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 Gen Physiol ; 156(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38832889

ABSTRACT

Voltage-gated ion channels are responsible for the electrical excitability of neurons and cardiomyocytes. Thus, they are obvious targets for pharmaceuticals aimed to modulate excitability. Compounds activating voltage-gated potassium (KV) channels are expected to reduce excitability. To search for new KV-channel activators, we performed a high-throughput screen of 10,000 compounds on a specially designed Shaker KV channel. Here, we report on a large family of channel-activating compounds with a carboxyl (COOH) group as the common motif. The most potent COOH activators are lipophilic (4 < LogP <7) and are suggested to bind at the interface between the lipid bilayer and the channel's positively charged voltage sensor. The negatively charged form of the COOH-group compounds is suggested to open the channel by electrostatically pulling the voltage sensor to an activated state. Several of the COOH-group compounds also activate the therapeutically important KV7.2/7.3 channel and can thus potentially be developed into antiseizure drugs. The COOH-group compounds identified in this study are suggested to act via the same site and mechanism of action as previously studied COOH-group compounds, such as polyunsaturated fatty acids and resin acids, but distinct from sites for several other types of potassium channel-activating compounds.


Subject(s)
Ion Channel Gating , Animals , Ion Channel Gating/drug effects , Shaker Superfamily of Potassium Channels/metabolism , KCNQ2 Potassium Channel/metabolism , KCNQ2 Potassium Channel/agonists , Potassium Channels, Voltage-Gated/metabolism , Potassium Channels, Voltage-Gated/drug effects , KCNQ3 Potassium Channel/metabolism , Humans , Xenopus laevis
2.
Brain ; 144(9): 2863-2878, 2021 10 22.
Article in English | MEDLINE | ID: mdl-33768249

ABSTRACT

Spreading depolarization is a slowly propagating wave of massive cellular depolarization associated with acute brain injury and migraine aura. Genetic studies link depolarizing molecular defects in Ca2+ flux, Na+ current in interneurons, and glial Na+-K+ ATPase with spreading depolarization susceptibility, emphasizing the important roles of synaptic activity and extracellular ionic homeostasis in determining spreading depolarization threshold. In contrast, although gene mutations in voltage-gated potassium ion channels that shape intrinsic membrane excitability are frequently associated with epilepsy susceptibility, it is not known whether epileptogenic mutations that regulate membrane repolarization also modify spreading depolarization threshold and propagation. Here we report that the Kcnq2/Kv7.2 potassium channel subunit, frequently mutated in developmental epilepsy, is a spreading depolarization modulatory gene with significant control over the seizure-spreading depolarization transition threshold, bi-hemispheric cortical expression, and diurnal temporal susceptibility. Chronic DC-band cortical EEG recording from behaving conditional Kcnq2 deletion mice (Emx1cre/+::Kcnq2flox/flox) revealed spontaneous cortical seizures and spreading depolarization. In contrast to the related potassium channel deficient model, Kv1.1-KO mice, spontaneous cortical spreading depolarizations in Kcnq2 cKO mice are tightly coupled to the terminal phase of seizures, arise bilaterally, and are observed predominantly during the dark phase. Administration of the non-selective Kv7.2 inhibitor XE991 to Kv1.1-KO mice partly reproduced the Kcnq2 cKO-like spreading depolarization phenotype (tight seizure coupling and bilateral symmetry) in these mice, indicating that Kv7.2 currents can directly and actively modulate spreading depolarization properties. In vitro brain slice studies confirmed that Kcnq2/Kv7.2 depletion or pharmacological inhibition intrinsically lowers the cortical spreading depolarization threshold, whereas pharmacological Kv7.2 activators elevate the threshold to multiple depolarizing and hypometabolic spreading depolarization triggers. Together these results identify Kcnq2/Kv7.2 as a distinctive spreading depolarization regulatory gene, and point to spreading depolarization as a potentially significant pathophysiological component of KCNQ2-linked epileptic encephalopathy syndromes. Our results also implicate KCNQ2/Kv7.2 channel activation as a potential adjunctive therapeutic target to inhibit spreading depolarization incidence.


Subject(s)
Brain/physiology , Cortical Spreading Depression/physiology , KCNQ2 Potassium Channel/metabolism , Nerve Tissue Proteins/metabolism , Anilides/pharmacology , Animals , Brain/drug effects , Bridged Bicyclo Compounds/pharmacology , Carbamates/pharmacology , Cortical Spreading Depression/drug effects , KCNQ2 Potassium Channel/agonists , KCNQ2 Potassium Channel/deficiency , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/deficiency , Organ Culture Techniques , Phenylenediamines/pharmacology
3.
J Assoc Res Otolaryngol ; 22(2): 127-139, 2021 04.
Article in English | MEDLINE | ID: mdl-33575914

ABSTRACT

Exposure to loud noise can cause hearing loss and tinnitus in mice and humans. In mice, one major underlying mechanism of noise-induced tinnitus is hyperactivity of auditory brainstem neurons, due at least in part, to decreased Kv7.2/3 (KCNQ2/3) potassium channel activity. In our previous studies, we used a reflex-based mouse model of tinnitus and showed that administration of a non-specific KCNQ channel activator, immediately after noise trauma, prevented the development of noise-induced tinnitus, assessed 1 week after trauma. Subsequently, we developed RL-81, a very potent and highly specific activator of KCNQ2/3 channels. Here, to test the timing window within which RL-81 prevents tinnitus in mice, we modified and employed an operant animal model of tinnitus, where mice are trained to move in response to sound but not move in silence. Mice with behavioral evidence of tinnitus are expected to move in silence. We validated this mouse model by testing the effect of salicylate, which is known to induce tinnitus. We found that transient administration of RL-81 1 week after noise exposure did not affect hearing loss but reduced significantly the percentage of mice with behavioral evidence of tinnitus, assessed 2 weeks after noise exposure. Our results indicate that RL-81 is a promising drug candidate for further development for the treatment of noise-induced tinnitus.


