Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
J Neurosci ; 43(43): 7073-7083, 2023 10 25.
Article in English | MEDLINE | ID: mdl-37648450

ABSTRACT

Neuronal Kv7 voltage-gated potassium channels generate the M-current and regulate neuronal excitability. Here, we report that dehydroepiandrosterone sulfate (DHEAS) is an endogenous Kv7 channel modulator that attenuates Gq-coupled receptor-induced M-current suppression. DHEAS reduced muscarinic agonist-induced Kv7-current suppression of Kv7.1, Kv7.2, Kv7.4, or Kv7.5 homomeric currents and endogenous M-currents in rat sympathetic ganglion neurons. However, DHEAS per se did not alter the voltage dependence of these Kv7 homomeric channels or the m1 receptor-induced activation of phospholipase C or protein kinase C. DHEAS-treated Kv7.2 homomeric currents became resistant to depletion of phosphatidylinositol 4,5-bisphosphate (PIP2) induced by voltage-activated phosphatase, Ci-VSP or eVSP. Our computational models predicted a novel binding site for DHEAS in the cytoplasmic domain of Kv7 subunits. A single-point mutation of the predicted key histidine into cysteine in the rat Kv7.2 subunit, rKv7.2(H558C), resulted in a loss of effects of DHEAS on muscarinic Kv7 current suppression. Furthermore, in vivo administration of DHEAS in mice of both sexes reduced late phase pain responses in the formalin paw test. However, it did not have effects on early phase responses in the formalin paw test or responses in the hot plate test. Coadministration of a selective Kv7 inhibitor, XE991, and DHEAS eliminated analgesic effects of DHEAS in late phase responses in the formalin paw test. Collectively, these results suggest that DHEAS attenuates M-current suppression by stabilizing PIP2-Kv7 subunit interaction and can mitigate inflammatory pain.SIGNIFICANCE STATEMENT M-current suppression induced by stimulation of Gq-coupled receptors is a form of Kv7 current modulation that can reversibly increase neuronal excitability. This study demonstrates that DHEAS, an endogenous steroid hormone, is a novel Kv7 channel modulator that can attenuate M-current suppression without affecting basal Kv7 channel kinetics. Administration of DHEAS in vivo alleviated inflammatory pain in rodents. These results suggest that the degree of M-current suppression can be dynamically regulated by small molecules. Therefore, this novel form of Kv7 channel regulation holds promising potential as a therapeutic target for sensitized nervous activities, such as inflammatory pain.


Subject(s)
KCNQ2 Potassium Channel , Muscarinic Agonists , Male , Female , Mice , Rats , Animals , Dehydroepiandrosterone Sulfate , KCNQ2 Potassium Channel/metabolism , Muscarinic Agonists/pharmacology , Pain/drug therapy , Formaldehyde , KCNQ3 Potassium Channel/genetics , KCNQ3 Potassium Channel/metabolism
2.
World Neurosurg ; 148: e581-e588, 2021 04.
Article in English | MEDLINE | ID: mdl-33476779

ABSTRACT

BACKGROUND: The purpose of this study was to compare clinical results of microendoscopic laminectomy (MEL) with those of unilateral biportal endoscopic laminectomy (UBEL) in patients with single-level lumbar spinal canal stenosis. METHODS: The subjects consisted of 181 patients who underwent MEL (139 cases) and UBEL (42 cases) who were followed up for at least 6 months. All patients had lumber canal stenosis for 1 level. Outcomes of the patients were assessed with the duration of surgery, the bone resection area in 3-dimensional computed tomography, the facet preservation rates in computed tomography axial imagery, Visual Analog Scale (VAS) for low back pain, the Oswestry Disability Index, and the EuroQol 5-Dimensions questionnaire. RESULTS: The bone resection area in 3-dimensional computed tomography was 1.5 for MEL versus 1.0 cm2 for UBEL (P < 0.05). The facet preservation rates on the advancing side and the opposite side were 78% versus 86% (advancing side: MEL vs. UBEL) and 85% versus 94% (opposite side) (P < 0.05). The VAS (low back pain) score, VAS (leg pain), Oswestry Disability Index, and EuroQol 5-Dimension questionnaire significantly dropped in both groups at the final period (P < 0.05), however, exhibiting no difference between the 2 groups at each period. MEL resulted in greater numbers of complications, including 5 cases of hematoma paralysis, 8 cases of dura injury, 2 cases of reoperation, as opposed to zero cases of hematoma paralysis and only 2 cases of dura injury resulting from UBEL. CONCLUSIONS: The UBEL method is a more useful technique than the MEL method as it requires a smaller bone resection area and produces fewer complications.


