Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 196
1.
Protein Sci ; 33(6): e4995, 2024 Jun.
Article En | MEDLINE | ID: mdl-38747377

Membrane proteins play critical physiological roles as receptors, channels, pumps, and transporters. Despite their importance, however, low expression levels often hamper the experimental characterization of membrane proteins. We present an automated and web-accessible design algorithm called mPROSS (https://mPROSS.weizmann.ac.il), which uses phylogenetic analysis and an atomistic potential, including an empirical lipophilicity scale, to improve native-state energy. As a stringent test, we apply mPROSS to the Kv1.2-Kv2.1 paddle chimera voltage-gated potassium channel. Four designs, encoding 9-26 mutations relative to the parental channel, were functional and maintained potassium-selective permeation and voltage dependence in Xenopus oocytes with up to 14-fold increase in whole-cell current densities. Additionally, single-channel recordings reveal no significant change in the channel-opening probability nor in unitary conductance, indicating that functional expression levels increase without impacting the activity profile of individual channels. Our results suggest that the expression levels of other dynamic channels and receptors may be enhanced through one-shot design calculations.


Xenopus laevis , Animals , Algorithms , Kv1.2 Potassium Channel/genetics , Kv1.2 Potassium Channel/metabolism , Kv1.2 Potassium Channel/chemistry , Oocytes/metabolism , Phylogeny , Shab Potassium Channels/metabolism , Shab Potassium Channels/genetics , Shab Potassium Channels/chemistry , Mutation , Xenopus
2.
Transl Res ; 263: 15-27, 2024 01.
Article En | MEDLINE | ID: mdl-37607607

Nerve injury-induced alternations of gene expression in primary sensory neurons of the dorsal root ganglion (DRG) are molecular basis of neuropathic pain genesis. Transcription factors regulate gene expression. In this study, we examined whether early B cell factor 1 (EBF1), a transcription factor, in the DRG, participated in neuropathic pain caused by chronic constriction injury (CCI) of the sciatic nerve. EBF1 was distributed exclusively in the neuronal nucleus and coexpressed with cytoplasmic/membrane Kv1.2 in individual DRG neurons. The expression of Ebf1 mRNA and protein was time-dependently downregulated in the ipsilateral lumbar (L) 3/4 DRGs after unilateral CCI. Rescuing this downregulation through microinjection of the adeno-associated virus 5 expressing full-length Ebf1 mRNA into the ipsilateral L3/4 DRGs reversed the CCI-induced decrease of DRG Kv1.2 expression and alleviated the development and maintenance of mechanical, heat and cold hypersensitivities. Conversely, mimicking the downregulation of DRG EBF1 through microinjection of AAV5-expressing Ebf1 shRNA into unilateral L3/4 DRGs produced a reduction of Kv1.2 expression in the ipsilateral L3/4 DRGs, spontaneous pain, and the enhanced responses to mechanical, heat and cold stimuli in naive mice. Mechanistically, EBF1 not only bound to the Kcna2 gene (encoding Kv1.2) promoter but also directly activated its activity. CCI decreased the EBF1 binding to the Kcna2 promoter in the ipsilateral L3/4 DRGs. Our findings suggest that DRG EBF1 downregulation contributes to neuropathic pain likely by losing its binding to Kcna2 promoter and subsequently silencing Kv1.2 expression in primary sensory neurons. Exogenous EBF1 administration may mitigate neuropathic pain by rescuing DRG Kv1.2 expression.


Neuralgia , Transcription Factors , Animals , Mice , Gene Expression Regulation , Hyperalgesia/genetics , Neuralgia/genetics , RNA, Messenger/metabolism , Sensory Receptor Cells , Transcription Factors/genetics , Kv1.2 Potassium Channel/metabolism
3.
J Chem Inf Model ; 63(10): 3043-3053, 2023 05 22.
Article En | MEDLINE | ID: mdl-37143234

Peptide toxins that adopt the ShK fold can inhibit the voltage-gated potassium channel KV1.3 with IC50 values in the pM range and are therefore potential leads for drugs targeting autoimmune and neuroinflammatory diseases. Nuclear magnetic resonance (NMR) relaxation measurements and pressure-dependent NMR have shown that, despite being cross-linked by disulfide bonds, ShK itself is flexible in solution. This flexibility affects the local structure around the pharmacophore for the KV1.3 channel blockade and, in particular, the relative orientation of the key Lys and Tyr side chains (Lys22 and Tyr23 in ShK) and has implications for the design of KV1.3 inhibitors. In this study, we have performed molecular dynamics (MD) simulations on ShK and a close homologue, HmK, to probe the conformational space occupied by the Lys and Tyr residues, and docked the different conformations with a recently determined cryo-EM structure of the KV1.3 channel. Although ShK and HmK have 60% sequence identity, their dynamic behaviors are quite different, with ShK sampling a broad range of conformations over the course of a 5 µs MD simulation, while HmK is relatively rigid. We also investigated the importance of conformational dynamics, in particular the distance between the side chains of the key dyad Lys22 and Tyr23, for binding to KV1.3. Although these peptides have quite different dynamics, the dyad in both adopts a similar configuration upon binding, revealing a conformational selection upon binding to KV1.3 in the case of ShK. Both peptides bind to KV1.3 with Lys22 occupying the pore of the channel. Intriguingly, the more flexible peptide, ShK, binds with significantly higher affinity than HmK.