Subject(s)
Hearing Loss , KCNQ2 Potassium Channel/agonists , KCNQ3 Potassium Channel/agonists , Noise/adverse effects , Tinnitus , Animals , Hearing Loss/drug therapy , Hearing Loss/etiology , Mice , Tinnitus/drug therapy , Tinnitus/etiology
4.
J Pharmacol Exp Ther ; 377(1): 20-28, 2021 04.
Article in English | MEDLINE | ID: mdl-33431609

ABSTRACT

Treatment of chronic pain remains an unmet medical need. The neuronal voltage-gated potassium Kv7/KCNQ/M channel has been implicated as a therapeutic target for chronic pain. However, whether pharmacological activation of the Kv7 channel can alleviate pain remains elusive. In this study, we show that selective activation of native M-currents by a novel channel opener SCR2682 reduces repetitive firings of dorsal root ganglia (DRG) sensory neurons. Intraperitoneal administration of SCR2682 relieves mechanical allodynia and thermal hyperalgesia in rat models of pain induced by complete Freund's adjuvant (CFA) or spared nerve injury (SNI) in a dose-dependent manner without affecting locomotor activity. The antinociceptive efficacy of SCR2682 can be reversed by the channel-specific blocker XE991. Furthermore, SCR2682 increases Kv7.2/KCNQ2 mRNA and protein expression in DRG neurons from rats in the SNI model of neuropathic pain. Taken together, pharmacological activation of neuronal Kv7 channels by opener SCR2682 can alleviate pain in rats, thus possessing therapeutic potential for chronic pain or hyperexcitability-related neurologic disorders. SIGNIFICANCE STATEMENT: A novel voltage-gated potassium Kv7 channel opener SCR2682 inhibits action potential firings in dorsal root ganglia sensory neurons and exhibits efficacy in antinociception, thus possessing a developmental potential for treatment of chronic pain or epilepsy.


Subject(s)
Analgesics/therapeutic use , Chronic Pain/drug therapy , KCNQ2 Potassium Channel/metabolism , Membrane Transport Modulators/therapeutic use , Pyridines/therapeutic use , Action Potentials , Analgesics/pharmacology , Animals , Cells, Cultured , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/physiology , KCNQ2 Potassium Channel/agonists , Male , Membrane Transport Modulators/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley
5.
Cell Res ; 31(1): 52-61, 2021 01.
Article in English | MEDLINE | ID: mdl-32884139

ABSTRACT

The voltage-gated potassium channel KCNQ2 is responsible for M-current in neurons and is an important drug target to treat epilepsy, pain and several other diseases related to neuronal hyper-excitability. A list of synthetic compounds have been developed to directly activate KCNQ2, yet our knowledge of their activation mechanism is limited, due to lack of high-resolution structures. Here, we report cryo-electron microscopy (cryo-EM) structures of the human KCNQ2 determined in apo state and in complex with two activators, ztz240 or retigabine, which activate KCNQ2 through different mechanisms. The activator-bound structures, along with electrophysiology analysis, reveal that ztz240 binds at the voltage-sensing domain and directly stabilizes it at the activated state, whereas retigabine binds at the pore domain and activates the channel by an allosteric modulation. By accurately defining ligand-binding sites, these KCNQ2 structures not only reveal different ligand recognition and activation mechanisms, but also provide a structural basis for drug optimization and design.


Subject(s)
KCNQ2 Potassium Channel/metabolism , Ligands , Action Potentials/drug effects , Benzamides/chemistry , Benzamides/metabolism , Benzamides/pharmacology , Binding Sites , Carbamates/chemistry , Carbamates/metabolism , Carbamates/pharmacology , Cryoelectron Microscopy , Humans , KCNQ2 Potassium Channel/agonists , KCNQ2 Potassium Channel/chemistry , KCNQ2 Potassium Channel/genetics , Molecular Dynamics Simulation , Mutagenesis , Phenylenediamines/chemistry , Phenylenediamines/metabolism , Phenylenediamines/pharmacology , Protein Binding , Protein Structure, Tertiary , Pyridines/chemistry , Pyridines/metabolism , Pyridines/pharmacology
6.
Eur J Pharmacol ; 887: 173440, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32745603

ABSTRACT

Activation of the voltage-gated Kv7 channels holds therapeutic promise in several neurological and psychiatric disorders, including epilepsy, schizophrenia, and depression. Here, we present a pharmacological characterization of Lu AA41178, a novel, pan-selective Kv7.2-7.5 opener, using both in vitro assays and a broad range of in vivo assays with relevance to epilepsy, schizophrenia, and depression. Electrophysiological characterization in Xenopus oocytes expressing human Kv7.2-Kv7.5 confirmed Lu AA41178 as a pan-selective opener of Kv7 channels by significantly left-shifting the activation threshold. Additionally, Lu AA41178 was tested in vitro for off-target effects, demonstrating a clean Kv7-selective profile, with no impact on common cardiac ion channels, and no potentiating activity on GABAA channels. Lu AA41178 was evaluated across preclinical in vivo assays with relevance to neurological and psychiatric disorders. In the maximum electroshock seizure threshold test and PTZ seizure threshold test, Lu AA41178 significantly increased the seizure thresholds in mice, demonstrating anticonvulsant efficacy. Lu AA41178 demonstrated antipsychotic-like activity by reducing amphetamine-induced hyperlocomotion in mice as well as lowering conditioned avoidance responses in rats. In the mouse forced swim test, a model with antidepressant predictivity, Lu AA41178 significantly reduced immobility. Additionally, behavioral effects typically observed with Kv7 openers was also characterized. In vivo assays were accompanied by plasma and brain exposures, revealing minimum effective plasma levels <1000 ng/ml. Lu AA41178, a potent opener of neuronal Kv7 channels demonstrate efficacy in assays of epilepsy, schizophrenia and depression and might serve as a valuable tool for exploring the role of Kv7 channels in both neurological and psychiatric disorders.


