Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
J Neurosci ; 44(21)2024 May 22.
Article in English | MEDLINE | ID: mdl-38664011

ABSTRACT

Fragile X syndrome (FXS) arises from the loss of fragile X messenger ribonucleoprotein (FMRP) needed for normal neuronal excitability and circuit functions. Recent work revealed that FMRP contributes to mossy fiber long-term potentiation by adjusting the Kv4 A-type current availability through interactions with a Cav3-Kv4 ion channel complex, yet the mechanism has not yet been defined. In this study using wild-type and Fmr1 knock-out (KO) tsA-201 cells and cerebellar sections from male Fmr1 KO mice, we show that FMRP associates with all subunits of the Cav3.1-Kv4.3-KChIP3 complex and is critical to enabling calcium-dependent shifts in Kv4.3 inactivation to modulate the A-type current. Specifically, upon depolarization Cav3 calcium influx activates dual-specific phosphatase 1/6 (DUSP1/6) to deactivate ERK1/2 (ERK) and lower phosphorylation of Kv4.3, a signaling pathway that does not function in Fmr1 KO cells. In Fmr1 KO mouse tissue slices, cerebellar granule cells exhibit a hyperexcitable response to membrane depolarizations. Either incubating Fmr1 KO cells or in vivo administration of a tat-conjugated FMRP N-terminus fragment (FMRP-N-tat) rescued Cav3-Kv4 function and granule cell excitability, with a decrease in the level of DUSP6. Together these data reveal a Cav3-activated DUSP signaling pathway critical to the function of a FMRP-Cav3-Kv4 complex that is misregulated in Fmr1 KO conditions. Moreover, FMRP-N-tat restores function of this complex to rescue calcium-dependent control of neuronal excitability as a potential therapeutic approach to alleviating the symptoms of FXS.


Subject(s)
Calcium , Fragile X Mental Retardation Protein , Fragile X Syndrome , Mice, Knockout , Neurons , Animals , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Mice , Male , Fragile X Syndrome/metabolism , Fragile X Syndrome/genetics , Fragile X Syndrome/physiopathology , Neurons/metabolism , Calcium/metabolism , Mice, Inbred C57BL , Shal Potassium Channels/metabolism , Shal Potassium Channels/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism
2.
PLoS One ; 16(3): e0248688, 2021.
Article in English | MEDLINE | ID: mdl-33755670

ABSTRACT

Voltage-gated potassium (Kv) channels regulate the membrane potential and conductance of excitable cells to control the firing rate and waveform of action potentials. Even though Kv channels have been intensely studied for over 70 year, surprisingly little is known about how specific channels expressed in various neurons and their functional properties impact neuronal network activity and behavior in vivo. Although many in vivo genetic manipulations of ion channels have been tried, interpretation of these results is complicated by powerful homeostatic plasticity mechanisms that act to maintain function following perturbations in excitability. To better understand how Kv channels shape network function and behavior, we have developed a novel optogenetic technology to acutely regulate Kv channel expression with light by fusing the light-sensitive LOV domain of Vaucheria frigida Aureochrome 1 to the N-terminus of the Kv1 subunit protein to make an Opto-Kv1 channel. Recording of Opto-Kv1 channels expressed in Xenopus oocytes, mammalian cells, and neurons show that blue light strongly induces the current expression of Opto-Kv1 channels in all systems tested. We also find that an Opto-Kv1 construct containing a dominant-negative pore mutation (Opto-Kv1(V400D)) can be used to down-regulate Kv1 currents in a blue light-dependent manner. Finally, to determine whether Opto-Kv1 channels can elicit light-dependent behavioral effect in vivo, we targeted Opto-Kv1 (V400D) expression to Kv1.3-expressing mitral cells of the olfactory bulb in mice. Exposure of the bulb to blue light for 2-3 hours produced a significant increase in sensitivity to novel odors after initial habituation to a similar odor, comparable to behavioral changes seen in Kv1.3 knockout animals. In summary, we have developed novel photoactivatable Kv channels that provide new ways to interrogate neural circuits in vivo and to examine the roles of normal and disease-causing mutant Kv channels in brain function and behavior.


Subject(s)
Neurons , Shaker Superfamily of Potassium Channels/metabolism , Action Potentials , Animals , HEK293 Cells , Humans , Membrane Potentials , Mice , Mice, Transgenic , Neurons/cytology , Neurons/metabolism , Oocytes/cytology , Oocytes/metabolism , Xenopus laevis
3.
Front Cell Neurosci ; 8: 82, 2014.
Article in English | MEDLINE | ID: mdl-24723849

