Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Neuropharmacology ; 254: 109994, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38750803

ABSTRACT

Neuronal voltage-gated KCNQ (Kv7) channels, expressed centrally and peripherally, mediate low-threshold and non-inactivating M-currents responsible for the control of tonic excitability of mammalian neurons. Pharmacological opening of KCNQ channels has been reported to generate analgesic effects in animal models of neuropathic pain. Here, we examined the possible involvement of central KCNQ channels in the analgesic effects of retigabine, a KCNQ channel opener. Behaviorally, intraperitoneally applied retigabine exerted analgesic effects on thermal and mechanical hypersensitivity in male mice developing neuropathic pain after partial sciatic nerve ligation, which was antagonized by the KCNQ channel blocker XE991 preadministered intraperitoneally and intrathecally. Intrathecally applied retigabine also exerted analgesic effects that were inhibited by intrathecally injected XE991. We then explored the synaptic mechanisms underlying the analgesic effects of retigabine in the spinal dorsal horn. Whole-cell recordings were made from dorsal horn neurons in spinal slices with attached dorsal roots from adult male mice developing neuropathic pain, and the effects of retigabine on miniature and afferent-evoked postsynaptic currents were examined. Retigabine reduced the amplitude of A-fiber-mediated EPSCs without affecting C-fiber-mediated excitatory synaptic transmission. A-fiber-mediated EPSCs remained unaltered by retigabine in the presence of XE991, consistently with the behavioral findings. The frequency and amplitude of mEPSCs were not affected by retigabine. Thus, opening of KCNQ channels in the central terminals of primary afferent A-fibers inhibits excitatory synaptic transmission in the spinal dorsal horn, most likely contributing to the analgesic effect of retigabine.


Subject(s)
Analgesics , Anthracenes , Carbamates , KCNQ Potassium Channels , Phenylenediamines , Animals , Male , Carbamates/pharmacology , Phenylenediamines/pharmacology , KCNQ Potassium Channels/antagonists & inhibitors , KCNQ Potassium Channels/drug effects , Anthracenes/pharmacology , Mice , Analgesics/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Neuralgia/drug therapy , Posterior Horn Cells/drug effects , Nerve Fibers, Myelinated/drug effects , Nerve Fibers, Myelinated/physiology , Spinal Cord Dorsal Horn/drug effects
2.
J Biol Chem ; 299(2): 102819, 2023 02.
Article in English | MEDLINE | ID: mdl-36549648

ABSTRACT

Zinc (Zn) is an essential trace element; it serves as a cofactor for a great number of enzymes, transcription factors, receptors, and other proteins. Zinc is also an important signaling molecule, which can be released from intracellular stores into the cytosol or extracellular space, for example, during synaptic transmission. Amongst cellular effects of zinc is activation of Kv7 (KCNQ, M-type) voltage-gated potassium channels. Here, we investigated relationships between Kv7 channel inhibition by Ca2+/calmodulin (CaM) and zinc-mediated potentiation. We show that Zn2+ ionophore, zinc pyrithione (ZnPy), can prevent or reverse Ca2+/CaM-mediated inhibition of Kv7.2. In the presence of both Ca2+ and Zn2+, the Kv7.2 channels lose most of their voltage dependence and lock in an open state. In addition, we demonstrate that mutations that interfere with CaM binding to Kv7.2 and Kv7.3 reduced channel membrane abundance and activity, but these mutants retained zinc sensitivity. Moreover, the relative efficacy of ZnPy to activate these mutants was generally greater, compared with the WT channels. Finally, we show that zinc sensitivity was retained in Kv7.2 channels assembled with mutant CaM with all four EF hands disabled, suggesting that it is unlikely to be mediated by CaM. Taken together, our findings indicate that zinc is a potent Kv7 stabilizer, which may protect these channels from physiological inhibitory effects of neurotransmitters and neuromodulators, protecting neurons from overactivity.