Cnidarian Venoms , Sea Anemones , Animals , Kv1.3 Potassium Channel/chemistry , Kv1.3 Potassium Channel/metabolism , Cnidarian Venoms/chemistry , Cnidarian Venoms/metabolism , Cnidarian Venoms/pharmacology , Sea Anemones/chemistry , Sea Anemones/metabolism , Peptides/chemistry , Molecular Conformation , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/chemistry , Kv1.2 Potassium Channel/metabolism
4.
ACS Chem Neurosci ; 13(23): 3342-3351, 2022 12 07.
Article En | MEDLINE | ID: mdl-36417797

Demyelination, the loss of the insulating sheath of neurons, causes failed or slowed neuronal conduction and contributes to the neurological symptoms in multiple sclerosis, traumatic brain and spinal cord injuries, stroke, and dementia. In demyelinated neurons, the axonal potassium channels Kv1.1 and Kv1.2, generally under the myelin sheath, become exposed and upregulated. Therefore, imaging these channels using positron emission tomography can provide valuable information for disease diagnosis and monitoring. Here, we describe a novel tracer for Kv1 channels, [11C]3-methyl-4-aminopyridine ([11C]3Me4AP). [11C]3Me4AP was efficiently synthesized via Pd(0)-Cu(I) comediated Stille cross-coupling of a stannyl precursor containing a free amino group. Evaluation of its imaging properties in rats and nonhuman primates showed that [11C]3Me4AP has a moderate brain permeability and slow kinetics. Additional evaluation in monkeys showed that the tracer is metabolically stable and that a one-tissue compartment model can accurately model the regional brain time-activity curves. Compared to the related tracers [18F]3-fluoro-4-aminopyridine ([18F]3F4AP) and [11C]3-methoxy-4-aminopyridine ([11C]3MeO4AP), [11C]3Me4AP shows lower initial brain uptake, which indicates reduced permeability to the blood-brain barrier and slower kinetics, suggesting higher binding affinity consistent with in vitro studies. While the slow kinetics and strong binding affinity resulted in a tracer with less favorable properties for imaging the brain than its predecessors, these properties may make 3Me4AP useful as a therapeutic.


4-Aminopyridine , Brain , Demyelinating Diseases , Kv1.1 Potassium Channel , Kv1.2 Potassium Channel , Molecular Imaging , Radioactive Tracers , Animals , Rats , 4-Aminopyridine/analogs & derivatives , 4-Aminopyridine/chemical synthesis , 4-Aminopyridine/pharmacokinetics , Brain/diagnostic imaging , Brain/metabolism , Permeability , Kv1.1 Potassium Channel/metabolism , Kv1.2 Potassium Channel/metabolism , Demyelinating Diseases/diagnostic imaging , Molecular Imaging/methods , Primates , Blood-Brain Barrier/metabolism
5.
J Biol Chem ; 298(11): 102467, 2022 11.
Article En | MEDLINE | ID: mdl-36087839

Among voltage-gated potassium channel (KV) isoforms, KV1.6 is one of the most widespread in the nervous system. However, there are little data concerning its physiological significance, in part due to the scarcity of specific ligands. The known high-affinity ligands of KV1.6 lack selectivity, and conversely, its selective ligands show low affinity. Here, we present a designer peptide with both high affinity and selectivity to KV1.6. Previously, we have demonstrated that KV isoform-selective peptides can be constructed based on the simplistic α-hairpinin scaffold, and we obtained a number of artificial Tk-hefu peptides showing selective blockage of KV1.3 in the submicromolar range. We have now proposed amino acid substitutions to enhance their activity. As a result, we have been able to produce Tk-hefu-11 that shows an EC50 of ≈70 nM against KV1.3. Quite surprisingly, Tk-hefu-11 turns out to block KV1.6 with even higher potency, presenting an EC50 of ≈10 nM. Furthermore, we have solved the peptide structure and used molecular dynamics to investigate the determinants of selective interactions between artificial α-hairpinins and KV channels to explain the dramatic increase in KV1.6 affinity. Since KV1.3 is not highly expressed in the nervous system, we hope that Tk-hefu-11 will be useful in studies of KV1.6 and its functions.