ABSTRACT

Auxiliary subunits are non-conducting, modulatory components of the multi-protein ion channel complexes that underlie normal neuronal signaling. They interact with the pore-forming α-subunits to modulate surface distribution, ion conductance, and channel gating properties. For the somatodendritic subthreshold A-type potassium (ISA) channel based on Kv4 α-subunits, two types of auxiliary subunits have been extensively studied: Kv channel-interacting proteins (KChIPs) and dipeptidyl peptidase-like proteins (DPLPs). KChIPs are cytoplasmic calcium-binding proteins that interact with intracellular portions of the Kv4 subunits, whereas DPLPs are type II transmembrane proteins that associate with the Kv4 channel core. Both KChIPs and DPLPs genes contain multiple start sites that are used by various neuronal populations to drive the differential expression of functionally distinct N-terminal variants. In turn, these N-terminal variants generate tremendous functional diversity across the nervous system. Here, we focus our review on (1) the molecular mechanism underlying the unique properties of different N-terminal variants, (2) the shaping of native ISA properties by the concerted actions of KChIPs and DPLP variants, and (3) the surprising ways that KChIPs and DPLPs coordinate the activity of multiple channels to fine-tune neuronal excitability. Unlocking the unique contributions of different auxiliary subunit N-terminal variants may provide an important opportunity to develop novel targeted therapeutics to treat numerous neurological disorders.

4.
PLoS One ; 9(3): e93315, 2014.
Article in English | MEDLINE | ID: mdl-24675763

ABSTRACT

Reactive oxygen species (ROS) regulate ion channels, modulate neuronal excitability, and contribute to the etiology of neurodegenerative disorders. ROS differentially suppress fast "ball-and-chain" N-type inactivation of cloned Kv1 and Kv3 potassium channels but not of Kv4 channels, likely due to a lack of reactive cysteines in Kv4 N-termini. Recently, we discovered that N-type inactivation of Kv4 channel complexes can be independently conferred by certain N-terminal variants of Kv4 auxiliary subunits (DPP6a, DPP10a). Here, we report that both DPP6a and DPP10a, like Kv subunits with redox-sensitive N-type inactivation, contain a highly conserved cysteine in their N-termini (Cys-13). To test if N-type inactivation mediated by DPP6a or DPP10a is redox sensitive, Xenopus oocyte recordings were performed to examine the effects of two common oxidants, tert-butyl hydroperoxide (tBHP) and diamide. Both oxidants markedly modulate DPP6a- or DPP10a-conferred N-type inactivation of Kv4 channels, slowing the overall inactivation and increasing the peak current. These functional effects are fully reversed by the reducing agent dithiothreitol (DTT) and appear to be due to a selective modulation of the N-type inactivation mediated by these auxiliary subunits. Mutation of DPP6a Cys-13 to serine eliminated the tBHP or diamide effects, confirming the importance of Cys-13 to the oxidative regulation. Biochemical studies designed to elucidate the underlying molecular mechanism show no evidence of protein-protein disulfide linkage formation following cysteine oxidation. Instead, using a biotinylated glutathione (BioGEE) reagent, we discovered that oxidation by tBHP or diamide leads to S-glutathionylation of Cys-13, suggesting that S-glutathionylation underlies the regulation of fast N-type inactivation by redox. In conclusion, our studies suggest that Kv4-based A-type current in neurons may show differential redox sensitivity depending on whether DPP6a or DPP10a is highly expressed, and that the S-glutathionylation mechanism may play a previously unappreciated role in mediating excitability changes and neuropathologies associated with ROS.


Subject(s)
Glutathione/metabolism , Oocytes/metabolism , Protein Subunits/metabolism , Reactive Oxygen Species/pharmacology , Shal Potassium Channels/metabolism , Action Potentials/drug effects , Amino Acid Sequence , Animals , Cattle , Diamide/pharmacology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Dithiothreitol/pharmacology , Female , Gene Expression , Humans , Kv Channel-Interacting Proteins/genetics , Kv Channel-Interacting Proteins/metabolism , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Oocytes/cytology , Oocytes/drug effects , Patch-Clamp Techniques , Potassium Channels/genetics , Potassium Channels/metabolism , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sequence Homology, Amino Acid , Shal Potassium Channels/antagonists & inhibitors , Shal Potassium Channels/genetics , Transfection , Xenopus laevis , tert-Butylhydroperoxide/pharmacology
5.
PLoS One ; 8(11): e79891, 2013.
Article in English | MEDLINE | ID: mdl-24236164

ABSTRACT

N-type inactivation occurs when the N-terminus of a potassium channel binds into the open pore of the channel. This study examined the relationship between activation and steady state inactivation for mutations affecting the N-type inactivation properties of the Aplysia potassium channel AKv1 expressed in Xenopus oocytes. The results show that the traditional single-step model for N-type inactivation fails to properly account for the observed relationship between steady state channel activation and inactivation curves. We find that the midpoint of the steady state inactivation curve depends in part on a secondary interaction between the channel core and a region of the N-terminus just proximal to the pore blocking peptide that we call the Inactivation Proximal (IP) region. The IP interaction with the channel core produces a negative shift in the activation and inactivation curves, without blocking the pore. A tripeptide motif in the IP region was identified in a large number of different N-type inactivation domains most likely reflecting convergent evolution in addition to direct descent. Point mutating a conserved hydrophobic residue in this motif eliminates the gating voltage shift, accelerates recovery from inactivation and decreases the amount of pore block produced during inactivation. The IP interaction we have identified likely stabilizes the open state and positions the pore blocking region of the N-terminus at the internal opening to the transmembrane pore by forming a Pre-Block (P state) interaction with residues lining the side window vestibule of the channel.


