene Expression Regulation , Humans , Intracellular Space/metabolism , Protein Subunits/antagonists & inhibitors , Protein Subunits/chemistry , Protein Subunits/metabolism , Rats , Shaw Potassium Channels/chemistry
13.
J Neurosci ; 30(47): 15987-6001, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-21106837

ABSTRACT

Precise targeting of various voltage-gated ion channels to proper membrane domains is crucial for their distinct roles in neuronal excitability and synaptic transmission. How each channel protein is transported within the cytoplasm is poorly understood. Here, we report that KIF5/kinesin I transports Kv3.1 voltage-gated K(+) (Kv) channels through the axon initial segment (AIS) via direct binding. First, we have identified a novel interaction between Kv3.1 and KIF5, confirmed by immunoprecipitation from mouse brain lysates and by pull-down assays with exogenously expressed proteins. The interaction is mediated by a direct binding between the Kv3.1 N-terminal T1 domain and a conserved region in KIF5 tail domains, in which proper T1 tetramerization is crucial. Overexpression of this region of KIF5B markedly reduces axonal levels of Kv3.1bHA. In mature hippocampal neurons, endogenous Kv3.1b and KIF5 colocalize. Suppressing the endogenous KIF5B level by RNA interference significantly reduces the Kv3.1b axonal level. Furthermore, mutating the Zn(2+)-binding site within T1 markedly decreases channel axonal targeting and forward trafficking, likely through disrupting T1 tetramerization and hence eliminating the binding to KIF5 tail. The mutation also alters channel activity. Interestingly, coexpression of the YFP (yellow fluorescent protein)-tagged KIF5B assists dendritic Kv3.1a and even mutants with a faulty axonal targeting motif to penetrate the AIS. Finally, fluorescently tagged Kv3.1 channels colocalize and comove with KIF5B along axons revealed by two-color time-lapse imaging. Our findings suggest that the binding to KIF5 ensures properly assembled and functioning Kv3.1 channels to be transported into axons.


Subject(s)
Axons/metabolism , Kinesins/metabolism , Shaw Potassium Channels/metabolism , Animals , Animals, Newborn , Cells, Cultured , Gene Knockdown Techniques , HEK293 Cells , Humans , Kinesins/chemistry , Mice , Protein Binding/physiology , Protein Transport/physiology , Rats , Shaw Potassium Channels/chemistry
14.
PLoS One ; 4(10): e7474, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19830250

ABSTRACT

BACKGROUND: The ability to disrupt the function of a specific protein on a rapid time scale provides a powerful tool for biomedical research. Specific proteases provide a potential method to selectively cleave a chosen protein, but rapid control of protease activity is difficult. METHODOLOGY/PRINCIPAL FINDINGS: A heterologous expression system for rapid target-directed proteolysis in mammalian cells was developed. The system consists of an inducible NIa protease from the tobacco etch virus (TEVp) and a chosen protein into which a TEVp substrate recognition sequence (TRS) has been inserted. Inducible activity was conferred to the TEVp using rapamycin-controlled TEVp fragment complementation. TEVp activity was assayed using a FRET-based reporter construct. TEVp expression was well tolerated by mammalian cells and complete cleavage of the substrate was possible. Cleavage at 37 degrees C proceeded exponentially with a time constant of approximately 150 minutes. Attempts to improve cleavage efficiency were hampered by substantial background activity, which was attributed to inherent affinity between the TEVp fragments. A second TEVp assay, based on changes in inactivation of a modified K(V)3.4 channel, showed that functional properties of a channel can be using altered using an inducible TEVp system. Similar levels of background activity and variability were observed in both electrophysiological and FRET assays. CONCLUSIONS/SIGNIFICANCE: The results suggested that an optimum level of TEVp expression leading to sufficient inducible activity (with minimal background activity) exists but the variability in expression levels between cells makes the present system rather impractical for single cell experiments. The system is likely to be more suitable for experiments in which the cell-to-cell variability is less of an issue; for example, in experiments involving large populations of cells.