Potassium Channels, Voltage-Gated , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Amino Acid Sequence , Potassium Channel Blockers/chemistry , Peptides/chemistry , Ligands , Protein Isoforms/genetics , Protein Isoforms/metabolism , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/metabolism , Kv1.1 Potassium Channel/metabolism , Kv1.2 Potassium Channel/metabolism , Kv1.5 Potassium Channel/metabolism
6.
Proc Natl Acad Sci U S A ; 119(17): e2113675119, 2022 04 26.
Article En | MEDLINE | ID: mdl-35439054

We report on a heterozygous KCNA2 variant in a child with epilepsy. KCNA2 encodes KV1.2 subunits, which form homotetrameric potassium channels and participate in heterotetrameric channel complexes with other KV1-family subunits, regulating neuronal excitability. The mutation causes substitution F233S at the KV1.2 charge transfer center of the voltage-sensing domain. Immunocytochemical trafficking assays showed that KV1.2(F233S) subunits are trafficking deficient and reduce the surface expression of wild-type KV1.2 and KV1.4: a dominant-negative phenotype extending beyond KCNA2, likely profoundly perturbing electrical signaling. Yet some KV1.2(F233S) trafficking was rescued by wild-type KV1.2 and KV1.4 subunits, likely in permissible heterotetrameric stoichiometries: electrophysiological studies utilizing applied transcriptomics and concatemer constructs support that up to one or two KV1.2(F233S) subunits can participate in trafficking-capable heterotetramers with wild-type KV1.2 or KV1.4, respectively, and that both early and late events along the biosynthesis and secretion pathway impair trafficking. These studies suggested that F233S causes a depolarizing shift of ∼48 mV on KV1.2 voltage dependence. Optical tracking of the KV1.2(F233S) voltage-sensing domain (rescued by wild-type KV1.2 or KV1.4) revealed that it operates with modestly perturbed voltage dependence and retains pore coupling, evidenced by off-charge immobilization. The equivalent mutation in the Shaker K+ channel (F290S) was reported to modestly affect trafficking and strongly affect function: an ∼80-mV depolarizing shift, disrupted voltage sensor activation and pore coupling. Our work exposes the multigenic, molecular etiology of a variant associated with epilepsy and reveals that charge-transfer-center disruption has different effects in KV1.2 and Shaker, the archetypes for potassium channel structure and function.


Epilepsy , Cell Membrane/metabolism , Child , Epilepsy/genetics , Epilepsy/metabolism , Humans , Kv1.1 Potassium Channel/genetics , Kv1.2 Potassium Channel/genetics , Kv1.2 Potassium Channel/metabolism , Mutation , Potassium/metabolism , Potassium Channels/metabolism
7.
Bioengineered ; 12(1): 7470-7480, 2021 12.
Article En | MEDLINE | ID: mdl-34632937

Neuropathic pain is a kind of pain caused by damage to somatosensory nervous system. Currently, neuropathic pain is still a medical problem for clinicians. Ubiquitin conjugating enzyme E2B (Ube2b) is validated to be implicated with nerve function, but whether Ube2b can play a role in neuropathic pain is still elusive. In this work, we constructed chronic constriction injury (CCI) rat model by ligating the left sciatic nerve, Ube2b protein expression was confirmed to be decreased in spinal cord tissues of CCI rats via Western blot analysis and immunofluorescence (IF) staining. Moreover, Ube2b elevation alleviated the thermal hyperalgesia and mechanical hyperalgesia in CCI rats according to paw withdrawal thermal latency (PWTL) and paw withdrawal mechanic threshold (PWMT). In addition, Hematoxylin-eosin staining revealed that Ube2b elevation suppressed chronic sciatic nerve injury. All these data suggested that Ube2b could ameliorate neuropathic pain in CCI rats. Mechanically, Ube2b upregulation elevated the protein level of Kcna2 (potassium voltage-gated channel subfamily A member 2) and decreased the protein level of DNMT3a (DNA methyltransferase 3 alpha). Ube2b elevation could increase Kcna2 expression via suppressing DNMT3a. Rescue assays unveiled that Ube2b overexpression modulated-mechanical hyperalgesia and thermal hyperalgesia were reversed by Kcna2 depletion, indicating that Ube2b alleviated neuropathic pain via mediating Kcna2 via the regulation of DNMT3a. In summary, we found that Ube2b elevation ameliorated neuropathic pain through regulating Kcna2, which might offer a novel biomarker for the therapies of neuropathic pain.


Kv1.2 Potassium Channel/genetics , Neuralgia , Neurons, Afferent/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Animals , DNA Methyltransferase 3A/genetics , DNA Methyltransferase 3A/metabolism , Kv1.2 Potassium Channel/metabolism , Male , Neuralgia/genetics , Neuralgia/metabolism , Neuralgia/physiopathology , Rats , Rats, Sprague-Dawley , Ubiquitin-Conjugating Enzymes/metabolism , Up-Regulation/genetics
8.
ACS Appl Mater Interfaces ; 13(24): 27784-27795, 2021 Jun 23.
Article En | MEDLINE | ID: mdl-34126740

Graphene quantum dots (GQDs) are emerging as a versatile nanomaterial with numerous proposed biomedical applications. Despite the explosion in potential applications, the molecular interactions between GQDs and complex biomolecular systems, including potassium-ion (K+) channels, remain largely unknown. Here, we use molecular dynamics (MD) simulations and electrophysiology to study the interactions between GQDs and three representative K+ channels, which participate in a variety of physiological processes and are closely related to many disease states. Using MD simulations, we observed that GQDs adopt distinct contact poses with each of the three structurally distinct K+ channels. Our electrophysiological characterization of the effects of GQDs on channel currents revealed that GQDs interact with the extracellular voltage-sensing domain (VSD) of a Kv1.2 channel, augmenting current by left-shifting the voltage dependence of channel activation. In contrast, GQDs form a "lid" cluster over the extracellular mouth of inward rectifier Kir3.2, blocking the channel pore and decreasing the current in a concentration-dependent manner. Meanwhile, GQDs accumulate on the extracellular "cap domain" of K2P2 channels and have no apparent impact on the K+ flux through the channel. These results reveal a surprising multifaceted regulation of K+ channels by GQDs, which might help de novo design of nanomaterial-based channel probe openers/inhibitors that can be used to further discern channel function.