Subject(s)
Brain/drug effects , Disease Models, Animal , KCNQ2 Potassium Channel/agonists , Mental Disorders/drug therapy , Seizures/drug therapy , Animals , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Brain/metabolism , Dose-Response Relationship, Drug , Female , Humans , KCNQ2 Potassium Channel/metabolism , Male , Mental Disorders/metabolism , Mental Disorders/psychology , Mice , Mice, Inbred C57BL , Psychotropic Drugs/pharmacology , Psychotropic Drugs/therapeutic use , Rats , Rats, Wistar , Seizures/metabolism , Seizures/psychology , Treatment Outcome , Xenopus laevis
7.
Commun Biol ; 3(1): 356, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32641720

ABSTRACT

Many commonly consumed plants are used as folk medicines, often with unclear molecular mechanisms. Recent studies uncovered the ubiquitous and influential KCNQ family of voltage-gated potassium (Kv) channels as a therapeutic target for several medicinal plant compounds. Capers - immature flower buds of Capparis spinosa - have been consumed for food and medicinal purposes for millennia. Here, we show that caper extract hyperpolarizes cells expressing KCNQ1 or KCNQ2/3 Kv channels. Capers are the richest known natural source of quercetin, the most consumed dietary flavonoid. Quercetin potentiated KCNQ1/KCNE1, KCNQ2/3 and KCNQ4 currents but, unusually, not KCNQ5. Strikingly, quercetin augmented both activation and inactivation of KCNQ1, via a unique KCNQ activation mechanism involving sites atop the voltage sensor and in the pore. The findings uncover a novel potential molecular basis for therapeutic effects of quercetin-rich foods and a new chemical space for atypical modes of KCNQ channel modulation.


Subject(s)
KCNQ Potassium Channels/agonists , Quercetin/pharmacology , Animals , Binding Sites , Capparis/chemistry , KCNQ Potassium Channels/chemistry , KCNQ2 Potassium Channel/agonists , KCNQ3 Potassium Channel/agonists , Oocytes , Patch-Clamp Techniques , Plant Extracts/pharmacology , Protein Structure, Tertiary , Rutin/pharmacology , Xenopus laevis
8.
Neuropharmacology ; 168: 108012, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32067988

ABSTRACT

Although the lateral habenula (LHb) is involved in the regulation of multiple brain functions and this region expresses abundant M-type potassium channel (M-channel) subunits Kv7.2 and Kv7.3, the role of M-channels in regulating working memory is unclear, particularly in Parkinson's disease (PD). Here we tested the effects of activation and blockade of LHb M-channels on working memory by the T-maze rewarded alternation test in rats with unilateral 6-hydroxydopamine lesions of the substantia nigra compacta (SNc). The SNc lesion induced working memory impairment, increased the firing rate of LHb neurons, decreased dopamine (DA) level in the ventral medial prefrontal cortex (vmPFC) and reduced the expression of Kv7.2 subunit in the LHb. Intra-LHb injection of M-channel activator retigabine induced enhancement of working memory in SNc sham-lesioned and SNc-lesioned rats; conversely, the injection of M-channel blocker XE-991 impaired working memory in the two groups of rats. However, doses producing significant effects in SNc-lesioned rats were higher than those in SNc sham-lesioned rats. Further, intra-LHb injection of retigabine decreased the firing rate of LHb neurons and increased release of DA and serotonin (5-HT) in the vmPFC, while XE-991 increased the firing rate and decreased DA and 5-HT release in the two groups of rats. Compared with SNc sham-lesioned rats, the duration of M-channel activation and blockade action on the firing rate of the neurons and release of DA and 5-HT was significantly shortened in SNc-lesioned rats, which was consistent with reduced expression of Kv7.2 subunit in the LHb after lesioning the SNc. Collectively, these findings suggest involvement of LHb Kv7.2 subunit-containing M-channels in working memory impairment in SNc-lesioned rats, and that enhanced or impaired working memory after activation or blockade of M-channels in the LHb is related to the changes in the firing activity of LHb neurons and DA and 5-HT release in the vmPFC.