Subject(s)
Calcium , Calmodulin , Intracellular Space , KCNQ Potassium Channels , Zinc , Calcium Signaling , Calmodulin/metabolism , KCNQ Potassium Channels/antagonists & inhibitors , KCNQ Potassium Channels/chemistry , KCNQ Potassium Channels/genetics , KCNQ Potassium Channels/metabolism , Mutation , Protein Binding/genetics , Zinc/pharmacology , Zinc/metabolism , Intracellular Space/metabolism , Calcium/metabolism , KCNQ2 Potassium Channel/antagonists & inhibitors , KCNQ2 Potassium Channel/chemistry , KCNQ2 Potassium Channel/genetics , KCNQ2 Potassium Channel/metabolism , KCNQ3 Potassium Channel/antagonists & inhibitors , KCNQ3 Potassium Channel/chemistry , KCNQ3 Potassium Channel/genetics , KCNQ3 Potassium Channel/metabolism
3.
Eur J Pharmacol ; 906: 174278, 2021 Sep 05.
Article in English | MEDLINE | ID: mdl-34174270

ABSTRACT

KCNQ-encoded (KV7) potassium channels are diversely distributed in the human tissues, associated with many physiological processes and pathophysiological conditions. These channels are increasingly used as drug targets for treating diseases. More selective and potent molecules on various types of the KV7 channels are desirable for appropriate therapies. The recent knowledge of the structure and function of human KCNQ-encoded channels makes it more feasible to achieve these goals. This review discusses the role and mechanism of action of many molecules in modulating the function of the KCNQ-encoded potassium channels in the heart and nervous system. The effects of these compounds on KV7 channels help to understand their involvement in many diseases, and to search for more selective and potent ligands to be used in the treatment of many disorders such as various types of cardiac arrhythmias, epilepsy, and pain.


Subject(s)
Analgesics/pharmacology , Anti-Arrhythmia Agents/pharmacology , Anticonvulsants/pharmacology , KCNQ Potassium Channels/antagonists & inhibitors , Analgesics/therapeutic use , Animals , Anti-Arrhythmia Agents/therapeutic use , Anticonvulsants/therapeutic use , Arrhythmias, Cardiac/drug therapy , Epilepsy/drug therapy , Heart/drug effects , Humans , KCNQ Potassium Channels/metabolism , Ligands , Myocardium/metabolism , Neurons/drug effects , Neurons/metabolism , Pain/drug therapy
4.
J Neurophysiol ; 126(1): 1-10, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34038189

ABSTRACT

The exercise pressor reflex (EPR) originates in skeletal muscle and is activated by exercise-induced signals to increase arterial blood pressure and cardiac output. Muscle ischemia can elicit the EPR, which can be inappropriately activated in patients with peripheral vascular disease or heart failure to increase the incidence of myocardial infarction. We seek to better understand the receptor/channels that control excitability of group III and group IV muscle afferent fibers that give rise to the EPR. Bradykinin (BK) is released within contracting muscle and can evoke the EPR. However, the mechanism is incompletely understood. KV7 channels strongly regulate neuronal excitability and are inhibited by BK. We have identified KV7 currents in muscle afferent neurons by their characteristic activation/deactivation kinetics, enhancement by the KV7 activator retigabine, and block by KV7 specific inhibitor XE991. The blocking of KV7 current by different XE991 concentrations suggests that the KV7 current is generated by both KV7.2/7.3 (high affinity) and KV7.5 (low affinity) channels. The KV7 current was inhibited by 300 nM BK in neurons with diameters consistent with both group III and group IV afferents. The inhibition of KV7 by BK could be a mechanism by which this metabolic mediator generates the EPR. Furthermore, our results suggest that KV7 channel activators such as retigabine, could be used to reduce cardiac stress resulting from the exacerbated EPR in patients with cardiovascular disease.NEW & NOTEWORTHY KV7 channels control neuronal excitability. We show that these channels are expressed in muscle afferents and generate currents that are blocked by XE991 and bradykinin (BK). The XE991 block suggests that KV7 current is generated by KV7.2/3 and KV7.5 channels. The BK inhibition of KV7 channels may explain how BK activates the exercise pressor reflex (EPR). Retigabine can enhance KV7 current, which could help control the inappropriately activated EPR in patients with cardiovascular disease.