Subject(s)
Potassium Channels/chemistry , Potassium Channels/metabolism , Protein Interaction Domains and Motifs , Algorithms , Amino Acid Motifs , Amino Acid Sequence , Animals , Conserved Sequence , Drosophila , Ion Channel Gating , Kinetics , Models, Biological , Models, Molecular , Molecular Sequence Data , Mutation , Oocytes/metabolism , Potassium Channels/genetics , Protein Conformation , Sequence Alignment , Xenopus
6.
PLoS One ; 8(4): e62695, 2013.
Article in English | MEDLINE | ID: mdl-23638135

ABSTRACT

N-type inactivation is produced by the binding of a potassium channel's N-terminus within the open pore, blocking conductance. Previous studies have found that introduction of negative charges into N-terminal inactivation domains disrupts inactivation; however, the Aplysia AKv1 N-type inactivation domain contains two negatively charged residues, E2 and E9. Rather than being unusual, sequence analysis shows that this N-terminal motif is highly conserved among Kv1 sequences across many phyla. Conservation analysis shows some tolerance at position 9 for other charged residues, like D9 and K9, whereas position 2 is highly conserved as E2. To examine the functional importance of these residues, site directed mutagenesis was performed and effects on inactivation were recorded by two electrode voltage clamp in Xenopus oocytes. We find that inclusion of charged residues at positions 2 and 9 prevents interactions with non-polar sites along the inactivation pathway increasing the efficiency of pore block. In addition, E2 appears to have additional specific electrostatic interactions that stabilize the inactivated state likely explaining its high level of conservation. One possible explanation for E2's unique importance, consistent with our data, is that E2 interacts electrostatically with a positive charge on the N-terminal amino group to stabilize the inactivation domain at the block site deep within the pore. Simple electrostatic modeling suggests that due to the non-polar environment in the pore in the blocked state, even a 1 Å larger separation between these charges, produced by the E2D substitution, would be sufficient to explain the 65× reduced affinity of the E2D N-terminus for the pore. Finally, our studies support a multi-step, multi-site N-type inactivation model where the N-terminus interacts deep within the pore in an extended like structure placing the most N-terminal residues 35% of the way across the electric field in the pore blocked state.


Subject(s)
Protein Interaction Domains and Motifs , Shaker Superfamily of Potassium Channels/chemistry , Shaker Superfamily of Potassium Channels/metabolism , Static Electricity , Amino Acid Substitution/genetics , Amino Acids/chemistry , Animals , Membrane Potentials/physiology , Mutation , Oocytes/metabolism , Shaker Superfamily of Potassium Channels/genetics
7.
J Neurochem ; 126(4): 462-72, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23692269

ABSTRACT

K channel-interacting proteins (KChIPs) enhance functional expression of Kv4 channels by binding to an N-terminal regulatory region located in the first 40 amino acids of Kv4.2 that we call the functional expression regulating N-terminal (FERN) domain. Mutating two residues in the FERN domain to alanines, W8A and F11A, disrupts KChIP binding and regulation of Kv4.2 without eliminating the FERN domain's control of basal expression level or regulation by DPP6. When Kv4.2(W8A,F11A) is co-expressed with wild type Kv4.2 and KChIP3 subunits, a dominant negative effect is seen where the current expression is reduced to levels normally seen without KChIP addition. The dominant negative effect correlates with heteromultimeric channels remaining on intracellular membranes despite KChIP binding to non-mutant Kv4.2 subunits. In contrast, the deletion mutant Kv4.2(Δ1-40), eliminating both KChIP binding and the FERN domain, has no dominant negative effect even though the maximal conductance level is 5x lower than seen with KChIP3. The 5x increased expression seen with KChIP integration into the channel is fully apparent even when a reduced number of KChIP subunits are incorporated as long as all FERN domains are bound. Our results support the hypothesis that KChIPs enhances Kv4.2 functional expression by a 1 : 1 suppression of the N-terminal FERN domain and by producing additional positive regulatory effects on functional channel expression.


Subject(s)
Kv Channel-Interacting Proteins/genetics , Kv Channel-Interacting Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Shal Potassium Channels/genetics , Shal Potassium Channels/metabolism , Animals , CHO Cells , COS Cells , Chlorocebus aethiops , Cloning, Molecular , Cricetinae , Green Fluorescent Proteins/genetics , Intracellular Membranes/metabolism , Ion Channel Gating/physiology , Kv Channel-Interacting Proteins/chemistry , Membrane Potentials/physiology , Mutagenesis/physiology , Protein Binding/physiology , Protein Structure, Tertiary , Protein Subunits/genetics , Protein Subunits/metabolism , Protein Transport/physiology , Repressor Proteins/chemistry , Shal Potassium Channels/chemistry
8.
PLoS One ; 8(4): e60831, 2013.
Article in English | MEDLINE | ID: mdl-23593319