Subject(s)
Habenula/metabolism , KCNQ2 Potassium Channel/biosynthesis , Memory, Short-Term/physiology , Parkinsonian Disorders/metabolism , Animals , Habenula/drug effects , KCNQ2 Potassium Channel/agonists , KCNQ2 Potassium Channel/antagonists & inhibitors , Male , Membrane Transport Modulators/pharmacology , Memory, Short-Term/drug effects , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced , Potassium Channel Blockers/pharmacology , Protein Subunits/biosynthesis , Rats , Rats, Sprague-Dawley
9.
J Med Chem ; 63(1): 163-185, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31815462

ABSTRACT

Kv7 K+ channels represent attractive pharmacological targets for the treatment of different neurological disorders, including epilepsy. In this paper, 42 conformationally restricted analogues of the prototypical Kv7 activator retigabine have been synthesized and tested by electrophysiological patch-clamp experiments as Kv7 agonists. When compared to retigabine (0.93 ± 0.43 µM), the EC50s for Kv7.2 current enhancements by compound 23a (0.08 ± 0.04 µM) were lower, whereas no change in potency was observed for 24a (0.63 ± 0.07 µM). In addition, compared to retigabine, 23a and 24a showed also higher potency in activating heteromeric Kv7.2/Kv7.3 and homomeric Kv7.4 channels. Molecular modeling studies provided new insights into the chemical features required for optimal interaction at the binding site. Stability studies evidenced improved chemical stability of 23a and 24a in comparison with retigabine. Overall, the present results highlight that the N5-alkylamidoindole moiety provides a suitable pharmacophoric scaffold for the design of chemically stable, highly potent and selective Kv7 agonists.


Subject(s)
Indoles/pharmacology , KCNQ2 Potassium Channel/agonists , KCNQ3 Potassium Channel/agonists , Animals , CHO Cells , Carbamates/chemistry , Cricetulus , Indoles/chemical synthesis , Indoles/metabolism , KCNQ2 Potassium Channel/genetics , KCNQ2 Potassium Channel/metabolism , KCNQ3 Potassium Channel/metabolism , Microsomes, Liver/metabolism , Models, Molecular , Molecular Conformation , Mutation , Phenylenediamines/chemistry , Protein Binding , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Xenopus laevis
10.
Eur J Pharmacol ; 858: 172474, 2019 Sep 05.
Article in English | MEDLINE | ID: mdl-31238068

ABSTRACT

The Kv7 family of voltage-dependent non-inactivating potassium channels is composed of five members, of which four are expressed in the CNS. Kv7.2, 7.3 and 7.5 are responsible for the M-current, which plays a critical role in the regulation of neuronal excitability. Stimulation of M1 muscarinic acetylcholine receptor, M1 receptor, increases neuronal excitability by suppressing the M-current generated by the Kv7 channel family. The M-current modulation via M1 receptor is well-described in in vitro assays using cell lines and in native rodent tissue. However, this mechanism was not yet reported in human induced pluripotent stem cells (hiPSC) derived neurons. In the present study, we investigated the effects of both agonists and antagonists of Kv7.2/7.3 channel and M1 receptor in hiPSC derived neurons and in primary rat cortical neuronal cells. The role of M1 receptors in the modulation of neuronal excitability could be demonstrated in both rat primary and hiPSC neurons. The M1 receptors agonist, xanomeline, increased neuronal excitability in both rat cortical and the hiPSC neuronal cells. Furthermore, M1 receptor agonist-induced neuronal excitability in vitro was reduced by an agonist of Kv7.2/7.3 in both neuronal cells. These results show that hiPSC derived neurons recreate the modulation of the M-current by the muscarinic receptor in hiPSC neurons similarly to rat native neurons. Thus, hiPSC neurons could be a useful human-based cell assay for characterization of drugs that affect neuronal excitability and/or induce seizure activity by modulation of M1 receptors or inhibition of Kv7 channels.


Subject(s)
Electrophysiological Phenomena , Induced Pluripotent Stem Cells/cytology , KCNQ2 Potassium Channel/metabolism , KCNQ3 Potassium Channel/metabolism , Neurons/cytology , Receptor, Muscarinic M1/metabolism , Animals , Electrophysiological Phenomena/drug effects , Gene Expression Regulation/drug effects , Humans , KCNQ2 Potassium Channel/agonists , KCNQ2 Potassium Channel/antagonists & inhibitors , KCNQ2 Potassium Channel/genetics , KCNQ3 Potassium Channel/agonists , KCNQ3 Potassium Channel/antagonists & inhibitors , KCNQ3 Potassium Channel/genetics , Muscarinic Antagonists/pharmacology , Neurons/drug effects , Neurons/metabolism , Potassium Channel Blockers/pharmacology , Rats , Receptor, Muscarinic M1/agonists , Receptor, Muscarinic M1/antagonists & inhibitors
11.
FASEB J ; 33(8): 9154-9166, 2019 08.
Article in English | MEDLINE | ID: mdl-31063701

ABSTRACT

Voltage-gated Kv7/KCNQ/M potassium channels play an essential role in the control of membrane potential and neuronal excitability. Activation of the neuronal Kv7/KCNQ/M-current represents an attractive therapeutic strategy for treatment of hyperexcitability-related neuropsychiatric disorders such as epilepsy, pain, and depression, which is an unmet medical need. In this study, we synthesized and characterized a novel compound, N-(4-(2-bromo-6,7-dihydrothieno[3,2-c]pyridin-5(4H)-yl)-2,6-dimethylphenyl)-3,3-dimethylbutanamide (SCR2682) 2,6-dimethyl-4-(piperidin-yl) phenyl)-amide derivative, that exhibits selective and potent activation of neuronal Kv7/KCNQ/M-channels. Whole-cell patch-clamp recordings of human embryonic kidney 293 cells expressing Kv7.2/Kv7.3 channels show that SCR2682 selectively activates the channel current in a dose-dependent manner with an EC50 of 9.8 ± 0.4 nM, which is ∼100-fold more potent than a U.S. Food and Drug Administration-approved antiepileptic drug (retigabine) for treatment of partial epilepsy. SCR2682 shifts voltage-dependent activation of the Kv7.2/7.3 current toward more negative membrane potential, to about -37 mV (V1/2). SCR2682 also activates the native M-current in rat hippocampal or cortical neurons, causing marked hyperpolarization and potent inhibition of neuronal firings. Mechanistically, mutating the tryptophan residue 236 located at the fifth transmembrane segment of Kv7.2 abolishes the chemical activation of the channel by SCR2682. Furthermore, intraperitoneal or intragastric administration of SCR2682 results in a dose-dependent inhibition of seizures by maximal electroshock. Taken together, our findings demonstrate that a novel small molecule, SCR2682, selectively and potently activates neuronal Kv7 channels and reverses epileptic seizures in rodents. Thus, SCR2682 may warrant further evaluation for clinical development of antiepileptic therapy.-Zhang, F., Liu, Y., Tang, F., Liang, B., Chen, H., Zhang, H., Wang, K. Electrophysiological and pharmacological characterization of a novel and potent neuronal Kv7 channel opener SCR2682 for antiepilepsy.