Subject(s)
KCNQ Potassium Channels/physiology , Muscle Contraction/physiology , Muscle, Skeletal/physiology , Physical Conditioning, Animal/physiology , Reflex/physiology , Animals , Anthracenes/pharmacology , Anticonvulsants/pharmacology , Carbamates/pharmacology , Dose-Response Relationship, Drug , KCNQ Potassium Channels/antagonists & inhibitors , Male , Muscle Contraction/drug effects , Muscle, Skeletal/drug effects , Phenylenediamines/pharmacology , Rats , Rats, Sprague-Dawley , Reflex/drug effects
5.
Mol Cell ; 81(1): 25-37.e4, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33238160

ABSTRACT

Among the five KCNQ channels, also known as the Kv7 voltage-gated potassium (Kv) channels, KCNQ2-KCNQ5 control neuronal excitability. Dysfunctions of KCNQ2-KCNQ5 are associated with neurological disorders such as epilepsy, deafness, and neuropathic pain. Here, we report the cryoelectron microscopy (cryo-EM) structures of human KCNQ4 and its complexes with the opener retigabine or the blocker linopirdine at overall resolutions of 2.5, 3.1, and 3.3 Å, respectively. In all structures, a phosphatidylinositol 4,5-bisphosphate (PIP2) molecule inserts its head group into a cavity within each voltage-sensing domain (VSD), revealing an unobserved binding mode for PIP2. Retigabine nestles in each fenestration, inducing local shifts. Instead of staying within the central pore, linopirdine resides in a cytosolic cavity underneath the inner gate. Electrophysiological analyses of various mutants corroborated the structural observations. Our studies reveal the molecular basis for the modulatory mechanism of neuronal KCNQ channels and provide a framework for structure-facilitated drug discovery targeting these important channels.


Subject(s)
Carbamates/pharmacology , Indoles/pharmacology , KCNQ Potassium Channels , Phenylenediamines/pharmacology , Pyridines/pharmacology , Animals , Cryoelectron Microscopy , Humans , KCNQ Potassium Channels/agonists , KCNQ Potassium Channels/antagonists & inhibitors , KCNQ Potassium Channels/genetics , KCNQ Potassium Channels/metabolism , Mutation , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Domains , Sf9 Cells , Spodoptera
6.
Eur J Pharmacol ; 885: 173393, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32712094

ABSTRACT

Secretory diarrhea is one of the most common types of diarrhea with high morbidity and mortality. Previous studies showed that inhibition of cystic fibrosis transmembrane conductance regulator (CFTR) Cl- channels alleviated fluid loss in secretory diarrheas. This study aimed to identify novel CFTR inhibitors from fungal metabolites and explore its underlying mechanisms and potential utility in secretory diarrheas. Electrophysiological analyses in human intestinal epithelial (T84) cells were performed to investigate the effect and mechanism of fungal metabolites on CFTR-mediated Cl- secretion. Anti-diarrheal efficacy and the effect of compound on fluid absorption were investigated in mouse closed-loop models. We found that the screening identified arthropsolide A, a fungal metabolite from an endophytic fungus Roussoella sp. PSU-H51, as an inhibitor of CFTR-mediated Cl- secretion in T84 cells (IC50 ~0.8 µM). Arthropsolide A inhibited both CFTR and cAMP-activated basolateral K+ channels. Arthropsolide A had no effect on Na+-K+ ATPase activity. Interestingly, the inhibitory effect of arthropsolide A on CFTR was attenuated by cell depolarization and AMPK inhibition independent of multi-drug resistance protein 4, phosphodiesterases, and protein phosphatases. Importantly, arthropsolide A suppressed cholera toxin (CT)-induced Cl- secretion in T84 cells and CT-induced intestinal fluid secretion in mice by ~75% without affecting intestinal fluid absorption. Taken together, arthropsolide A represents a novel class of fungal metabolites that acts as a potent CFTR inhibitor. Further development of this class of compounds may provide a therapy for secretory diarrheas.