G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Graphite/metabolism , Kv1.2 Potassium Channel/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Quantum Dots/metabolism , Animals , G Protein-Coupled Inwardly-Rectifying Potassium Channels/chemistry , Graphite/chemistry , HEK293 Cells , Humans , Kv1.2 Potassium Channel/chemistry , Mice , Molecular Dynamics Simulation , Potassium Channels, Tandem Pore Domain/chemistry , Protein Binding , Protein Domains , Quantum Dots/chemistry , Rats
9.
Int J Mol Sci ; 22(6)2021 Mar 10.
Article En | MEDLINE | ID: mdl-33802230

Pathogenic variants in KCNA2, encoding for the voltage-gated potassium channel Kv1.2, have been identified as the cause for an evolving spectrum of neurological disorders. Affected individuals show early-onset developmental and epileptic encephalopathy, intellectual disability, and movement disorders resulting from cerebellar dysfunction. In addition, individuals with a milder course of epilepsy, complicated hereditary spastic paraplegia, and episodic ataxia have been reported. By analyzing phenotypic, functional, and genetic data from published reports and novel cases, we refine and further delineate phenotypic as well as functional subgroups of KCNA2-associated disorders. Carriers of variants, leading to complex and mixed channel dysfunction that are associated with a gain- and loss-of-potassium conductance, more often show early developmental abnormalities and an earlier onset of epilepsy compared to individuals with variants resulting in loss- or gain-of-function. We describe seven additional individuals harboring three known and the novel KCNA2 variants p.(Pro407Ala) and p.(Tyr417Cys). The location of variants reported here highlights the importance of the proline(405)-valine(406)-proline(407) (PVP) motif in transmembrane domain S6 as a mutational hotspot. A novel case of self-limited infantile seizures suggests a continuous clinical spectrum of KCNA2-related disorders. Our study provides further insights into the clinical spectrum, genotype-phenotype correlation, variability, and predicted functional impact of KCNA2 variants.


Databases, Nucleic Acid , Genotype , Kv1.2 Potassium Channel , Mutation, Missense , Nervous System Diseases , Amino Acid Substitution , Female , Humans , Kv1.2 Potassium Channel/genetics , Kv1.2 Potassium Channel/metabolism , Male , Nervous System Diseases/genetics , Nervous System Diseases/metabolism
10.
Int J Biol Macromol ; 178: 143-153, 2021 May 01.
Article En | MEDLINE | ID: mdl-33636268

Novel degraded potassium channel-modulatory peptides were recently found in thermally processed scorpions, but their pharmacological properties remain unclear. Here, we identified a full-length scorpion toxin (i.e., BmKcug2) and its four truncated analogs (i.e., BmKcug2-P1, BmKcug2-P2, BmKcug2-P3 and BmKcug2-P4) with three conserved disulfide bonds in processed scorpion medicinal material by mass spectrometry. The pharmacological experiments revealed that the recombinant BmKcug2 and BmKcug2-P1 could selectively inhibit the human Kv1.2 and human Kv1.3 potassium channels, while the other three analogs showed a much weaker inhibitory effect on potassium channels. BmKcug2 inhibited hKv1.2 and hKv1.3 channels, with IC50 values of 45.6 ± 5.8 nM and 215.2 ± 39.7 nM, respectively, and BmKcug2-P1 inhibited hKv1.2 and hKv1.3, with IC50 values of 89.9 ± 9.6 nM and 1142.4 ± 64.5 nM, respectively. The chromatographic analysis and pharmacological properties of BmKcug2 and BmKcug2-P1 boiled in water for different times further strongly supported their good thermal stability. Structural and functional dissection indicated that one amino acid, i.e., Tyr36, determined the differential affinities of BmKcug2 and four BmKcug2 analogs. Altogether, this research investigated the different pharmacological properties of BmKcug2 and its truncated analogs, and the findings highlighted the diversity of K+ channel blockers from various scorpion species through thermal processing.