Subject(s)
Endoscopy/methods , Laminectomy/methods , Microsurgery/methods , Spinal Stenosis/surgery , Aged , Disability Evaluation , Female , Follow-Up Studies , Humans , Low Back Pain/etiology , Male , Middle Aged , Pain Measurement , Postoperative Complications/epidemiology , Retrospective Studies , Spinal Stenosis/diagnostic imaging , Spine/diagnostic imaging , Spine/surgery , Surveys and Questionnaires , Tomography, X-Ray Computed , Treatment Outcome
3.
Antioxidants (Basel) ; 9(11)2020 Nov 13.
Article in English | MEDLINE | ID: mdl-33202933

ABSTRACT

Opening of large conductance calcium-activated and voltage-dependent potassium (BKCa) channels hyperpolarizes plasma membranes of smooth muscle (SM) to cause vasodilation, underling a key mechanism for mediating uterine artery (UA) dilation in pregnancy. Hydrogen sulfide (H2S) has been recently identified as a new UA vasodilator, yet the mechanism underlying H2S-induced UA dilation is unknown. Here, we tested whether H2S activated BKCa channels in human UA smooth muscle cells (hUASMC) to mediate UA relaxation. Multiple BKCa subunits were found in human UA in vitro and hUASMC in vitro, and high ß1 and γ1 proteins were localized in SM cells in human UA. Baseline outward currents, recorded by whole-cell and single-channel patch clamps, were significantly inhibited by specific BKCa blockers iberiotoxin (IBTX) or tetraethylammonium, showing specific BKCa activity in hUASMC. H2S dose (NaHS, 1-1000 µM)-dependently potentiated BKCa currents and open probability. Co-incubation with a Ca2+ blocker nifedipine (5 µM) or a chelator (ethylene glycol-bis (ß-aminoethyl ether)-N,N,N',N'-tetraacetic acid (EGTA), 5 mM) did not alter H2S-potentiated BKCa currents and open probability. NaHS also dose-dependently relaxed phenylephrine pre-constricted freshly prepared human UA rings, which was inhibited by IBTX. Thus, H2S stimulated human UA relaxation at least partially via activating SM BKCa channels independent of extracellular Ca2+.

4.
Photobiomodul Photomed Laser Surg ; 38(8): 507-511, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32780687

ABSTRACT

Background: Transsacral epiduroscopic laser decompression (SELD) is a very noninvasive surgery, so it is effective for elderly patients and athletes and is a new and minimally invasive therapeutic technique that may be useful in many patients with discogenic low-back pain (LBP) having high signal intensity zone (HIZ) in magnetic resonance imaging (MRI). We investigated the clinical outcomes of SELD in Japanese patients with discogenic LBP having HIZ as a first trial. Methods: The subjects consisted of 52 patients who underwent SELD and were followed up for at least 6 months. All patients with LBP with HIZ were operative using the SELD technique. Outcomes of the patients were assessed with visual analogue scale (VAS) for LBP, the Oswestry disability index (ODI), and the EuroQol 5 dimension (EQ-5D). Statistical analyses were carried out using a paired t-test. A p-value of <0.05 was considered significant. For statistical analysis, we used the SPSS software program. Results: At 12 months after the procedure, the average VAS score for LBP fell to 1.2 from 5.6 (p-value <0.05). The ODI score also dropped from the preoperative level of 22.3 to 8.8. The EQ-5D score also significantly increased from the preoperative level of 0.865 (SD 0.10) to 0.950 (SD 0.05). Eight cases of intraoperative cervical pain were observed as complications with no cases of hematomas, infections, and postoperative neurosis was observed. Conclusions: SELD provides a novel minimally invasive technique capable of performing multilevel intervertebral surgery. We believe that SELD is an effective method of treating discogenic LBP due to HIZs.


Subject(s)
Intervertebral Disc Displacement/surgery , Laser Therapy , Low Back Pain/surgery , Decompression, Surgical/methods , Denervation/methods , Endoscopy , Female , Humans , Intervertebral Disc Displacement/complications , Japan , Low Back Pain/etiology , Male , Middle Aged , Sacrum
5.
Front Physiol ; 11: 513, 2020.
Article in English | MEDLINE | ID: mdl-32523549

ABSTRACT

Neuronal Kv7 channel generates a low voltage-activated potassium current known as the M-current. The M-current can be suppressed by various neurotransmitters that activate Gq-coupled receptors. Because the M-current stabilizes membrane potential at the resting membrane potential, its suppression transiently increase neuronal excitability. However, its physiological and pathological roles in vivo is not well understood to date. This review summarizes the molecular mechanism underlying M-current suppression, and why it remained elusive for many years. I also summarize how regulation of neuronal Kv7 channel contributes to anti-seizure action of valproic acid through inhibition of palmitoylation of a Kv7 channel binding protein, and discuss about a potential link with anti-seizure mechanisms of medium chain triglyceride ketogenic diet.