Subject(s)
Antidiarrheals/pharmacology , Chlorides/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Intestines/drug effects , Spiro Compounds/pharmacology , Animals , Antidiarrheals/therapeutic use , Cell Line , Cell Polarity/drug effects , Cholera Toxin/antagonists & inhibitors , Cholera Toxin/pharmacology , Drug Resistance , Fungi/metabolism , Humans , KCNQ Potassium Channels/antagonists & inhibitors , Male , Mice , Mice, Inbred ICR , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Spiro Compounds/therapeutic use
7.
Eur J Pharmacol ; 866: 172820, 2020 Jan 05.
Article in English | MEDLINE | ID: mdl-31760069

ABSTRACT

Recently, we found cardioprotective effects of ischemic preconditioning (IPC), and from a blocker of KCNQ voltage-gated K+ channels (KV7), XE991 (10,10-bis(4-pyridinylmethyl)-9(10H)-anthracenone), in isolated rat hearts. The purpose of the present study was to investigate the cardiovascular effects of IPC and XE991 and whether they are cardioprotective in intact rats. In conscious rats, we measured the effect of the KV7 channel blocker XE991 on heart rate and blood pressure by use of telemetry. In anesthetized rats, cardiac ischemia was induced by occluding the left coronary artery, and the animals received IPC (2 × 5 min of occlusion), XE991, or a combination. After a 2 h reperfusion period, the hearts were excised, and the area at risk and infarct size were determined. In both anesthetized and conscious rats, XE991 increased blood pressure, and the highest dose (7.5 mg/kg) of XE991 also increased heart rate, and 44% of conscious rats died. XE991 induced marked changes in the electrocardiogram (e.g., increased PR interval and prolonged QTC interval) without changing cardiac action potentials. The infarct size to area at risk ratio was reduced from 53 ± 2% (n = 8) in the vehicle compared to 36 ± 3% in the IPC group (P < 0.05, n = 9). XE991 (0.75 mg/kg) treatment alone or on top of IPC failed to reduce myocardial infarct size. Similar to the effect in isolated hearts, locally applied IPC was cardioprotective in intact animals exposed to ischemia-reperfusion. Systemic administration of XE991 failed to protect the heart against ischemia-reperfusion injury suggesting effects on the autonomic nervous system counteracting the cardioprotection in intact animals.


Subject(s)
Ischemic Preconditioning, Myocardial , KCNQ Potassium Channels/antagonists & inhibitors , Myocardial Reperfusion Injury/drug therapy , Potassium Channel Blockers/pharmacology , Action Potentials/drug effects , Animals , Anthracenes/pharmacology , Blood Pressure/drug effects , Electrocardiography/drug effects , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Potassium Channel Blockers/therapeutic use , Rats , Rats, Wistar
8.
Int J Mol Sci ; 20(18)2019 Sep 08.
Article in English | MEDLINE | ID: mdl-31500374

ABSTRACT

Alcohol causes diverse acute and chronic symptoms that often lead to critical health problems. Exposure to ethanol alters the activities of sympathetic neurons that control the muscles, eyes, and blood vessels in the brain. Although recent studies have revealed the cellular targets of ethanol, such as ion channels, the molecular mechanism by which alcohol modulates the excitability of sympathetic neurons has not been determined. Here, we demonstrated that ethanol increased the discharge of membrane potentials in sympathetic neurons by inhibiting the M-type or Kv7 channel consisting of the Kv7.2/7.3 subunits, which were involved in determining the membrane potential and excitability of neurons. Three types of sympathetic neurons, classified by their threshold of activation and firing patterns, displayed distinct sensitivities to ethanol, which were negatively correlated with the size of the Kv7 current that differs depending on the type of neuron. Using a heterologous expression system, we further revealed that the inhibitory effects of ethanol on Kv7.2/7.3 currents were facilitated or diminished by adjusting the amount of plasma membrane phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2). These results suggested that ethanol and PI(4,5)P2 modulated gating of the Kv7 channel in superior cervical ganglion neurons in an antagonistic manner, leading to regulation of the membrane potential and neuronal excitability, as well as the physiological functions mediated by sympathetic neurons.