Kv1.2 Potassium Channel/metabolism , Kv1.3 Potassium Channel/metabolism , Potassium Channel Blockers/pharmacology , Scorpion Venoms/pharmacology , Animals , HEK293 Cells , Humans , Scorpions
11.
J Neurochem ; 156(3): 367-378, 2021 02.
Article En | MEDLINE | ID: mdl-32621322

Voltage-gated potassium channels (Kv) are important regulators of neuronal excitability for its role of regulating resting membrane potential and repolarization. Recent studies show that Kv channels participate in neuropathic pain, but the detailed underlying mechanisms are far from being clear. In this study, we used siRNA, miR-137 agomir, and antagomir to regulate the expression of Kv1.2 in spinal cord and dorsal root ganglia (DRG) of naïve and chronic constriction injury (CCI) rats. Kv currents and neuron excitability in DRG neurons were examined by patch-clamp whole-cell recording to verify the change in Kv1.2 function. The results showed that Kv1.2 was down-regulated in DRG and spinal dorsal horn (SDH) by CCI. Knockdown of Kv1.2 by intrathecally injecting Kcna2 siRNA induced significant mechanical and thermal hypersensitivity in naïve rats. Concomitant with the down-regulation of Kv1.2 was an increase in the expression of the miR-137. The targeting and regulating of miR-137 on Kcna2 was verified by dual-luciferase reporter system and intrathecal injecting miR-137 agomir. Furthermore, rescuing the expression of Kv1.2 in CCI rats, achieved through inhibiting miR-137, restored the abnormal Kv currents and excitability in DRG neurons, and alleviated mechanical allodynia and thermal hyperalgesia. These results indicate that the miR-137-mediated Kv1.2 impairment is a crucial etiopathogenesis for the nerve injury-induced neuropathic pain and can be a novel potential therapeutic target for neuropathic pain management.


Kv1.2 Potassium Channel/metabolism , Neuralgia/metabolism , Peripheral Nerve Injuries/metabolism , Animals , Epigenesis, Genetic , Ganglia, Spinal/metabolism , Male , MicroRNAs/metabolism , Neuralgia/etiology , Neurons/metabolism , Peripheral Nerve Injuries/complications , Rats , Rats, Sprague-Dawley , Sciatic Nerve/injuries , Spinal Cord Dorsal Horn/metabolism
12.
BMC Cardiovasc Disord ; 20(1): 337, 2020 07 14.
Article En | MEDLINE | ID: mdl-32664860

BACKGROUND: High blood glucose impairs voltage-gated K+ (Kv) channel-mediated vasodilation in rat coronary artery smooth muscle cells (CSMCs) via oxidative stress. Advanced glycation end product (AGE) and receptor for AGE (RAGE) axis has been found to impair coronary dilation by reducing Kv channel activity in diabetic rat small coronary arteries (RSCAs). However, its underlying mechanism remain unclear. Here, we used isolated arteries and primary CSMCs to investigate the effect of AGE incubation on Kv channel-mediated coronary dilation and the possible involvement of peroxisome proliferators-activated receptor (PPAR) -γ pathway. METHODS: The RSCAs and primary CSMCs were isolated, cultured, and treated with bovine serum albumin (BSA), AGE-BSA, alagrebrium (ALA, AGE cross-linking breaker), pioglitazone (PIO, PPAR-γ activator) and/or GW9662 (PPAR-γ inhibitor). The groups were accordingly divided as control, BSA, AGE, AGE + ALA, AGE + PIO, or AGE + PIO + GW9662. Kv channel-mediated dilation was analyzed using wire myograph. Histology and immunohistochemistry of RSCAs were performed. Western blot was used to detect the protein expression of RAGE, major Kv channel subunits expressed in CSMCs (Kv1.2 and Kv1.5), PPAR-γ, and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-2 (NOX-2). RESULTS: AGE markedly reduced Forskolin-induced Kv channel-mediated dilation of RSCAs by engaging with RAGE, and ALA or PIO significantly reversed the functional loss of Kv channel. In both RSCAs and CSMCs, AGE reduced Kv1.2/1.5 expression, increased RAGE and NOX-2 expression, and inhibited PPAR-γ expression, while ALA or PIO treatment partially reversed the inhibiting effects of AGE on Kv1.2/1.5 expression, accompanied by the downregulation of RAGE and decreased oxidative stress. Meanwhile, silencing of RAGE with siRNA remarkably alleviated the AGE-induced downregulation of Kv1.2/1.5 expression in CSMCs. CONCLUSION: AGE reduces the Kv channel expression in CSMCs and further impairs the Kv channel-mediated dilation in RSCAs. The AGE/RAGE axis may enhance oxidative stress by inhibiting the downstream PPAR-γ pathway, thus playing a critical role in the dysfunction of Kv channels.


Glycation End Products, Advanced/pharmacology , Kv1.2 Potassium Channel/metabolism , Kv1.5 Potassium Channel/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , PPAR gamma/metabolism , Serum Albumin, Bovine/pharmacology , Vasodilation/drug effects , Anilides/pharmacology , Animals , Cells, Cultured , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Kv1.2 Potassium Channel/genetics , Kv1.5 Potassium Channel/genetics , Male , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Oxidative Stress/drug effects , PPAR gamma/drug effects , Pioglitazone/pharmacology , Rats, Sprague-Dawley , Signal Transduction
13.
Eur J Pharmacol ; 882: 173281, 2020 Sep 05.
Article En | MEDLINE | ID: mdl-32562800