6.
J Neurosci ; 40(30): 5847-5856, 2020 07 22.
Article in English | MEDLINE | ID: mdl-32554550

ABSTRACT

The M-current is a low voltage-activated potassium current generated by neuronal Kv7 channels. A prominent role of the M-current is to a create transient increase of neuronal excitability in response to neurotransmitters through the suppression of this current. Accordingly, M-current suppression is assumed to be involved in higher brain functions including learning and memory. However, there is little evidence supporting such a role to date. To address this gap, we examined behavioral tasks to assess learning and memory in homozygous Kv7.2 knock-in mice, Kv7.2(S559A), which show reduced M-current suppression while maintaining a normal basal M-current activity in neurons. We found that Kv7.2(S559A) mice had normal object location memory and contextual fear memory, but impaired long-term object recognition memory. Furthermore, short-term memory for object recognition was intact in Kv7.2(S559A) mice. The deficit in long-term object recognition memory was restored by the administration of a selective Kv7 channel inhibitor, XE991, when delivered during the memory consolidation phase. Lastly, c-Fos induction 2 h after training in Kv7.2(S559A) mice was normal in the hippocampus, which corresponds to intact object location memory, but was reduced in the perirhinal cortex, which corresponds to impaired long-term object recognition memory. Together, these results support the overall conclusion that M-current suppression is important for memory consolidation of specific types of memories.SIGNIFICANCE STATEMENT Dynamic regulation of neuronal excitation is a fundamental mechanism for information processing in the brain, which is mediated by changes in synaptic transmissions or by changes in ion channel activity. Some neurotransmitters can facilitate action potential firing by suppression of a low voltage-activated potassium current, M-current. We demonstrate that M-current suppression is critical for establishment of long-term object recognition memory, but is not required for establishment of hippocampus-dependent location memory or contextual memory. This study suggests that M-current suppression is important for stable encoding of specific types of memories.


Subject(s)
KCNQ2 Potassium Channel/physiology , Memory Consolidation/physiology , Recognition, Psychology/physiology , Smell/physiology , Amino Acid Sequence , Animals , Fear/physiology , Fear/psychology , Female , Male , Memory Consolidation/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Odorants , Recognition, Psychology/drug effects , Smell/drug effects
7.
Proc Natl Acad Sci U S A ; 115(49): E11465-E11474, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30455320

ABSTRACT

A-kinase anchoring proteins (AKAPs) shape second-messenger signaling responses by constraining protein kinase A (PKA) at precise intracellular locations. A defining feature of AKAPs is a helical region that binds to regulatory subunits (RII) of PKA. Mining patient-derived databases has identified 42 nonsynonymous SNPs in the PKA-anchoring helices of five AKAPs. Solid-phase RII binding assays confirmed that 21 of these amino acid substitutions disrupt PKA anchoring. The most deleterious side-chain modifications are situated toward C-termini of AKAP helices. More extensive analysis was conducted on a valine-to-methionine variant in the PKA-anchoring helix of AKAP18. Molecular modeling indicates that additional density provided by methionine at position 282 in the AKAP18γ isoform deflects the pitch of the helical anchoring surface outward by 6.6°. Fluorescence polarization measurements show that this subtle topological change reduces RII-binding affinity 8.8-fold and impairs cAMP responsive potentiation of L-type Ca2+ currents in situ. Live-cell imaging of AKAP18γ V282M-GFP adducts led to the unexpected discovery that loss of PKA anchoring promotes nuclear accumulation of this polymorphic variant. Targeting proceeds via a mechanism whereby association with the PKA holoenzyme masks a polybasic nuclear localization signal on the anchoring protein. This led to the discovery of AKAP18ε: an exclusively nuclear isoform that lacks a PKA-anchoring helix. Enzyme-mediated proximity-proteomics reveal that compartment-selective variants of AKAP18 associate with distinct binding partners. Thus, naturally occurring PKA-anchoring-defective AKAP variants not only perturb dissemination of local second-messenger responses, but also may influence the intracellular distribution of certain AKAP18 isoforms.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/genetics , Membrane Proteins/metabolism , A Kinase Anchor Proteins/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation, Enzymologic , Genome-Wide Association Study , Humans , Membrane Proteins/genetics , Models, Molecular , Polymorphism, Single Nucleotide , Protein Binding , Protein Conformation , Protein Isoforms , Protein Transport
8.
Epilepsia ; 59(10): 1908-1918, 2018 10.
Article in English | MEDLINE | ID: mdl-30146722