ABSTRACT

Dipeptidyl Peptidase-like Protein 6 (DPP6) is widely expressed in the brain where it co-assembles with Kv4 channels and KChIP auxiliary subunits to regulate the amplitude and functional properties of the somatodendritic A-current, ISA. Here we show that in cerebellar granule (CG) cells DPP6 also regulates resting membrane potential and input resistance by increasing the amplitude of the IK(SO) resting membrane current. Pharmacological analysis shows that DPP6 acts through the control of a channel with properties matching the K2P channel TASK-3. Heterologous expression and co-immunoprecipitation shows that DPP6 co-expression with TASK-3 results in the formation of a protein complex that enhances resting membrane potassium conductance. The co-regulation of resting and voltage-gated channels by DPP6 produces coordinate shifts in resting membrane potential and A-current gating that optimize the sensitivity of ISA inactivation gating to subthreshold fluctuations in resting membrane potential.


Subject(s)
Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Potassium Channels/metabolism , Action Potentials , Animals , Cell Line , Cells, Cultured , Cerebellum/cytology , Cerebellum/metabolism , Cricetinae , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Gene Expression , Membrane Potentials , Mice , Models, Neurological , Neurons/physiology , Potassium Channels/genetics , RNA Interference , Shal Potassium Channels/metabolism
9.
PLoS One ; 7(6): e38205, 2012.
Article in English | MEDLINE | ID: mdl-22675523

ABSTRACT

Dipeptidyl peptidase-like protein 6 (DPP6) proteins co-assemble with Kv4 channel α-subunits and Kv channel-interacting proteins (KChIPs) to form channel protein complexes underlying neuronal somatodendritic A-type potassium current (I(SA)). DPP6 proteins are expressed as N-terminal variants (DPP6a, DPP6K, DPP6S, DPP6L) that result from alternative mRNA initiation and exhibit overlapping expression patterns. Here, we study the role DPP6 variants play in shaping the functional properties of I(SA) found in cerebellar granule (CG) cells using quantitative RT-PCR and voltage-clamp recordings of whole-cell currents from reconstituted channel complexes and native I(SA) channels. Differential expression of DPP6 variants was detected in rat CG cells, with DPP6K (41 ± 3%)>DPP6a (33 ± 3%)>>DPP6S (18 ± 2%)>DPP6L (8 ± 3%). To better understand how DPP6 variants shape native neuronal I(SA), we focused on studying interactions between the two dominant variants, DPP6K and DPP6a. Although previous studies did not identify unique functional effects of DPP6K, we find that the unique N-terminus of DPP6K modulates the effects of KChIP proteins, slowing recovery and producing a negative shift in the steady-state inactivation curve. By contrast, DPP6a uses its distinct N-terminus to directly confer rapid N-type inactivation independently of KChIP3a. When DPP6a and DPP6K are co-expressed in ratios similar to those found in CG cells, their distinct effects compete in modulating channel function. The more rapid inactivation from DPP6a dominates during strong depolarization; however, DPP6K produces a negative shift in the steady-state inactivation curve and introduces a slow phase of recovery from inactivation. A direct comparison to the native CG cell I(SA) shows that these mixed effects are present in the native channels. Our results support the hypothesis that the precise expression and co-assembly of different auxiliary subunit variants are important factors in shaping the I(SA) functional properties in specific neuronal populations.


Subject(s)
Cerebellum/cytology , Cytoplasmic Granules/enzymology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Ion Channel Gating/physiology , Multiprotein Complexes/metabolism , Shal Potassium Channels/metabolism , Alternative Splicing/genetics , Amino Acid Sequence , Animals , Conserved Sequence/genetics , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/chemistry , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Evolution, Molecular , Kinetics , Molecular Sequence Data , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Xenopus laevis
10.
J Neurosci ; 30(25): 8551-65, 2010 Jun 23.
Article in English | MEDLINE | ID: mdl-20573902

ABSTRACT

In cerebellar granule (CG) cells and many other neurons, A-type potassium currents play an important role in regulating neuronal excitability, firing patterns, and activity-dependent plasticity. Protein biochemistry has identified dipeptidyl peptidase-like protein 6 (DPP6) as an auxiliary subunit of Kv4-based A-type channels and thus a potentially important regulator of neuronal excitability. In this study, we used an RNA interference (RNAi) strategy to examine the role DPP6 plays in forming and shaping the electrophysiological properties of CG cells. DPP6 RNAi delivered by lentiviral vectors effectively disrupts DPP6 protein expression in CG cells. In response to the loss of DPP6, I(SA) peak conductance amplitude is reduced by >85% in parallel with a dramatic reduction in the level of I(SA) channel protein complex found in CG cells. The I(SA) channels remaining in CG cells after suppression of DPP6 show alterations in gating similar to Kv4 channels expressed in heterologous systems without DPP6. In addition to these effects on A-type current, we find that loss of DPP6 has additional effects on input resistance and Na(+) channel conductance that combine with the effects on I(SA) to produce a global change in excitability. Overall, DPP6 expression seems to be critical for the expression of a high-frequency electrophysiological phenotype in CG cells by increasing leak conductance, A-type current levels and kinetics, and Na(+) current amplitude.