Voltage-gated potassium (Kv) channels are the largest superfamily of potassium (K) channels. A variety of Kv channels are expressed in the vascular smooth muscle cells (SMC). Studies have shown that gestational diabetes mellitus (GDM) and pregnancy-induced hypertension (PIH) cause various changes in the human umbilical vein (HUV). Recently, we have shown that 4-AP, a nonspecific Kv1-4 channel inhibitor, significantly decreases vasorelaxation induced by K channel opener pinacidil in vascular SMCs of the HUVs from normal pregnancies, but not in GDM and PIH. The goal of this study was to provide more detailed insight in the Kv channel subtypes involved in pinacidil-induced vasodilation of HUVs, as well as to investigate potential alterations of their function and expression during GDM and PIH. Margatoxin, a specific blocker of Kv1.2 and Kv1.3 channels, significantly antagonized pinacidil-induced vasorelaxation in normal pregnancy, while in HUVs from GDM and PIH that was not the case, indicating damage of Kv1.2 and Kv1.3 channel function. Immunohistochemistry and Western blot revealed similar expression of Kv1.2 channels in all groups. The expression of Kv1.3 subunit was significantly decreased in PIH, while it remained unchanged in GDM compared to normal pregnancy. Phrixotoxin, specific blocker of Kv4.2 and Kv4.3 channels, did not antagonize response to pinacidil in any of the groups. The major novel findings show that margatoxin antagonized pinacidil-induced relaxation in normal pregnancy, but not in GDM and PIH. Decreased expression of Kv1.3 channels in HUV during PIH may be important pathophysiological mechanism contributing to an increased risk of adverse pregnancy outcomes.


Hypertension, Pregnancy-Induced/metabolism , Kv1.3 Potassium Channel/metabolism , Muscle, Smooth, Vascular/metabolism , Umbilical Veins/metabolism , Adult , Antihypertensive Agents/pharmacology , Diabetes, Gestational/metabolism , Female , Humans , Kv1.2 Potassium Channel/metabolism , Pinacidil/pharmacology , Pregnancy , Young Adult
14.
Sci Rep ; 10(1): 6954, 2020 04 24.
Article En | MEDLINE | ID: mdl-32332769

Low threshold voltage activated Kv1 potassium channels play key roles in regulating action potential (AP) threshold, neural excitability, and synaptic transmission. Kv1 channels are highly expressed in the cerebellum and mutations of human Kv1 genes are associated to episodic forms of ataxia (EAT-1). Besides the well-established role of Kv1 channels in controlling the cerebellar basket-Purkinje cells synapses, Kv1 channels are expressed by the deep cerebellar nuclear neurons (DCNs) where they regulate the activity of principal DCNs carrying the cerebellar output. DCNs include as well GABAergic neurons serving important functions, such as those forming the inhibitory nucleo-olivary pathway, the nucleo-cortical DCNs providing feed-back inhibition to the cerebellar cortex, and those targeting principal DCNs, but whether their function is regulated by Kv1 channels remains unclear. Here, using cerebellar slices from mature GAD67-GFP mice to identify putative GABAergic-DCNs (GAD + DCN) we show that specific Kv1 channel blockers (dendrotoxin-alpha/I/K, DTXs) hyperpolarized the threshold of somatic action potentials, increased the spontaneous firing rate and hampered evoked high frequency repetitive responses of GAD + DCNs. Moreover, DTXs induced somatic depolarization and tonic firing in previously silent, putative nucleo-cortical DCNs. These results reveal a novel role of Kv1 channels in regulating GABAergic-DCNs activity and thereby, cerebellar function at multiple levels.


Action Potentials/physiology , Cerebellar Nuclei/metabolism , GABAergic Neurons/metabolism , Kv1.1 Potassium Channel/metabolism , Kv1.2 Potassium Channel/metabolism , Shaker Superfamily of Potassium Channels/metabolism , Animals , Cerebellar Nuclei/cytology , Kv1.1 Potassium Channel/genetics , Kv1.2 Potassium Channel/genetics , Mice , Neurons/cytology , Neurons/metabolism
15.
J Gen Physiol ; 152(6)2020 06 01.
Article En | MEDLINE | ID: mdl-32110806

Slow inactivation has been described in multiple voltage-gated K+ channels and in great detail in the Drosophila Shaker channel. Structural studies have begun to facilitate a better understanding of the atomic details of this and other gating mechanisms. To date, the only voltage-gated potassium channels whose structure has been solved are KvAP (x-ray diffraction), the KV1.2-KV2.1 "paddle" chimera (x-ray diffraction and cryo-EM), KV1.2 (x-ray diffraction), and ether-à-go-go (cryo-EM); however, the structural details and mechanisms of slow inactivation in these channels are unknown or poorly characterized. Here, we present a detailed study of slow inactivation in the rat KV1.2 channel and show that it has some properties consistent with the C-type inactivation described in Shaker. We also study the effects of some mutations that are known to modulate C-type inactivation in Shaker and show that qualitative and quantitative differences exist in their functional effects, possibly underscoring subtle but important structural differences between the C-inactivated states in Shaker and KV1.2.