ABSTRACT

OBJECTIVES: The M-current is a low-threshold voltage-gated potassium current generated by Kv7 subunits that regulates neural excitation. It is important to note that M-current suppression, induced by activation of Gq-coupled neurotransmitter receptors, can dynamically regulate the threshold of action-potential firing and firing frequency. Here we sought to directly examine whether M-current suppression is involved in seizures and epileptogenesis. METHODS: Kv7.2 knock-in mice lacking the key protein kinase C (PKC) phosphorylation acceptor site for M-current suppression were generated by introducing an alanine substitution at serine residue 559 of mouse Kv7.2, mKv7.2(S559A). Basic electrophysiologic properties of the M-current between wild-type and Kv7.2(S559A) knock-in mice were analyzed in primary cultured neurons. Homozygous Kv7.2(S559A) knock-in mice were used to evaluate the protective effect of mutant Kv7.2 channel against chemoconvulsant-induced seizures. In addition, pilocarpine-induced neuronal damage and spontaneously recurrent seizures were evaluated after equivalent chemoconvulsant-induced status epilepticus was achieved by coadministration of the M-current-specific channel inhibitor, XE991. RESULT: Neurons from Kv7.2(S559A) knock-in mice showed normal basal M-currents. Knock-in mice displayed reduced M-current suppression when challenged by a muscarinic agonist, oxotremorine-M. Kv7.2(S559A) mice were resistant to chemoconvulsant-induced seizures with no mortality. Administration of XE991 transiently exacerbated seizures in knock-in mice equivalent to those of wild-type mice. Valproate, which disrupts neurotransmitter-induced M-current suppression, showed no additional anticonvulsant effect in Kv7.2(S559A) mice. After experiencing status epilepticus, Kv7.2(S559A) knock-in mice did not show seizure-induced cell death or spontaneous recurring seizures. SIGNIFICANCE: This study provides evidence that neurotransmitter-induced suppression of M-current generated by Kv7.2-containing channels exacerbates behavioral seizures. In addition, prompt recovery of M-current after status epilepticus prevents subsequent neuronal death and the development of spontaneously recurrent seizures. Therefore, prompt restoration of M-current activity may have a therapeutic benefit for epilepsy.


Subject(s)
Gene Expression Regulation/genetics , KCNQ2 Potassium Channel/genetics , Membrane Potentials/genetics , Mutation/genetics , Status Epilepticus , Animals , Anticonvulsants/therapeutic use , Cells, Cultured , Cerebral Cortex/cytology , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Glutamate Decarboxylase/metabolism , KCNQ2 Potassium Channel/metabolism , Male , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscarinic Agonists/toxicity , Neurons/drug effects , Neurons/physiology , Pilocarpine/toxicity , Proto-Oncogene Proteins c-fos/metabolism , Status Epilepticus/chemically induced , Status Epilepticus/pathology , Status Epilepticus/prevention & control
9.
J Pharmacol Exp Ther ; 362(1): 177-185, 2017 07.
Article in English | MEDLINE | ID: mdl-28483800

ABSTRACT

M-channel inhibitors, especially XE991, are being used increasingly in animal experiments; however, insufficient characterization of XE991 at times confounds the interpretation of results when using this compound. Here, we demonstrate that XE991 and linopirdine are state-dependent inhibitors that favor the activated-subunit of neuronal Kv7/KCNQ channels. We performed patch-clamp experiments on homomeric Kv7.2 or heteromeric Kv7.2/3 channels expressed in Chinese hamster ovary cells to characterize XE991 and linopirdine. Neither inhibitor was efficacious around the resting membrane potential of cells in physiologic conditions. Inhibition of Kv7.2 and Kv7.2/3 channels by XE991 was closely related with channel activation. When the voltage dependence of activation was left-shifted by retigabine or right-shifted by the mutation, Kv7.2(R214D), the shift in half-activation voltage proportionally coincided with the shift in the half-effective potential for XE991 inhibition. Inhibition kinetics during XE991 wash-in was facilitated at depolarized potentials. Ten-minute washout of XE991 resulted in ∼30% current recovery, most of which was attributed to surface transport of Kv7.2 channels. Linopirdine also exhibited similar inhibition characteristics, with the exception of near- complete current recovery after washout at depolarized potentials. Inhibition kinetics of both XE991 and linopirdine was not as sensitive to changes in voltage as would be predicted by open- channel inhibition. Instead, they were well explained by binding to a single activated subunit. The characteristics of XE991 and linopirdine should be taken into account when these M-channel inhibitors are used in experiments.


Subject(s)
Anthracenes/pharmacology , Indoles/pharmacology , KCNQ1 Potassium Channel/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Pyridines/pharmacology , Animals , CHO Cells , Carbamates/pharmacology , Cricetinae , Cricetulus , KCNQ1 Potassium Channel/genetics , KCNQ2 Potassium Channel/drug effects , Kinetics , Membrane Potentials/drug effects , Mutation , Patch-Clamp Techniques , Phenylenediamines/pharmacology , Protein Subunits/drug effects , Rats
10.
Cell Mol Life Sci ; 74(3): 495-508, 2017 02.
Article in English | MEDLINE | ID: mdl-27645822

ABSTRACT

Neuronal Kv7 channels underlie a voltage-gated non-inactivating potassium current known as the M-current. Due to its particular characteristics, Kv7 channels show pronounced control over the excitability of neurons. We will discuss various factors that have been shown to drastically alter the activity of this channel such as protein and phospholipid interactions, phosphorylation, calcium, and numerous neurotransmitters. Kv7 channels locate to key areas for the control of action potential initiation and propagation. Moreover, we will explore the dynamic surface expression of the channel modulated by neurotransmitters and neural activity. We will also focus on known principle functions of neural Kv7 channels: control of resting membrane potential and spiking threshold, setting the firing frequency, afterhyperpolarization after burst firing, theta resonance, and transient hyperexcitability from neurotransmitter-induced suppression of the M-current. Finally, we will discuss the contribution of altered Kv7 activity to pathologies such as epilepsy and cognitive deficits.