Subject(s)
Action Potentials/physiology , Cerebellum/physiology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Nerve Tissue Proteins/metabolism , Neurons/physiology , Potassium Channels/metabolism , Analysis of Variance , Blotting, Western , Cell Line , Cerebellum/cytology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Electrophysiology , Genetic Vectors , Hippocampus/cytology , Hippocampus/physiology , Humans , Lentivirus , Nerve Tissue Proteins/genetics , Potassium Channels/genetics , RNA Interference , Shal Potassium Channels/physiology
11.
Channels (Austin) ; 3(6): 448-61, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19901547

ABSTRACT

The somatodendritic subthreshold A-type K(+) current in neurons (I(SA)) depends on its kinetic and voltage-dependent properties to regulate membrane excitability, action potential repetitive firing, and signal integration. Key functional properties of the K(V)4 channel complex underlying I(SA) are determined by dipeptidyl peptidase-like proteins known as dipeptidyl peptidase 6 (DPP6) and dipeptidyl peptidase 10 (DPP10). Among the multiple known DPP10 isoforms with alternative N-terminal sequences, DPP10a confers exceptionally fast inactivation to K(V)4.2 channels. To elucidate the molecular basis of this fast inactivation, we investigated the structure-function relationship of the DPP10a N-terminal region and its interaction with the K(V)4.2 channel. Here, we show that DPP10a shares a conserved N-terminal sequence (MNQTA) with DPP6a (aka DPP6-E), which also induces fast inactivation. Deletion of the NQTA sequence in DPP10a eliminates this dramatic fast inactivation, and perfusion of MNQTA peptide to the cytoplasmic face of inside-out patches inhibits the K(V)4.2 current. DPP10a-induced fast inactivation exhibits competitive interactions with internally applied tetraethylammonium (TEA), and elevating the external K(+) concentration accelerates recovery from DPP10a-mediated fast inactivation. These results suggest that fast inactivation induced by DPP10a or DPP6a is mediated by a common N-terminal inactivation motif via a pore-blocking mechanism. This mechanism may offer an attractive target for novel pharmacological interventions directed at impairing I(SA) inactivation and reducing neuronal excitability.


Subject(s)
Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/physiology , Shal Potassium Channels/antagonists & inhibitors , Amino Acid Motifs , Amino Acid Sequence , Animals , Conserved Sequence , DNA, Complementary , Neurons/metabolism , Oocytes , Patch-Clamp Techniques , Plasmids , Protein Binding , Rats , Rats, Sprague-Dawley , Xenopus laevis
12.
J Gen Physiol ; 134(1): 15-34, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19528261

ABSTRACT

N-type inactivation of voltage-gated potassium channels is an autoinhibitory process that occurs when the N terminus binds within the channel pore and blocks conduction. N-type inactivation and recovery occur with single-exponential kinetics, consistent with a single-step reaction where binding and block occur simultaneously. However, recent structure-function studies have suggested the presence of a preinactivated state whose formation and loss regulate inactivation and recovery kinetics. Our studies on N-type inactivation of the Shaker-type AKv1 channel support a multiple-step inactivation process involving a series of conformational changes in distinct regions of the N terminus that we have named the polar, flex, and latch regions. The highly charged polar region forms interactions with the surface of the channel leading up to the side window openings between the T1 domain and the channel transmembrane domains, before the rate-limiting step occurs. This binding culminates with a specific electrostatic interaction between R18 and EDE161-163 located at the entrance to the side windows. The latch region appears to work together with the flex region to block the pore after polar region binding occurs. Analysis of tail currents for a latch region mutant shows that both blocked and unblocked states exist after the rate-limiting transition is passed. Our results suggest that at least two intermediate states exist for N-type inactivation: a polar region-bound state that is formed before the rate-limiting step, and a pre-block state that is formed by the flex and latch regions during the rate-limiting step.


Subject(s)
Cell Membrane/chemistry , Cell Membrane/physiology , Ion Channel Gating/physiology , Oocytes/physiology , Shaker Superfamily of Potassium Channels/chemistry , Shaker Superfamily of Potassium Channels/physiology , Animals , Cells, Cultured , Porosity , Xenopus laevis
13.
J Biol Chem ; 283(51): 36046-59, 2008 Dec 19.
Article in English | MEDLINE | ID: mdl-18957440