Ion Channel Gating , Kv1.2 Potassium Channel , Animals , Kv1.2 Potassium Channel/metabolism , Rats , Xenopus laevis/metabolism
16.
J Phys Chem B ; 123(38): 7984-7998, 2019 09 26.
Article En | MEDLINE | ID: mdl-31441655

Ion channels in cell membranes control entry and exit of ions; their gating (opening and closing) is key to their functioning. It is known that protons can pass through the voltage-sensing domain (VSD) of channels such as Kv1.2. Quantum calculations for a section of the VSD show the steps protons take in responding to voltage and show no major displacement of the protein backbone with voltage change; 70 amino acids are included, 42 with side chains (9 directly in the proton path), 28 as backbone only, and 24 water molecules. Protons provide much of the gating current, the capacitative current immediately preceding channel opening with significant additional contributions from charge transfer to other groups. Most gating models, in contrast, require major protein displacement during gating. Energy terms without classical analogues (exchange plus correlation energy, which are greater than thermal energy) show that quantum calculations are required. Energy as a function of voltage for a key proton transfer leads to, approximately, the correct voltage for channel opening. Calculated total charge transfer (not only protons) for gating is reasonable compared to experimental values. We are also able to account, at least qualitatively, for two mutations, one with the gating current curve left-shifted and one right-shifted, and show the alternate proton paths that are required to account for these.


Kv1.2 Potassium Channel/chemistry , Quantum Theory , Animals , Arginine/chemistry , Crystallography, X-Ray , Hydrogen Bonding , Kv1.2 Potassium Channel/genetics , Kv1.2 Potassium Channel/metabolism , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protons , Rats , Water/chemistry
17.
Proc Natl Acad Sci U S A ; 116(37): 18700-18709, 2019 09 10.
Article En | MEDLINE | ID: mdl-31444298

Voltage-dependent potassium channels (Kvs) gate in response to changes in electrical membrane potential by coupling a voltage-sensing module with a K+-selective pore. Animal toxins targeting Kvs are classified as pore blockers, which physically plug the ion conduction pathway, or as gating modifiers, which disrupt voltage sensor movements. A third group of toxins blocks K+ conduction by an unknown mechanism via binding to the channel turrets. Here, we show that Conkunitzin-S1 (Cs1), a peptide toxin isolated from cone snail venom, binds at the turrets of Kv1.2 and targets a network of hydrogen bonds that govern water access to the peripheral cavities that surround the central pore. The resulting ectopic water flow triggers an asymmetric collapse of the pore by a process resembling that of inherent slow inactivation. Pore modulation by animal toxins exposes the peripheral cavity of K+ channels as a novel pharmacological target and provides a rational framework for drug design.


Cell Membrane/drug effects , Drosophila Proteins/antagonists & inhibitors , Ion Channel Gating/drug effects , Kv1.2 Potassium Channel/antagonists & inhibitors , Mollusk Venoms/toxicity , Shaker Superfamily of Potassium Channels/antagonists & inhibitors , Animals , Cell Membrane/metabolism , Crystallography, X-Ray , Drosophila Proteins/genetics , Drosophila Proteins/isolation & purification , Drosophila Proteins/metabolism , Drug Design , Female , Hydrogen Bonding/drug effects , Kv1.2 Potassium Channel/genetics , Kv1.2 Potassium Channel/isolation & purification , Kv1.2 Potassium Channel/metabolism , Lethal Dose 50 , Molecular Docking Simulation , Molecular Dynamics Simulation , Mollusk Venoms/chemistry , Mutation , Oocytes , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Shaker Superfamily of Potassium Channels/genetics , Shaker Superfamily of Potassium Channels/isolation & purification , Shaker Superfamily of Potassium Channels/metabolism , Water/chemistry , Water/metabolism , Xenopus laevis
18.
J Neurosci ; 39(33): 6595-6607, 2019 08 14.
Article En | MEDLINE | ID: mdl-31182635

Expressional changes of pain-associated genes in primary sensory neurons of DRG are critical for neuropathic pain genesis. DNA methyltransferase (DNMT)-triggered DNA methylation silences gene expression. We show here that DNMT1, a canonical maintenance methyltransferase, acts as the de novo DNMT and is required for neuropathic pain genesis likely through repressing at least DRG Kcna2 gene expression in male mice. Peripheral nerve injury upregulated DNMT1 expression in the injured DRG through the transcription factor cAMP response element binding protein-triggered transcriptional activation of Dnmt1 gene. Blocking this upregulation prevented nerve injury-induced DNA methylation within the promoter and 5'-untranslated region of Kcna2 gene, rescued Kcna2 expression and total Kv current, attenuated hyperexcitability in the injured DRG neurons, and alleviated nerve injury-induced pain hypersensitivities. Given that Kcna2 is a key player in neuropathic pain, our findings suggest that DRG DNMT1 may be a potential target for neuropathic pain management.SIGNIFICANCE STATEMENT In the present study, we reported that DNMT1, a canonical DNA maintenance methyltransferase, is upregulated via the activation of the transcription factor CREB in the injured DRG after peripheral nerve injury. This upregulation was responsible for nerve injury-induced de novo DNA methylation within the promoter and 5'-untranslated region of the Kcna2 gene, reductions in Kcna2 expression and Kv current and increases in neuronal excitability in the injured DRG. Since pharmacological inhibition or genetic knockdown of DRG DNMT1 alleviated nerve injury-induced pain hypersensitivities, DRG DNMT1 contributes to neuropathic pain genesis partially through repression of DRG Kcna2 gene expression.


DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Epigenetic Repression/physiology , Kv1.2 Potassium Channel/metabolism , Neuralgia/metabolism , Neurons, Afferent/metabolism , Animals , Ganglia, Spinal/metabolism , Male , Mice , Mice, Inbred C57BL , Peripheral Nerve Injuries/metabolism
19.
Physiol Rep ; 7(12): e14147, 2019 07.
Article En | MEDLINE | ID: mdl-31222975

Expression of Kv1.2 within Kv1.x potassium channel complexes is critical in maintaining appropriate neuronal excitability and determining the threshold for action potential firing. This is attributed to the interaction of Kv1.2 with a hitherto unidentified protein that confers bimodal channel activation gating, allowing neurons to adapt to repetitive trains of stimulation and protecting against hyperexcitability. One potential protein candidate is the sigma-1 receptor (Sig-1R), which regulates other members of the Kv1.x channel family; however, the biophysical nature of the interaction between Sig-1R and Kv1.2 has not been elucidated. We hypothesized that Sig-1R may regulate Kv1.2 and may further act as the unidentified modulator of Kv1.2 activation. In transiently transfected HEK293 cells, we found that ligand activation of the Sig-1R modulates Kv1.2 current amplitude. More importantly, Sig-1R interacts with Kv1.2 in baseline conditions to influence bimodal activation gating. These effects are abolished in the presence of the auxiliary subunit Kvß2 and when the Sig-1R mutation underlying ALS16 (Sig-1R-E102Q), is expressed. These data suggest that Kvß2 occludes the interaction of Sig-1R with Kv1.2, and that E102 may be a residue critical for Sig-1R modulation of Kv1.2. The results of this investigation describe an important new role for Sig-1R in the regulation of neuronal excitability and introduce a novel mechanism of pathophysiology in Sig-1R dysfunction.


Kv1.2 Potassium Channel/physiology , Receptors, sigma/physiology , Cells, Cultured , Electrophysiological Phenomena/drug effects , Electrophysiological Phenomena/physiology , HEK293 Cells , Humans , Ion Channel Gating/physiology , Kv1.2 Potassium Channel/drug effects , Kv1.2 Potassium Channel/metabolism , Patch-Clamp Techniques/methods , Phenazocine/analogs & derivatives , Phenazocine/antagonists & inhibitors , Phenazocine/pharmacology , Receptors, sigma/agonists , Receptors, sigma/metabolism , Shaker Superfamily of Potassium Channels/physiology , Sigma-1 Receptor
20.
Nan Fang Yi Ke Da Xue Xue Bao ; 39(5): 579-585, 2019 May 30.
Article Zh | MEDLINE | ID: mdl-31140423

OBJECTIVE: To investigate the changes in the expression of voltage-gated potassium channel subunit KCNA2 in the dorsal root ganglion (DRG) neurons of rats with osteoarthritis (OA) pain induced by sodium monoiodoacetate and explore the mechanism. METHODS: A total of 156 adult male Sprague-Dawley rats were randomly divided into blank control group, saline group and intra-articular monoiodoacetate injection-induced OA group. The paw withdrawal mechanical threshold (PWMT) was measured before and at 1, 2, 4, and 6 weeks after monoiodoacetate injection. At 4 weeks after the injection, the pathological changes in the knee joints were analyzed using HE staining and Safranin O-Fast Green staining, and the expression of activating transcription factor 3 (ATF-3) and inducible nitric oxide synthase (iNOS) in the DRG neurons were detected by immunofluorescence staining. The expression of Kcna2 mRNA in the DRG neurons was detected by RT-qPCR at 1, 2, 4 and 6 weeks after the injection. The expression of KCNA2 in the DRG was measured by Western blotting, and the methylation level of Kcna2 promoter region was measured by MSPCR at 4 weeks after the injection. RESULTS: The PWMT of the rats in OA group was significantly decreased at 2, 4, and 6 weeks after the injection as compared with the baseline (P < 0.05 or P < 0.001) as well as the control group (P < 0.05 or P < 0.001). Four weeks after the intra-articular injection, fractures and defects on the surface of the articular cartilage, bone hyperplasia, and blurred tidal line were observed in the rats in OA group, but no obvious pathological changes were detected in the control or saline groups. Compared with those in the control group, the expressions of ATF-3 and iNOS were significantly increased (P < 0.01) at 4 weeks after injection; the expression of Kcna2 mRNA at 2, 4 and 6 weeks and the expression of KCNA2 protein at 4 weeks were all significantly decreased (P < 0.05 or P < 0.01), and the methylation level of Kcna2 gene was significantly increased at 4 weeks after the injection in OA group (P < 0.01). CONCLUSIONS: The expression of KCNA2 is decreased in the DRG neurons of rats with OA pain likely as a result of enhanced methylation of Kcna2 promoter region.


Ganglia, Spinal , Kv1.2 Potassium Channel/metabolism , Osteoarthritis , Pain , Animals , Disease Models, Animal , Knee Joint , Male , Osteoarthritis/complications , Osteoarthritis/metabolism , Pain/etiology , Pain/metabolism , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley
...