Subject(s)
Action Potentials , Brain/physiology , KCNQ Potassium Channels/metabolism , Membrane Potentials , Animals , Brain/physiopathology , Calcium/metabolism , Calmodulin/metabolism , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Epilepsy/metabolism , Epilepsy/physiopathology , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , KCNQ Potassium Channels/analysis , Neurons/metabolism , Neurons/pathology , Signal Transduction
11.
J Cell Sci ; 128(22): 4235-45, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26446259

ABSTRACT

Neuronal excitability is strictly regulated by various mechanisms, including modulation of ion channel activity and trafficking. Stimulation of m1 muscarinic acetylcholine receptor (also known as CHRM1) increases neuronal excitability by suppressing the M-current generated by the Kv7/KCNQ channel family. We found that m1 muscarinic acetylcholine receptor stimulation also triggers surface transport of KCNQ subunits. This receptor-induced surface transport was observed with KCNQ2 as well as KCNQ3 homomeric channels, but not with Kv3.1 channels. Deletion analyses identified that a conserved domain in a proximal region of the N-terminal tail of KCNQ protein is crucial for this surface transport--the translocation domain. Proteins that bind to this domain were identified as α- and ß-tubulin and collapsin response mediator protein 2 (CRMP-2; also known as DPYSL2). An inhibitor of casein kinase 2 (CK2) reduced tubulin binding to the translocation domain, whereas an inhibitor of glycogen synthase kinase 3 (GSK3) facilitated CRMP-2 binding to the translocation domain. Consistently, treatment with the GSK3 inhibitor enhanced receptor-induced KCNQ2 surface transport. M-current recordings from neurons showed that treatment with a GSK3 inhibitor shortened the duration of muscarinic suppression and led to over-recovery of the M-current. These results suggest that m1 muscarinic acetylcholine receptor stimulates surface transport of KCNQ channels through a CRMP-2-mediated pathway.


Subject(s)
Acetylcholine/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , KCNQ2 Potassium Channel/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptor, Muscarinic M1/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetulus , Humans , Molecular Sequence Data
12.
J Clin Invest ; 125(10): 3904-14, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26348896

ABSTRACT

Valproic acid (VPA) has been widely used for decades to treat epilepsy; however, its mechanism of action remains poorly understood. Here, we report that the anticonvulsant effects of nonacute VPA treatment involve preservation of the M-current, a low-threshold noninactivating potassium current, during seizures. In a wide variety of neurons, activation of Gq-coupled receptors, such as the m1 muscarinic acetylcholine receptor, suppresses the M-current and induces hyperexcitability. We demonstrated that VPA treatment disrupts muscarinic suppression of the M-current and prevents resultant agonist-induced neuronal hyperexcitability. We also determined that VPA treatment interferes with M-channel signaling by inhibiting palmitoylation of a signaling scaffold protein, AKAP79/150, in cultured neurons. In a kainate-induced murine seizure model, administration of a dose of an M-channel inhibitor that did not affect kainate-induced seizure transiently eliminated the anticonvulsant effects of VPA. Retigabine, an M-channel opener that does not open receptor-suppressed M-channels, provided anticonvulsant effects only when administered prior to seizure induction in control animals. In contrast, treatment of VPA-treated mice with retigabine induced anticonvulsant effects even when administered after seizure induction. Together, these results suggest that receptor-induced M-current suppression plays a role in the pathophysiology of seizures and that preservation of the M-current during seizures has potential as an effective therapeutic strategy.