ABSTRACT

Kv channel-interacting proteins (KChIPs) are auxiliary subunits of the heteromultimeric channel complexes that underlie neuronal I(SA), the subthreshold transient K(+) current that dynamically regulates membrane excitability, action potential firing properties, and long term potentiation. KChIPs form cytoplasmic associations with the principal pore-forming Kv4 subunits and typically mediate enhanced surface expression and accelerated recovery from depolarization-induced inactivation. An exception is KChIP4a, which dramatically suppresses Kv4 inactivation while promoting neither surface expression nor recovery. These unusual properties are attributed to the effects of a K channel inactivation suppressor domain (KISD) encoded within the variable N terminus of KChIP4a. Here, we have functionally and biochemically characterized two brain KChIP isoforms, KChIP2x and KChIP3x (also known as KChIP3b) and show that they also contain a functional KISD. Like KChIP4a and in contrast with non-KISD-containing KChIPs, both KChIP2x and KChIP3x strongly suppress inactivation and slow activation and inhibit the typical increases in surface expression of Kv4.2 channels. We then examined the properties of the KISD to determine potential mechanisms for its action. Subcellular fractionation shows that KChIP4a, KChIP2x, and KChIP3x are highly associated with the membrane fraction. Fluorescent confocal imaging of enhanced green fluorescent proteins (eGFP) N-terminally fused with KISD in HEK293T cells indicates that KISDs of KChIP4a, KChIP2x, and KChIP3x all autonomously target eGFP to intracellular membranes. Cell surface biotinylation experiments on KChIP4a indicate that the N terminus is exposed extracellularly, consistent with a transmembrane KISD. In summary, KChIP4a, KChIP2x, and KChIP3x comprise a novel class of KChIP isoforms characterized by an unusual transmembrane domain at their N termini that modulates Kv4 channel gating and trafficking.


Subject(s)
Cell Membrane/metabolism , Ion Channel Gating/physiology , Kv Channel-Interacting Proteins/metabolism , Protein Subunits/metabolism , Shal Potassium Channels/metabolism , Animals , Cell Line , Cell Membrane/genetics , Humans , Kv Channel-Interacting Proteins/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary/physiology , Protein Subunits/genetics , Rats , Shal Potassium Channels/genetics
14.
Mol Cell Neurosci ; 35(4): 604-24, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17475505

ABSTRACT

Dipeptidyl peptidase-like proteins (DPLs) and Kv-channel-interacting proteins (KChIPs) join Kv4 pore-forming subunits to form multi-protein complexes that underlie subthreshold A-type currents (I(SA)) in neuronal somatodendritic compartments. Here, we characterize the functional effects and brain distributions of N-terminal variants belonging to the DPL dipeptidyl peptidase 10 (DPP10). In the Kv4.2+KChIP3+DPP10 channel complex, all DPP10 variants accelerate channel gating kinetics; however, the splice variant DPP10a produces uniquely fast inactivation kinetics that accelerates with increasing depolarization. This DPP10a-specific inactivation dominates in co-expression studies with KChIP4a and other DPP10 isoforms. Real-time qRT-PCR and in situ hybridization analyses reveal differential expression of DPP10 variants in rat brain. DPP10a transcripts are prominently expressed in the cortex, whereas DPP10c and DPP10d mRNAs exhibit more diffuse distributions. Our results suggest that DPP10a underlies rapid inactivation of cortical I(SA), and the regulation of isoform expression may contribute to the variable inactivation properties of I(SA) across different brain regions.


Subject(s)
Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Shal Potassium Channels/metabolism , Amino Acid Sequence , Animals , Brain/anatomy & histology , Brain/metabolism , Humans , In Situ Hybridization , Ion Channel Gating , Isoenzymes/genetics , Isoenzymes/metabolism , Kv Channel-Interacting Proteins/genetics , Kv Channel-Interacting Proteins/metabolism , Membrane Potentials/physiology , Molecular Sequence Data , Patch-Clamp Techniques , Protein Subunits/genetics , Protein Subunits/metabolism , Rats , Rats, Sprague-Dawley , Sequence Alignment , Shal Potassium Channels/genetics , Xenopus laevis
15.
J Biol Chem ; 282(18): 13637-47, 2007 May 04.
Article in English | MEDLINE | ID: mdl-17331952

ABSTRACT

The thiol-based redox regulation of proteins plays a central role in cellular signaling. Here, we investigated the redox regulation at the Zn(2+) binding site (HX(5)CX(20)CC) in the intracellular T1-T1 inter-subunit interface of a Kv4 channel. This site undergoes conformational changes coupled to voltage-dependent gating, which may be sensitive to oxidative stress. The main results show that internally applied nitric oxide (NO) inhibits channel activity profoundly. This inhibition is reversed by reduced glutathione and suppressed by intracellular Zn(2+), and at least two Zn(2+) site cysteines are required to observe the NO-induced inhibition (Cys-110 from one subunit and Cys-132 from the neighboring subunit). Biochemical evidence suggests strongly that NO induces a disulfide bridge between Cys-110 and Cys-132 in intact cells. Finally, further mutational studies suggest that intra-subunit Zn(2+) coordination involving His-104, Cys-131, and Cys-132 protects against the formation of the inhibitory disulfide bond. We propose that the interfacial T1 Zn(2+) site of Kv4 channels acts as a Zn(2+)-dependent redox switch that may regulate the activity of neuronal and cardiac A-type K(+) currents under physiological and pathological conditions.