Subject(s)
Endopeptidases/chemistry , Genetic Techniques , Potyvirus/metabolism , Protein Engineering/methods , Proteins/chemistry , Sirolimus/chemistry , Cell Line , Electrophysiology/methods , Fluorescence Resonance Energy Transfer/methods , Humans , Kinetics , Models, Chemical , Shaw Potassium Channels/chemistry , Temperature , Time Factors
15.
Biosci Rep ; 29(5): 301-13, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-18937645

ABSTRACT

Mammalian brains contain relatively high amounts of common and uncommon sialylated N-glycan structures. Sialic acid linkages were identified for voltage-gated potassium channels, Kv3.1, 3.3, 3.4, 1.1, 1.2 and 1.4, by evaluating their electrophoretic migration patterns in adult rat brain membranes digested with various glycosidases. Additionally, their electrophoretic migration patterns were compared with those of NCAM (neural cell adhesion molecule), transferrin and the Kv3.1 protein heterologously expressed in B35 neuroblastoma cells. Metabolic labelling of the carbohydrates combined with glycosidase digestion reactions were utilized to show that the N-glycan of recombinant Kv3.1 protein was capped with an oligo/poly-sialyl unit. All three brain Kv3 glycoproteins, like NCAM, were terminated with alpha2,3-linked sialyl residues, as well as atypical alpha2,8-linked sialyl residues. Additionally, at least one of their antennae was terminated with an oligo/poly-sialyl unit, similar to recombinant Kv3.1 and NCAM. In contrast, brain Kv1 glycoproteins consisted of sialyl residues with alpha2,8-linkage, as well as sialyl residues linked to internal carbohydrate residues of the carbohydrate chains of the N-glycans. This type of linkage was also supported for Kv3 glycoproteins. To date, such a sialyl linkage has only been identified in gangliosides, not N-linked glycoproteins. We conclude that all six Kv channels (voltage-gated K+ channels) contribute to the alpha2,8-linked sialylated N-glycan pool in mammalian brain and furthermore that their N-glycan structures contain branched sialyl residues. Identification of these novel and unique sialylated N-glycan structures implicate a connection between potassium channel activity and atypical sialylated N-glycans in modulating and fine-tuning the excitable properties of neurons in the nervous system.


Subject(s)
N-Acetylneuraminic Acid/metabolism , Neurons/metabolism , Polysaccharides/chemistry , Shaw Potassium Channels/chemistry , Shaw Potassium Channels/metabolism , Animals , Brain/metabolism , Cells, Cultured , Deuterium/metabolism , Neuroblastoma/pathology , Rats , Rats, Sprague-Dawley , Transfection
16.
BMC Neurosci ; 9: 52, 2008 Jun 19.
Article in English | MEDLINE | ID: mdl-18565223

ABSTRACT

BACKGROUND: Voltage-gated ion channels are membrane proteins containing a selective pore that allows permeable ions to transit the membrane in response to a change in the transmembrane voltage. The typical selectivity filter in potassium channels is formed by a tetrameric arrangement of the carbonyl groups of the conserved amino-acid sequence Gly-Tyr-Gly. This canonical pore is opened or closed by conformational changes that originate in the voltage sensor (S4), a transmembrane helix with a series of positively charged amino acids. This sensor moves through a gating pore formed by elements of the S1, S2 and S3 helices, across the plane of the membrane, without allowing ions to pass through the membrane at that site. Recently, synthetic mutagenesis studies in the Drosophila melanogaster Shaker channel and analysis of human disease-causing mutations in sodium channels have identified amino acid residues that are integral parts of the gating-pore; when these residues are mutated the proteins allow a non-specific cation current, known as the omega current, to pass through the gating-pore with relatively low selectivity. RESULTS: The N.at-Kv3.2 potassium channel has an unusual weak inward rectifier phenotype. Several mutations of two amino acids in the voltage sensing (S4) transmembrane helix change the phenotype to a typical delayed rectifier. The inward rectifier channels (wild-type and mutant) are sensitive to 4-aminopyridine (4-AP) but not tetra-ethyl ammonium (TEA), whereas the delayed rectifier mutants are sensitive to TEA but not 4-AP. The inward rectifier channels also manifest low cation selectivity. The relative selectivity for different cations is sensitive to specific mutations in the S4 helix, CONCLUSION: N.at-Kv3.2, a naturally occurring potassium channel of the Kv3 sequence family, mediates ion permeation through a modified gating pore, not the canonical, highly selective pore typical of potassium channels. This channel has evolved to yield qualitatively different ion permeability when compared to all other members of this gene family.