Subject(s)
Anticonvulsants/pharmacology , KCNQ2 Potassium Channel/physiology , Valproic Acid/pharmacology , A Kinase Anchor Proteins/genetics , A Kinase Anchor Proteins/metabolism , A Kinase Anchor Proteins/physiology , Action Potentials/drug effects , Animals , Anthracenes/pharmacology , Anticonvulsants/therapeutic use , Carbamates/pharmacology , Cells, Cultured , Drug Interactions , Female , Hippocampus/cytology , Humans , KCNQ2 Potassium Channel/drug effects , Kainic Acid/toxicity , Lipoylation/drug effects , Male , Mice , Mice, Inbred C57BL , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/pharmacology , Neurons/drug effects , Neurons/metabolism , Phenylenediamines/pharmacology , Phosphorylation/drug effects , Potassium Channel Blockers/pharmacology , Protein Processing, Post-Translational/drug effects , Rats , Receptor, Muscarinic M1/drug effects , Receptor, Muscarinic M1/physiology , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Seizures/chemically induced , Seizures/drug therapy , Seizures/physiopathology , Signal Transduction/drug effects , Superior Cervical Ganglion/cytology , Valproic Acid/therapeutic use
13.
J Biol Chem ; 289(16): 11536-11544, 2014 Apr 18.
Article in English | MEDLINE | ID: mdl-24627475

ABSTRACT

M-type potassium channels, encoded by the KCNQ family genes (KCNQ2-5), require calmodulin as an essential co-factor. Calmodulin bound to the KCNQ2 subunit regulates channel trafficking and stabilizes channel activity. We demonstrate that phosphorylation of calmodulin by protein kinase CK2 (casein kinase 2) rapidly and reversibly modulated KCNQ2 current. CK2-mediated phosphorylation of calmodulin strengthened its binding to KCNQ2 channel, caused resistance to phosphatidylinositol 4,5-bisphosphate depletion, and increased KCNQ2 current amplitude. Accordingly, application of CK2-selective inhibitors suppressed KCNQ2 current. This suppression was prevented by co-expression of CK2 phosphomimetic calmodulin mutants or pretreatment with a protein phosphatase inhibitor, calyculin A. We also demonstrated that functional CK2 and protein phosphatase 1 (PP1) were selectively tethered to the KCNQ2 subunit. We identified a functional KVXF consensus site for PP1 binding in the N-terminal tail of KCNQ2 subunit: mutation of this site augmented current density. CK2 inhibitor treatment suppressed M-current in rat superior cervical ganglion neurons, an effect negated by overexpression of phosphomimetic calmodulin or pretreatment with calyculin A Furthermore, CK2 inhibition diminished the medium after hyperpolarization by suppressing the M-current. These findings suggest that CK2-mediated phosphorylation of calmodulin regulates the M-current, which is tonically regulated by CK2 and PP1 anchored to the KCNQ2 channel complex.


Subject(s)
Calmodulin/metabolism , Casein Kinase II/metabolism , KCNQ2 Potassium Channel/metabolism , Neurons/metabolism , Protein Phosphatase 1/metabolism , Superior Cervical Ganglion/metabolism , Animals , CHO Cells , Calmodulin/genetics , Casein Kinase II/genetics , Cricetinae , Cricetulus , Enzyme Inhibitors/pharmacology , Humans , KCNQ2 Potassium Channel/genetics , Marine Toxins , Mutation , Neurons/cytology , Oxazoles/pharmacology , Protein Phosphatase 1/genetics , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Superior Cervical Ganglion/cytology
14.
PLoS One ; 8(12): e82290, 2013.
Article in English | MEDLINE | ID: mdl-24349250

ABSTRACT

All subtypes of KCNQ channel subunits (KCNQ1-5) require calmodulin as a co-factor for functional channels. It has been demonstrated that calmodulin plays a critical role in KCNQ channel trafficking as well as calcium-mediated current modulation. However, how calcium-bound calmodulin suppresses the M-current is not well understood. In this study, we investigated the molecular mechanism of KCNQ2 current suppression mediated by calcium-bound calmodulin. We show that calcium induced slow calmodulin dissociation from the KCNQ2 channel subunit. In contrast, in homomeric KCNQ3 channels, calcium facilitated calmodulin binding. We demonstrate that this difference in calmodulin binding was due to the unique cysteine residue in the KCNQ2 subunit at aa 527 in Helix B, which corresponds to an arginine residue in other KCNQ subunits including KCNQ3. In addition, a KCNQ2 channel associated protein AKAP79/150 (79 for human, 150 for rodent orthologs) also preferentially bound calcium-bound calmodulin. Therefore, the KCNQ2 channel complex was able to retain calcium-bound calmodulin either through the AKPA79/150 or KCNQ3 subunit. Functionally, increasing intracellular calcium by ionomycin suppressed currents generated by KCNQ2, KCNQ2(C527R) or heteromeric KCNQ2/KCNQ3 channels to an equivalent extent. This suggests that a change in the binding configuration, rather than dissociation of calmodulin, is responsible for KCNQ current suppression. Furthermore, we demonstrate that KCNQ current suppression was accompanied by reduced KCNQ affinity toward phosphatidylinositol 4,5-bisphosphate (PIP2) when assessed by a voltage-sensitive phosphatase, Ci-VSP. These results suggest that a rise in intracellular calcium induces a change in the configuration of CaM-KCNQ binding, which leads to the reduction of KCNQ affinity for PIP2 and subsequent current suppression.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , KCNQ2 Potassium Channel/metabolism , A Kinase Anchor Proteins/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Ionomycin/pharmacology , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Binding/drug effects , Protein Subunits/metabolism , Rats
15.
Proc Natl Acad Sci U S A ; 109(45): 18577-82, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23090990