Subject(s)
Cysteine/metabolism , Nitric Oxide/metabolism , Shal Potassium Channels/metabolism , Zinc/metabolism , Animals , Cysteine/genetics , Disulfides/chemistry , Disulfides/metabolism , Ion Transport/physiology , Membrane Potentials/physiology , Mice , Mutation, Missense , Myocardium/metabolism , Nerve Tissue/metabolism , Nitric Oxide/chemistry , Oxidation-Reduction , Shal Potassium Channels/chemistry , Shal Potassium Channels/genetics , Xenopus , Zinc/chemistry
16.
J Neurochem ; 99(4): 1207-23, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17026528

ABSTRACT

The somatodendritic A-current, I(SA), in hippocampal CA1 pyramidal neurons regulates the processing of synaptic inputs and the amplitude of back propagating action potentials into the dendritic tree, as well as the action potential firing properties at the soma. In this study, we have used RNA interference and over-expression to show that expression of the Kv4.2 gene specifically regulates the I(SA) component of A-current in these neurons. In dissociated hippocampal pyramidal neuron cultures, or organotypic cultured CA1 pyramidal neurons, the expression level of Kv4.2 is such that the I(SA) channels are maintained in the population at a peak conductance of approximately 950 pS/pF. Suppression of Kv4.2 transcripts in hippocampal pyramidal neurons using an RNA interference vector suppresses I(SA) current by 60% in 2 days, similar to the effect of expressing dominant-negative Kv4 channel constructs. Increasing the expression of Kv4.2 in these neurons increases the level of I(SA) to 170% of the normal set point without altering the biophysical properties. Our results establish a specific role for native Kv4.2 transcripts in forming and maintaining I(SA) current at characteristic levels in hippocampal pyramidal neurons.


Subject(s)
Action Potentials/genetics , Hippocampus/metabolism , Pyramidal Cells/metabolism , Shal Potassium Channels/genetics , Shal Potassium Channels/metabolism , Action Potentials/drug effects , Animals , COS Cells , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cells, Cultured , Chlorocebus aethiops , Dendrites/metabolism , Down-Regulation/genetics , Gene Expression Regulation/genetics , Genetic Vectors/genetics , Hippocampus/drug effects , Hippocampus/ultrastructure , Microscopy, Electron, Transmission , Organ Culture Techniques , Pyramidal Cells/drug effects , Pyramidal Cells/ultrastructure , RNA Interference , Rats , Rats, Sprague-Dawley , Shal Potassium Channels/drug effects , Synaptic Transmission/genetics
17.
J Biol Chem ; 281(17): 11769-79, 2006 Apr 28.
Article in English | MEDLINE | ID: mdl-16513649

ABSTRACT

Small conductance, Ca2+-activated voltage-independent potassium channels (SK channels) are widely expressed in diverse tissues; however, little is known about the molecular regulation of SK channel subunits. Direct alteration of ion channel subunits by kinases is a candidate mechanism for functional modulation of these channels. We find that activation of cyclic AMP-dependent protein kinase (PKA) with forskolin (50 microm) causes a dramatic decrease in surface localization of the SK2 channel subunit expressed in COS7 cells due to direct phosphorylation of the SK2 channel subunit. PKA phosphorylation studies using the intracellular domains of the SK2 channel subunit expressed as glutathione S-transferase fusion protein constructs showed that both the amino-terminal and carboxyl-terminal regions are PKA substrates in vitro. Mutational analysis identified a single PKA phosphorylation site within the amino-terminal of the SK2 subunit at serine 136. Mutagenesis and mass spectrometry studies identified four PKA phosphorylation sites: Ser465 (minor site) and three amino acid residues Ser568, Ser569, and Ser570 (major sites) within the carboxyl-terminal region. A mutated SK2 channel subunit, with the three contiguous serines mutated to alanines to block phosphorylation at these sites, shows no decrease in surface expression after PKA stimulation. Thus, our findings suggest that PKA phosphorylation of these three sites is necessary for PKA-mediated reorganization of SK2 surface expression.


Subject(s)
Cell Membrane/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Colforsin/pharmacology , Cyclic AMP/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Mass Spectrometry , Molecular Sequence Data , Mutagenesis , Mutation/genetics , Phosphorylation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Small-Conductance Calcium-Activated Potassium Channels/genetics
18.
Biochemistry ; 45(6): 1663-72, 2006 Feb 14.
Article in English | MEDLINE | ID: mdl-16460013

ABSTRACT

Some eukaryotic voltage-gated K+ (Kv) channels contain an N-terminal inactivation peptide (IP), which mediates a fast inactivation process that limits channel function during membrane depolarization and thus shapes the action potential. We obtained sequence-specific nuclear magnetic resonance (NMR) assignments for the polypeptide backbone of a tetrameric N-terminal fragment (amino acids 1-181) of the Aplysia Kv1.1 channel. Additional NMR measurements show that the tetramerization domain 1 (T1) has the same globular structure in solution as previously determined by crystallography and that the IP (residues 1-20) and the linker (residues 21-65) are in a flexibly disordered, predominantly extended conformation. A potential contact site between the T1 domain and the flexible tail (residues 1-65) has been identified on the basis of chemical-shift changes of individual T1 domain amino acids, which map to the T1 surface near the interface between adjacent subunits. Paramagnetic perturbation experiments further indicate that, in the ensemble of solution conformers, there is at least a small population of species with the IP localized in close proximity to the proposed interacting residues of the T1 tetramer. Electrophysiological measurements show that all three mutations in this pocket that we tested slow the rate of inactivation and speed up recovery, as predicted from the preinactivation site model. These results suggest that specific, short-lived transient interactions between the T1 domain and the IP or the linker segment may play a role in defining the regulatory kinetics of fast channel inactivation.