Subject(s)
Anticonvulsants/pharmacology , KCNQ2 Potassium Channel/agonists , KCNQ3 Potassium Channel/agonists , Membrane Transport Modulators/pharmacology , Pyridines/pharmacology , Amino Acid Substitution , Animals , Anticonvulsants/chemical synthesis , Anticonvulsants/chemistry , Cells, Cultured , ERG1 Potassium Channel/antagonists & inhibitors , Epilepsy/drug therapy , HEK293 Cells , Humans , KCNQ Potassium Channels/agonists , KCNQ2 Potassium Channel/genetics , KCNQ2 Potassium Channel/metabolism , Male , Membrane Potentials/drug effects , Membrane Transport Modulators/chemical synthesis , Membrane Transport Modulators/chemistry , Mice , Mutagenesis, Site-Directed , Neurons/drug effects , Neurons/metabolism , Patch-Clamp Techniques , Pyridines/chemical synthesis , Pyridines/chemistry , Rats , Seizures/drug therapy
12.
Acta Physiol (Oxf) ; 218(1): 28-37, 2016 09.
Article in English | MEDLINE | ID: mdl-26914447

ABSTRACT

AIM: Polyunsaturated fatty acids have been reported to reduce neuronal excitability, in part by promoting inactivation of voltage-gated sodium and calcium channels. Effects on neuronal potassium channels are less explored and experimental data ambiguous. The aim of this study was to investigate anti-excitable effects of polyunsaturated fatty acids on the neuronal M-channel, important for setting the resting membrane potential in hippocampal and dorsal root ganglion neurones. METHODS: Effects of fatty acids and fatty acid analogues on mouse dorsal root ganglion neurones and on the human KV 7.2/3 channel expressed in Xenopus laevis oocytes were studied using electrophysiology. RESULTS: Extracellular application of physiologically relevant concentrations of the polyunsaturated fatty acid docosahexaenoic acid hyperpolarized the resting membrane potential (-2.4 mV by 30 µm) and increased the threshold current to evoke action potentials in dorsal root ganglion neurones. The polyunsaturated fatty acids docosahexaenoic acid, α-linolenic acid and eicosapentaenoic acid facilitated opening of the human M-channel, comprised of the heteromeric human KV 7.2/3 channel expressed in Xenopus oocytes, by shifting the conductance-vs.-voltage curve towards more negative voltages (by -7.4 to -11.3 mV by 70 µm). Uncharged docosahexaenoic acid methyl ester and monounsaturated oleic acid did not facilitate opening of the human KV 7.2/3 channel. CONCLUSIONS: These findings suggest that circulating polyunsaturated fatty acids, with a minimum requirement of multiple double bonds and a charged carboxyl group, dampen excitability by opening neuronal M-channels. Collectively, our data bring light to the molecular targets of polyunsaturated fatty acids and thus a possible mechanism by which polyunsaturated fatty acids reduce neuronal excitability.


Subject(s)
Fatty Acids, Unsaturated/pharmacology , KCNQ2 Potassium Channel/agonists , KCNQ3 Potassium Channel/agonists , Animals , Fatty Acids, Omega-3/pharmacology , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Hippocampus/drug effects , Humans , Ion Channel Gating/drug effects , Membrane Potentials/drug effects , Mice, Inbred C57BL , Neurons/drug effects , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Xenopus laevis
13.
J Gen Physiol ; 147(3): 229-41, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26880756

ABSTRACT

The anticonvulsant Retigabine is a KV7 channel agonist used to treat hyperexcitability disorders in humans. Retigabine shifts the voltage dependence for activation of the heteromeric KV7.2/KV7.3 channel to more negative potentials, thus facilitating activation. Although the molecular mechanism underlying Retigabine's action remains unknown, previous studies have identified the pore region of KV7 channels as the drug's target. This suggested that the Retigabine-induced shift in voltage dependence likely derives from the stabilization of the pore domain in an open (conducting) conformation. Testing this idea, we show that the heteromeric KV7.2/KV7.3 channel has at least two open states, which we named O1 and O2, with O2 being more stable. The O1 state was reached after short membrane depolarizations, whereas O2 was reached after prolonged depolarization or during steady state at the typical neuronal resting potentials. We also found that activation and deactivation seem to follow distinct pathways, suggesting that the KV7.2/KV7.3 channel activity displays hysteresis. As for the action of Retigabine, we discovered that this agonist discriminates between open states, preferentially acting on the O2 state and further stabilizing it. Based on these findings, we proposed a novel mechanism for the therapeutic effect of Retigabine whereby this drug reduces excitability by enhancing the resting potential open state stability of KV7.2/KV7.3 channels. To address this hypothesis, we used a model for action potential (AP) in Xenopus laevis oocytes and found that the resting membrane potential became more negative as a function of Retigabine concentration, whereas the threshold potential for AP firing remained unaltered.