ABSTRACT

Voltage-gated sodium (Na(V)) and potassium (K(V)) channels are critical components of neuronal action potential generation and propagation. Here, we report that Na(V)ß1 encoded by SCN1b, an integral subunit of Na(V) channels, coassembles with and modulates the biophysical properties of K(V)1 and K(V)7 channels, but not K(V)3 channels, in an isoform-specific manner. Distinct domains of Na(V)ß1 are involved in modulation of the different K(V) channels. Studies with channel chimeras demonstrate that Na(V)ß1-mediated changes in activation kinetics and voltage dependence of activation require interaction of Na(V)ß1 with the channel's voltage-sensing domain, whereas changes in inactivation and deactivation require interaction with the channel's pore domain. A molecular model based on docking studies shows Na(V)ß1 lying in the crevice between the voltage-sensing and pore domains of K(V) channels, making significant contacts with the S1 and S5 segments. Cross-modulation of Na(V) and K(V) channels by Na(V)ß1 may promote diversity and flexibility in the overall control of cellular excitability and signaling.


Subject(s)
Potassium Channels, Voltage-Gated/metabolism , Voltage-Gated Sodium Channel beta-1 Subunit/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Ion Channel Gating , Kinetics , Mice , Models, Molecular , PC12 Cells , Potassium Channels, Voltage-Gated/chemistry , Protein Binding , Protein Structure, Tertiary , Rats , Xenopus
16.
J Biol Chem ; 287(28): 23690-7, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22613709

ABSTRACT

ß(1)- and ß(2)-adrenergic receptors utilize different signaling mechanisms to control cardiac function. Recent studies demonstrated that ß(2)-adrenergic receptors (ß(2)ARs) colocalize with some ion channels that are critical for proper cardiac function. Here, we demonstrate that ß(2)ARs form protein complexes with the pacemaker HCN4 channel, as well as with other subtypes of HCN channels. The adrenergic receptor-binding site was identified at a proximal region of the N-terminal tail of the HCN4 channel. A synthetic peptide derived from the ß(2)AR-binding domain of the HCN4 channel disrupted interaction between HCN4 and ß(2)AR. In addition, treatment with this peptide prevented adrenergic augmentation of pacemaker currents and spontaneous contraction rates but did not affect adrenergic regulation of voltage-gated calcium currents. These results suggest that the ion channel-receptor complex is a critical mechanism in ion channel regulation.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/metabolism , Ion Channel Gating/physiology , Multiprotein Complexes/metabolism , Muscle Proteins/metabolism , Receptors, Adrenergic, beta-2/metabolism , Adrenergic beta-Agonists/pharmacology , Amino Acid Sequence , Animals , Animals, Newborn , Cells, Cultured , Cyclic Nucleotide-Gated Cation Channels/genetics , HEK293 Cells , HeLa Cells , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Immunoblotting , Isoproterenol/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Molecular Sequence Data , Muscle Proteins/genetics , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Patch-Clamp Techniques , Peptides/pharmacology , Potassium Channels , Protein Binding/drug effects , Protein Isoforms/genetics , Protein Isoforms/metabolism , Pyrimidines/pharmacology , Rats , Receptors, Adrenergic, beta-2/genetics , Sequence Homology, Amino Acid , Sinoatrial Node/cytology , Sinoatrial Node/drug effects , Sinoatrial Node/physiology
17.
EMBO J ; 31(14): 3147-56, 2012 May 29.
Article in English | MEDLINE | ID: mdl-22643219

ABSTRACT

Several neurotransmitters, including acetylcholine, regulate neuronal tone by suppressing a non-inactivating low-threshold voltage-gated potassium current generated by the M-channel. Agonist dependent control of the M-channel is mediated by calmodulin, activation of anchored protein kinase C (PKC), and depletion of the phospholipid messenger phosphatidylinositol 4,5-bisphosphate (PIP2). In this report, we show how this trio of second messenger responsive events acts synergistically and in a stepwise manner to suppress activity of the M-current. PKC phosphorylation of the KCNQ2 channel subunit induces dissociation of calmodulin from the M-channel complex. The calmodulin-deficient channel has a reduced affinity towards PIP2. This pathway enhances the effect of concomitant reduction of PIP2, which leads to disruption of the M-channel function. These findings clarify how a common lipid cofactor, such as PIP2, can selectively regulate ion channels.