Subject(s)
Eukaryotic Cells/chemistry , Shaker Superfamily of Potassium Channels/chemistry , Amino Acid Sequence , Animals , Binding Sites , Eukaryotic Cells/physiology , Humans , Kinetics , Magnetic Resonance Spectroscopy/methods , Protein Conformation , Protein Structure, Tertiary , Shaker Superfamily of Potassium Channels/physiology
19.
Am J Physiol Cell Physiol ; 290(1): C165-71, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16135544

ABSTRACT

Voltage-dependent (Kv)4.2-encoded A-type K+ channels play an important role in controlling neuronal excitability and are subject to modulation by various protein kinases, including ERK. In studies of ERK modulation, the organic compound U0126 is often used to suppress the activity of MEK, which is a kinase immediately upstream from ERK. We have observed that the inactivation time constant of heterologously expressed Kv4.2 channels was accelerated by U0126 at 1-20 microM. This effect, however, was not Kv4 family specific, because U0126 also converted noninactivating K+ currents mediated by Kv1.1 subunits into transient ones. To determine whether U0126 exerted these effects through kinase inhibition, we tested U0125, a derivative of U0126 that is less potent in MEK inhibition. At the same concentrations, U0125 had effects similar to those of U0126 on channel inactivation. Finally, we expressed a mutant form of Kv4.2 in which three identified ERK phosphorylation sites (T602, T607, and S616) were replaced with alanines. The inactivation of K+ currents mediated by this mutant was still accelerated by U0126. Our data favor the conclusion that the increase in the rate of channel inactivation by U0126 is likely to be independent of protein kinase inhibition and instead represents a direct action on channel gating.


Subject(s)
Butadienes/pharmacology , Enzyme Inhibitors/pharmacology , Ion Channel Gating/drug effects , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Nitriles/pharmacology , Shal Potassium Channels/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Dose-Response Relationship, Drug , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Molecular Sequence Data , Potassium/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Rats , Shal Potassium Channels/antagonists & inhibitors , Shal Potassium Channels/genetics , Transfection
20.
J Physiol ; 568(Pt 3): 767-88, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16123112

ABSTRACT

Kv4 pore-forming subunits are the principal constituents of the voltage-gated K+ channel underlying somatodendritic subthreshold A-type currents (I(SA)) in neurones. Two structurally distinct types of Kv4 channel modulators, Kv channel-interacting proteins (KChIPs) and dipeptidyl-peptidase-like proteins (DPLs: DPP6 or DPPX, DPP10 or DPPY), enhance surface expression and modify functional properties. Since KChIP and DPL distributions overlap in the brain, we investigated the potential coassembly of Kv4.2, KChIP3 and DPL proteins, and the contribution of DPLs to ternary complex properties. Immunoprecipitation results show that KChIP3 and DPP10 associate simultaneously with Kv4.2 proteins in rat brain as well as heterologously expressing Xenopus oocytes, indicating Kv4.2 + KChIP3 + DPP10 multiprotein complexes. Consistent with ternary complex formation, coexpression of Kv4.2, KChIP3 and DPP10 in oocytes and CHO cells results in current waveforms distinct from the arithmetic sum of Kv4.2 + KChIP3 and Kv4.2 + DPP10 currents. Furthermore, the Kv4.2 + KChIP3 + DPP10 channels recover from inactivation very rapidly (tau(rec) approximately 18-26 ms), closely matching that of native I(SA) and significantly faster than the recovery of Kv4.2 + KChIP3 or Kv4.2 + DPP10 channels. For comparison, identical triple coexpression experiments were performed using DPP6 variants. While most results are similar, the Kv4.2 + KChIP3 + DPP6 channels exhibit inactivation that slows with increasing membrane potential, resulting in inactivation slower than that of Kv4.2 + KChIP3 + DPP10 channels at positive voltages. In conclusion, the native neuronal subthreshold A-type channel is probably a macromolecular complex formed from Kv4 and a combination of both KChIP and DPL proteins, with the precise composition of channel alpha and auxiliary subunits underlying tissue and regional variability in I(SA) properties.


Subject(s)
Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/physiology , Ion Channel Gating/physiology , Kv Channel-Interacting Proteins/physiology , Oocytes/physiology , Potassium/metabolism , Shal Potassium Channels/physiology , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/chemistry , Kv Channel-Interacting Proteins/chemistry , Membrane Potentials/physiology , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Rats , Shal Potassium Channels/chemistry , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...