Subject(s)
Shaw Potassium Channels/genetics , Shaw Potassium Channels/metabolism , Turbellaria/metabolism , Amino Acid Sequence , Animals , Conserved Sequence , Drosophila/metabolism , Female , Ion Channel Gating/genetics , Models, Molecular , Ovum , Phenotype , Rats , Shaw Potassium Channels/chemistry
17.
J Biol Chem ; 283(32): 22283-94, 2008 Aug 08.
Article in English | MEDLINE | ID: mdl-18539595

ABSTRACT

Modulation of some Kv3 family potassium channels by protein kinase C (PKC) regulates their amplitude and kinetics and adjusts firing patterns of auditory neurons in response to stimulation. Nevertheless, little is known about the modulation of Kv3.3, a channel that is widely expressed throughout the nervous system and is the dominant Kv3 family member in auditory brainstem. We have cloned the cDNA for the Kv3.3 channel from mouse brain and have expressed it in a mammalian cell line and in Xenopus oocytes to characterize its biophysical properties and modulation by PKC. Kv3.3 currents activate at positive voltages and undergo inactivation with time constants of 150-250 ms. Activators of PKC increased current amplitude and removed inactivation of Kv3.3 currents, and a specific PKC pseudosubstrate inhibitor peptide prevented the effects of the activators. Elimination of the first 78 amino acids of the N terminus of Kv3.3 produced noninactivating currents suggesting that PKC modulates N-type inactivation, potentially by phosphorylation of sites in this region. To identify potential phosphorylation sites, we investigated the response of channels in which serines in this N-terminal domain were subjected to mutagenesis. Our results suggest that serines at positions 3 and 9 are potential PKC phosphorylation sites. Computer simulations of model neurons suggest that phosphorylation of Kv3.3 by PKC may allow neurons to maintain action potential height during stimulation at high frequencies, and may therefore contribute to stimulus-induced changes in the intrinsic excitability of neurons such as those of the auditory brainstem.


Subject(s)
Protein Kinase C/metabolism , Shaw Potassium Channels/antagonists & inhibitors , Shaw Potassium Channels/metabolism , Amino Acid Sequence , Animals , Brain/physiology , CHO Cells , Computer Simulation , Cricetinae , Cricetulus , Mice , Molecular Sequence Data , Mutation , Neurons/physiology , Phosphorylation , Recombinant Proteins/metabolism , Sequence Alignment , Serine/metabolism , Shaw Potassium Channels/chemistry , Shaw Potassium Channels/genetics
18.
Biochem Cell Biol ; 86(1): 21-30, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18364742

ABSTRACT

The N-glycan pool of mammalian brain contains remarkably high levels of sialylated N-glycans. This study provides the first evidence that voltage-gated K+ channels Kv3.1, Kv3.3, and Kv3.4, possess distinct sialylated N-glycan structures throughout the central nervous system of the adult rat. Electrophoretic migration patterns of Kv3.1, Kv3.3, and Kv3.4 glycoproteins from spinal cord, hypothalamus, thalamus, cerebral cortex, hippocampus, and cerebellum membranes digested with glycosidases were used to identify the various glycoforms. Differences in the migration of Kv3 proteins were attributed to the desialylated N-glycans. Expression levels of the Kv3 proteins were highest in cerebellum, whereas those of Kv3.1 and Kv3.3 were much lower in the other 5 regions. The lowest level of Kv3.1 was expressed in the hypothalamus, whereas the lowest levels of Kv3.3 were expressed in both thalamus and hypothalamus. The other regions expressed intermediate levels of Kv3.3, with spinal cord expressing the highest. The expression level of Kv3.4 in the hippocampus was slightly lower than that in cerebellum, and was closely followed by the other 4 regions, with spinal cord expressing the lowest level. We suggest that novel Kv3 glycoforms may endow differences in channel function and expression among regions throughout the central nervous system.