Subject(s)
Anticonvulsants/pharmacology , Carbamates/pharmacology , KCNQ2 Potassium Channel/agonists , KCNQ3 Potassium Channel/agonists , Membrane Potentials , Phenylenediamines/pharmacology , Animals , Humans , Ion Channel Gating , KCNQ2 Potassium Channel/chemistry , KCNQ2 Potassium Channel/metabolism , KCNQ3 Potassium Channel/chemistry , KCNQ3 Potassium Channel/metabolism , Protein Domains , Protein Multimerization , Xenopus
14.
Eur J Pharmacol ; 764: 633-642, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26134502

ABSTRACT

M/Kv7 K(+) channels, Ca(2+)-activated Cl(-) channels (CaCCs) and voltage gated Na(+) channels expressed in dorsal root ganglia (DRG) play an important role in nociception. Tannic acid has been proposed to be involved in multiple beneficial health effects; tannic acid has also been described to be analgesic. However the underlying mechanism is unknown. In this study, we investigated the effects of tannic acid on M/Kv7 K(+), Na(+) currents and CaCCs, and the effects on bradykinin-induced nociceptive behavior. A perforated patch technique was used. The bradykinin-induced rat pain model was used to assess the analgesic effect of tannic acid. We demonstrated that tannic acid enhanced M/Kv7 K(+) currents but inhibited bradykinin-induced activation of CaCC/TMEM16A currents in rat small DRG neurons. Tannic acid potentiated Kv7.2/7.3 and Kv7.2 currents expressed in HEK293B cells, with an EC50 of 7.38 and 5.40 µM, respectively. Tannic acid inhibited TTX-sensitive and TTX-insensitive currents of small DRG neurons with IC50 of 5.25 and 8.43 µM, respectively. Tannic acid also potently suppressed the excitability of small DRG neurons. Furthermore, tannic acid greatly reduced bradykinin-induced pain behavior of rats. This study thus demonstrates that tannic acid is an activator of M/Kv7 K(+) and an inhibitor of voltage-gated Na(+) channels and CaCC/TMEM16A, which may underlie its inhibitory effects on excitability of DRG neurons and its analgesic effect. Tannic acid could be a useful agent in treatment of inflammatory pain conditions such as osteoarthritis, rheumatic arthritis and burn pain.


Subject(s)
Analgesics/pharmacology , Chloride Channels/antagonists & inhibitors , Ganglia, Spinal/drug effects , KCNQ Potassium Channels/agonists , Nociception/drug effects , Nociceptive Pain/drug therapy , Sensory Receptor Cells/drug effects , Tannins/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channels/drug effects , Animals , Anoctamin-1 , Behavior, Animal/drug effects , Bradykinin , Chloride Channels/genetics , Chloride Channels/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Ganglia, Spinal/metabolism , Ganglia, Spinal/physiopathology , HEK293 Cells , Humans , KCNQ Potassium Channels/genetics , KCNQ Potassium Channels/metabolism , KCNQ2 Potassium Channel/agonists , KCNQ2 Potassium Channel/metabolism , KCNQ3 Potassium Channel/agonists , KCNQ3 Potassium Channel/metabolism , Membrane Potentials , Nociceptive Pain/chemically induced , Nociceptive Pain/metabolism , Nociceptive Pain/physiopathology , Nociceptive Pain/psychology , Rats, Sprague-Dawley , Sensory Receptor Cells/metabolism , Transfection , Voltage-Gated Sodium Channels/metabolism
15.
J Neurosci ; 35(23): 8829-42, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-26063916

ABSTRACT

Voltage-gated Kv7 (KCNQ) channels are voltage-dependent potassium channels that are activated at resting membrane potentials and therefore provide a powerful brake on neuronal excitability. Genetic or experience-dependent reduction of KCNQ2/3 channel activity is linked with disorders that are characterized by neuronal hyperexcitability, such as epilepsy and tinnitus. Retigabine, a small molecule that activates KCNQ2-5 channels by shifting their voltage-dependent opening to more negative voltages, is an US Food and Drug Administration (FDA) approved anti-epileptic drug. However, recently identified side effects have limited its clinical use. As a result, the development of improved KCNQ2/3 channel activators is crucial for the treatment of hyperexcitability-related disorders. By incorporating a fluorine substituent in the 3-position of the tri-aminophenyl ring of retigabine, we synthesized a small-molecule activator (SF0034) with novel properties. Heterologous expression of KCNQ2/3 channels in HEK293T cells showed that SF0034 was five times more potent than retigabine at shifting the voltage dependence of KCNQ2/3 channels to more negative voltages. Moreover, unlike retigabine, SF0034 did not shift the voltage dependence of either KCNQ4 or KCNQ5 homomeric channels. Conditional deletion of Kcnq2 from cerebral cortical pyramidal neurons showed that SF0034 requires the expression of KCNQ2/3 channels for reducing the excitability of CA1 hippocampal neurons. Behavioral studies demonstrated that SF0034 was a more potent and less toxic anticonvulsant than retigabine in rodents. Furthermore, SF0034 prevented the development of tinnitus in mice. We propose that SF0034 provides, not only a powerful tool for investigating ion channel properties, but, most importantly, it provides a clinical candidate for treating epilepsy and preventing tinnitus.