Subject(s)
Ion Channel Gating/physiology , KCNQ2 Potassium Channel/metabolism , Receptors, Muscarinic/metabolism , Second Messenger Systems/physiology , Animals , CHO Cells , Cricetinae , Cricetulus , HEK293 Cells , Humans , KCNQ2 Potassium Channel/genetics , Phosphatidylinositol 4,5-Diphosphate/genetics , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphorylation/physiology , Protein Kinase C/genetics , Protein Kinase C/metabolism , Rats , Receptors, Muscarinic/genetics
18.
PLoS One ; 6(10): e26338, 2011.
Article in English | MEDLINE | ID: mdl-22043317

ABSTRACT

We previously reported that some ATP competitive protein kinase C (PKC) inhibitors are either competitive or uncompetitive inhibitors with respect to substrate peptides. In this report, we demonstrate how the interactions between PKC and inhibitors change PKC activation kinetics. A substrate competitive inhibitor, bisindolylmaleimide I, targets activated PKC and stabilizes PKC in the activated conformation. This leads to transient activation and prolonged deactivation of PKC in the presence of bisindolylmaleimide I. In contrast, an uncompetitive substrate inhibitor, bisindolylmaleimide IV, targets quiescent PKC and stabilizes PKC in the quiescent conformation, which generates slower activation and suppressed translocation upon activation of PKC.


Subject(s)
Adenosine Triphosphate/metabolism , Binding, Competitive , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Animals , Humans , Indoles , Kinetics , Maleimides , Protein Conformation , Protein Stability
19.
Mol Cell ; 37(4): 541-50, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20188672

ABSTRACT

A-kinase anchoring proteins (AKAPs) coordinate cell signaling events. AKAP79 brings together different combinations of enzyme binding partners to customize the regulation of effector proteins. In neurons, muscarinic agonists mobilize an AKAP79-anchored pool of PKC that phosphorylates the KCNQ2 subunit of the M channel. This inhibits potassium permeability to enhance neuronal excitability. Using a dual fluorescent imaging/patch-clamp technique, we visualized AKAP79-anchored PKC phosphorylation of the kinase activity reporter CKAR concurrently with electrophysiological changes in KCNQ2 channels to show that AKAP79 synchronizes both signaling events to optimize the attenuation of M currents. AKAP79 also protects PKC from certain ATP-competitive inhibitors. Related studies suggest that context-dependent protein-protein interactions alter the susceptibility of another protein kinase, PDK1, to ATP analog inhibitors. This implies that intracellular binding partners not only couple individual molecular events in a cell signaling process but can also change the pharmacological profile of certain protein kinases.


Subject(s)
A Kinase Anchor Proteins/metabolism , Adenosine Triphosphate/analogs & derivatives , Protein Kinase C/chemistry , Protein Kinase C/metabolism , Protein Kinase Inhibitors/chemistry , A Kinase Anchor Proteins/genetics , Adenosine Triphosphate/metabolism , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , Cricetulus , KCNQ2 Potassium Channel/genetics , KCNQ2 Potassium Channel/metabolism , Models, Molecular , Muscarine/metabolism , Phosphorylation , Protein Binding , Protein Kinase C/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Pyruvate Dehydrogenase Acetyl-Transferring Kinase
20.
Proc Natl Acad Sci U S A ; 105(34): 12557-62, 2008 Aug 26.
Article in English | MEDLINE | ID: mdl-18711127

ABSTRACT

A-Kinase Anchoring Proteins (AKAPs) ensure the fidelity of second messenger signaling events by directing protein kinases and phosphatases toward their preferred substrates. AKAP150 brings protein kinase A (PKA), the calcium/calmodulin dependent phosphatase PP2B and protein kinase C (PKC) to postsynaptic membranes where they facilitate the phosphorylation dependent modulation of certain ion channels. Immunofluorescence and electrophysiological recordings were combined with behavioral analyses to assess whether removal of AKAP150 by gene targeting in mice changes the signaling environment to affect excitatory and inhibitory neuronal processes. Mislocalization of PKA in AKAP150 null hippocampal neurons alters the bidirectional modulation of postsynaptic AMPA receptors with concomitant changes in synaptic transmission and memory retention. AKAP150 null mice also exhibit deficits in motor coordination and strength that are consistent with a role for the anchoring protein in the cerebellum. Loss of AKAP150 in sympathetic cervical ganglion (SCG) neurons reduces muscarinic suppression of inhibitory M currents and provides these animals with a measure of resistance to seizures induced by the non-selective muscarinic agonist pilocarpine. These studies argue that distinct AKAP150-enzyme complexes regulate context-dependent neuronal signaling events in vivo.


Subject(s)
A Kinase Anchor Proteins/deficiency , Neurons/physiology , Animals , Cerebellum/cytology , Cyclic AMP-Dependent Protein Kinases/metabolism , Hippocampus/cytology , Mice , Mice, Knockout , Motor Skills Disorders/etiology , Muscarinic Agonists/pharmacology , Nerve Tissue Proteins , Receptors, AMPA/metabolism , Seizures , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...