Subject(s)
N-Acetylneuraminic Acid/chemistry , Polysaccharides/chemistry , Protein Isoforms , Shaw Potassium Channels , Animals , Brain/anatomy & histology , Brain/metabolism , Protein Conformation , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley , Shaw Potassium Channels/chemistry , Shaw Potassium Channels/metabolism
19.
J Neurosci ; 27(51): 14158-70, 2007 Dec 19.
Article in English | MEDLINE | ID: mdl-18094255

ABSTRACT

Kv3 (Shaw) channels regulate rapid spiking, transmitter release and dendritic integration of many central neurons. Crucial to functional diversity are the complex targeting patterns of channel proteins. However, the targeting mechanisms are not known. Here we report that the axon-dendrite targeting of Kv3.1 is controlled by a conditional interaction of a C-terminal axonal targeting motif (ATM) with the N-terminal T1 domain and adaptor protein ankyrin G. In cultured hippocampal neurons, although the two splice variants of Kv3.1, Kv3.1a and Kv3.1b, are differentially targeted to the somatodendritic and axonal membrane, respectively, the lysine-rich ATM is surprisingly common for both splice variants. The ATM not only directly binds to the T1 domain in a Zn2+-dependent manner, but also associates with the ankyrin-repeat domain of ankyrin G. However, the full-length channel proteins of Kv3.1b display stronger association to ankyrin G than those of Kv3.1a, suggesting that the unique splice domain at Kv3.1b C terminus influences ATM binding to T1 and ankyrin G. Because ankyrin G mainly resides at the axon initial segment, we propose that it may function as a barrier for axon-dendrite targeting of Kv3.1 channels. In support of this idea, disrupting ankyrin G function either by over-expressing a dominant-negative mutant or by siRNA knockdown decreases polarized axon-dendrite targeting of both Kv3.1a and Kv3.1b. We conclude that the conditional ATM masked by the T1 domain in Kv3.1a is exposed by the splice domain in Kv3.1b, and is subsequently recognized by ankyrin G to target Kv3.1b into the axon.


Subject(s)
Ankyrins/metabolism , Axons/metabolism , Dendrites/metabolism , Gene Targeting , Shaw Potassium Channels/metabolism , Amino Acid Motifs , Animals , Ankyrins/chemistry , Cell Line , Cells, Cultured , Gene Targeting/methods , Humans , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Structure, Tertiary , Rats , Shaw Potassium Channels/chemistry
20.
Biochim Biophys Acta ; 1770(4): 666-71, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17197096

ABSTRACT

Neuronal Kv3 voltage-gated K(+) channels have two absolutely conserved N-glycosylation sites. Here, it is shown that Kv3.1, 3.3, and 3.4 channels are N-glycosylated in rat brain. Digestion of total brain membranes with peptide N glycosidase F (PNGase F) produced faster migrating immunobands than those of undigested membranes. Additionally, partial PNGase F digests showed that both sites are occupied by oligosaccharides. Neuraminidase treatment produced a smaller immunoband shift relative to PNGase F treatment. These results indicate that both sites are highly available and occupied by N-linked oligosaccharides for Kv3.1, 3.3, and 3.4 in rat brain, and furthermore that at least one oligosaccharide is of complex type. Additionally, these results point to an extracytoplasmic S1-S2 linker in Kv3 proteins expressed in native membranes. We suggest that N-glycosylation processing of Kv3 channels is critical for the expression of K(+) currents at the surface of neurons, and perhaps contributes to the pathophysiology of congenital disorders of glycosylation.


Subject(s)
Brain/metabolism , Oligosaccharides/metabolism , Protein Processing, Post-Translational , Shaw Potassium Channels/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Brain/cytology , Carbohydrate Conformation , Cell Membrane/metabolism , Female , Glycosylation , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/metabolism , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , Neuraminidase/metabolism , Neurons/metabolism , Oligosaccharides/chemistry , Potassium Channels, Voltage-Gated/metabolism , Protein Conformation , Rats , Shaw Potassium Channels/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...