Subject(s)
Anticonvulsants/therapeutic use , Carbamates/therapeutic use , Epilepsy/drug therapy , KCNQ2 Potassium Channel/agonists , KCNQ2 Potassium Channel/metabolism , Phenylenediamines/therapeutic use , Tinnitus/prevention & control , Animals , Animals, Newborn , Anticonvulsants/chemistry , Carbamates/chemistry , Disease Models, Animal , Epilepsy/etiology , Epilepsy/genetics , Evoked Potentials, Auditory, Brain Stem/genetics , Female , HEK293 Cells , Homeodomain Proteins/genetics , Humans , In Vitro Techniques , KCNQ Potassium Channels/genetics , KCNQ2 Potassium Channel/genetics , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred ICR , Mice, Knockout , Mutation/genetics , Phenylenediamines/chemistry , Rats , Rats, Sprague-Dawley , Tinnitus/etiology , Transcription Factors/genetics
16.
Bioorg Med Chem Lett ; 25(21): 4941-4944, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-25987375

ABSTRACT

Facilitating activation, or delaying inactivation, of the native Kv7 channel reduces neuronal excitability, which may be beneficial in controlling spontaneous electrical activity during epileptic seizures. In an effort to identify a compound with such properties, the structure-activity relationship (SAR) and in vitro ADME for a series of heterocyclic Kv7.2-7.5 channel openers was explored. PF-05020182 (2) demonstrated suitable properties for further testing in vivo where it dose-dependently decreased the number of animals exhibiting full tonic extension convulsions in response to corneal stimulation in the maximal electroshock (MES) assay. In addition, PF-05020182 (2) significantly inhibited convulsions in the MES assay at doses tested, consistent with in vitro activity measure. The physiochemical properties, in vitro and in vivo activities of PF-05020182 (2) support further development as an adjunctive treatment of refractory epilepsy.


Subject(s)
Drug Discovery , Epilepsy/drug therapy , Ion Channel Gating/drug effects , KCNQ2 Potassium Channel/metabolism , Piperidines/pharmacology , Pyrimidines/pharmacology , Animals , Cell Line , Dose-Response Relationship, Drug , Electroshock , Humans , KCNQ2 Potassium Channel/agonists , Microsomes/drug effects , Molecular Structure , Piperidines/administration & dosage , Piperidines/chemistry , Pyrimidines/administration & dosage , Pyrimidines/chemistry , Rats , Structure-Activity Relationship
17.
J Pharmacol Exp Ther ; 351(3): 596-604, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25232191

ABSTRACT

Human ether-a-go-go-related gene (hERG) and KCNQ channels are two classes of voltage-gated potassium channels. Specific mutations have been identified that are causal for type II long QT (LQT2) syndrome, neonatal epilepsy, and benign familial neonatal convulsions. Increasing evidence from clinical studies suggests that LQT2 and epilepsy coexist in some patients. Therefore, an integral approach to investigating and treating the two diseases is likely more effective. In the current study, we found that NS1643 [1,3-bis-(2-hydroxy-5-trifluoromethyl-phenyl)-urea], a previously reported hERG activator, is also an activator of KCNQ channels. It potentiates the neuronal KCNQ2, KCNQ4, and KCNQ2/Q3 channels, but not the cardiac KCNQ1. The effects of NS1643 on the KCNQ2 channel include left shifting of voltage for reaching 50% of the maximum conductance and slowing of deactivation. Analysis of the dose-response curve of NS1643 revealed an EC50 value of 2.44 ± 0.25 µM. A hydrophobic phenylalanine (F137) located at the middle region of the voltage-sensing domain was identified as critical for NS1643 activity on KCNQ2. When testing NS1643 effects in rescuing LQT2 hERG mutants and the KCNQ2 BFNC mutants, we found it is particularly efficacious in some cases. Considering the substantial relationship between LQT2 and epilepsy, these findings reveal that NS1643 is a useful compound to elucidate the causal connection of LQT2 and epilepsy. More generally, this may provide a strategy in the development of therapeutics for LQT2 and epilepsy.


Subject(s)
Cresols/metabolism , Cresols/pharmacology , Epilepsy/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , KCNQ2 Potassium Channel/metabolism , Phenylurea Compounds/metabolism , Phenylurea Compounds/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Ether-A-Go-Go Potassium Channels/agonists , Humans , KCNQ2 Potassium Channel/agonists , Protein Structure, Secondary
18.
PLoS One ; 9(6): e100209, 2014.
Article in English | MEDLINE | ID: mdl-24956197

ABSTRACT

The voltage-gated potassium channels of the KV7 family (KV7.1-5) play important roles in controlling neuronal excitability and are therefore attractive targets for treatment of CNS disorders linked to hyperexcitability. One of the main challenges in developing KV7 channel active drugs has been to identify compounds capable of discriminating between the neuronally expressed subtypes (KV7.2-5), aiding the identification of the subunit composition of KV7 currents in various tissues, and possessing better therapeutic potential for particular indications. By taking advantage of the structure-activity relationship of acrylamide KV7 channel openers and the effects of these compounds on mutant KV7 channels, we have designed and synthesized a novel KV7 channel modulator with a unique profile. The compound, named SMB-1, is an inhibitor of KV7.2 and an activator of KV7.4. SMB-1 inhibits KV7.2 by reducing the current amplitude and increasing the time constant for the slow component of the activation kinetics. The activation of KV7.4 is seen as an increase in the current amplitude and a slowing of the deactivation kinetics. Experiments studying mutant channels with a compromised binding site for the KV7.2-5 opener retigabine indicate that SMB-1 binds within the same pocket as retigabine for both inhibition of KV7.2 and activation of KV7.4. SMB-1 may serve as a valuable tool for KV7 channel research and may be used as a template for further design of better subtype selective KV7 channel modulators. A compound with this profile could hold novel therapeutic potential such as the treatment of both positive and cognitive symptoms in schizophrenia.


Subject(s)
KCNQ Potassium Channels , KCNQ2 Potassium Channel , Membrane Transport Modulators/chemistry , Membrane Transport Modulators/pharmacology , Mutation, Missense , Amino Acid Substitution , Animals , Humans , KCNQ Potassium Channels/agonists , KCNQ Potassium Channels/antagonists & inhibitors , KCNQ Potassium Channels/genetics , KCNQ Potassium Channels/metabolism , KCNQ2 Potassium Channel/agonists , KCNQ2 Potassium Channel/antagonists & inhibitors , KCNQ2 Potassium Channel/genetics , KCNQ2 Potassium Channel/metabolism , Xenopus laevis
19.
Cell Res ; 23(9): 1106-18, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23797855

ABSTRACT

Voltage-gated potassium (Kv) channels derive their voltage sensitivity from movement of gating charges in voltage-sensor domains (VSDs). The gating charges translocate through a physical pathway in the VSD to open or close the channel. Previous studies showed that the gating charge pathways of Shaker and Kv1.2-2.1 chimeric channels are occluded, forming the structural basis for the focused electric field and gating charge transfer center. Here, we show that the gating charge pathway of the voltage-gated KCNQ2 potassium channel, activity reduction of which causes epilepsy, can accommodate various small molecule ligands. Combining mutagenesis, molecular simulation and electrophysiological recording, a binding model for the probe activator, ztz240, in the gating charge pathway was defined. This information was used to establish a docking-based virtual screening assay targeting the defined ligand-binding pocket. Nine activators with five new chemotypes were identified, and in vivo experiments showed that three ligands binding to the gating charge pathway exhibit significant anti-epilepsy activity. Identification of various novel activators by virtual screening targeting the pocket supports the presence of a ligand-binding site in the gating charge pathway. The capability of the gating charge pathway to accommodate small molecule ligands offers new insights into the gating charge pathway of the therapeutically relevant KCNQ2 channel.


Subject(s)
Anticonvulsants/metabolism , Benzamides/pharmacology , Epilepsy/metabolism , KCNQ2 Potassium Channel/metabolism , Pyridines/pharmacology , Shaker Superfamily of Potassium Channels/metabolism , Animals , Benzamides/metabolism , Binding Sites , CHO Cells , Cell Line , Cricetulus , Ion Channel Gating/physiology , KCNQ2 Potassium Channel/agonists , Male , Membrane Potentials/physiology , Mice , Patch-Clamp Techniques , Protein Binding , Pyridines/metabolism , Static Electricity
20.
Br J Pharmacol ; 169(6): 1279-89, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23581476

ABSTRACT

BACKGROUND AND PURPOSE: Retigabine is a recently approved antiepileptic agent which activates Kv7.2-7.5 potassium channels. It is emerging that these channels have an important role in vascular regulation, but the vascular effects of retigabine in the conscious state are unknown. Hence, in the present study we assessed the regional haemodynamic responses to retigabine in conscious rats. EXPERIMENTAL APPROACH: Male Sprague Dawley rats were chronically instrumented with pulsed Doppler flow probes to measure regional haemodynamic responses to retigabine under control conditions and during acute hypertension induced by infusion of angiotensin II and arginine vasopressin. Further experiments were performed, using the ß-adrenoceptor antagonists CGP 20712A, ICI 118551 and propranolol, to elucidate the roles of ß-adrenoceptors in the responses to retigabine in vivo and in vitro. KEY RESULTS: Under normotensive conditions, retigabine induced dose-dependent hypotension and hindquarters vasodilatation, with small, transient renal and mesenteric vasodilatations. In the acutely hypertensive state, the renal and mesenteric, but not hindquarters, vasodilatations were enhanced. The response of the hindquarters vascular bed to retigabine was mediated, in part, by ß2-adrenoceptors. However, in vitro experiments confirmed that retigabine did not act as a ß-adrenoceptor agonist. CONCLUSIONS AND IMPLICATIONS: We demonstrated that retigabine causes regionally specific vasodilatations, which are different under normotensive and hypertensive conditions, and are, in part, mediated by ß2-adrenoceptors in some vascular beds but not in others. These results broadly support previous findings and further indicate that Kv7 channels are a potential therapeutic target for the treatment of vascular diseases associated with inappropriate vasoconstriction.


Subject(s)
Antihypertensive Agents/therapeutic use , Carbamates/therapeutic use , Coronary Circulation/drug effects , Hypertension/drug therapy , KCNQ Potassium Channels/agonists , KCNQ2 Potassium Channel/agonists , Membrane Transport Modulators/therapeutic use , Phenylenediamines/therapeutic use , Adrenergic beta-2 Receptor Agonists/administration & dosage , Adrenergic beta-2 Receptor Agonists/adverse effects , Adrenergic beta-2 Receptor Agonists/pharmacology , Adrenergic beta-2 Receptor Agonists/therapeutic use , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Agonists/therapeutic use , Animals , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/adverse effects , Antihypertensive Agents/pharmacology , Arrhythmias, Cardiac/chemically induced , CHO Cells , Carbamates/administration & dosage , Carbamates/adverse effects , Carbamates/pharmacology , Cricetulus , Dose-Response Relationship, Drug , Humans , KCNQ Potassium Channels/metabolism , KCNQ2 Potassium Channel/metabolism , Male , Membrane Transport Modulators/administration & dosage , Membrane Transport Modulators/adverse effects , Membrane Transport Modulators/pharmacology , Phenylenediamines/administration & dosage , Phenylenediamines/adverse effects , Phenylenediamines/pharmacology , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Vasodilation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...