Subject(s)
Action Potentials , Ethanol/metabolism , KCNQ Potassium Channels/metabolism , Neurons/physiology , Phosphatidylinositol 4,5-Diphosphate/metabolism , Superior Cervical Ganglion/cytology , Biomarkers , Cell Membrane/metabolism , Cells, Cultured , Ethanol/pharmacology , Gene Expression , KCNQ Potassium Channels/antagonists & inhibitors , KCNQ Potassium Channels/genetics
9.
Bioorg Med Chem Lett ; 29(19): 126603, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31416667

ABSTRACT

Effective treatments of neuropathic pain have been a focus of many discovery programs. KCNQ (kv7) are voltage gated potassium channel openers that have the potential for the treatment of CNS disorders including neuropathic pain. Clinical studies have suggested agents such as Retigabine to be a modulator of pain-like effects such as hyperalgesia and allodynia. In this paper, we describe the discovery and evaluation of a series of novel pyrazolopyrimidines and their affinity for potassium channels KCNQ2/3. These pyrazolopyrimidines have also shown good efficacy in the capsaicin-induced acute and secondary mechanical allodynia model and excellent pharmacokinetic properties, which may be superior to Retigabine.


Subject(s)
Drug Design , Hyperalgesia/drug therapy , Ion Channel Gating/drug effects , KCNQ Potassium Channels/antagonists & inhibitors , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Humans , Pyrazoles/chemistry , Pyrimidines/chemistry
10.
Methods Mol Biol ; 2007: 79-87, 2019.
Article in English | MEDLINE | ID: mdl-31148107

ABSTRACT

Hydrogen sulfide (H2S) evokes vascular effects through several mechanisms including in wide part the activation of some ion channels such as ATP-sensitive potassium (KATP) channels and voltage-gated Kv7 potassium channels. Electrophysiological methods are very accurate, but they require high expertise and high specialized equipment. A more manageable fluorimetric technique which allows to record the membrane potential variations by the employment of an anionic bis-oxonol dye named DiBac4(3) with the administration of different blockers of several potassium channels could be useful to discover the targets of H2S-induced vascular hyperpolarization. Coupled with this technique, a fluorimetric detection (by the use of WSP-1 dye) of H2S generation in human vascular smooth muscle cells after H2S-donor administration could confirm the ability of these molecules to evoke the hyperpolarizing effect through the H2S release.


Subject(s)
Aorta/metabolism , Fluorometry , Hydrogen Sulfide , KCNQ Potassium Channels/metabolism , Membrane Potentials/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Aorta/cytology , Barbiturates/chemistry , Cell Line , Humans , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Isoxazoles/chemistry , KCNQ Potassium Channels/antagonists & inhibitors , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Potassium Channel Blockers/pharmacology
11.
Toxins (Basel) ; 11(2)2019 02 01.
Article in English | MEDLINE | ID: mdl-30717088

ABSTRACT

It was recently discovered that Ssm Spooky Toxin (SsTx) with 53 residues serves as a key killer factor in red-headed centipede's venom arsenal, due to its potent blockage of the widely expressed KCNQ channels to simultaneously and efficiently disrupt cardiovascular, respiratory, muscular, and nervous systems, suggesting that SsTx is a basic compound for centipedes' defense and predation. Here, we show that SsTx also inhibits KV1.3 channel, which would amplify the broad-spectrum disruptive effect of blocking KV7 channels. Interestingly, residue R12 in SsTx extends into the selectivity filter to block KV7.4, however, residue K11 in SsTx replaces this ploy when toxin binds on KV1.3. Both SsTx and its mutant SsTx_R12A inhibit cytokines production in T cells without affecting the level of KV1.3 expression. The results further suggest that SsTx is a key molecule for defense and predation in the centipedes' venoms and it evolves efficient strategy to disturb multiple physiological targets.


Subject(s)
Arthropod Venoms/pharmacology , KCNQ Potassium Channels/antagonists & inhibitors , Kv1.3 Potassium Channel/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Animals , Arthropods , CHO Cells , Cricetulus , Cytokines/metabolism , HEK293 Cells , Humans , KCNQ Potassium Channels/physiology , Kv1.3 Potassium Channel/physiology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
12.
Bioorg Med Chem Lett ; 28(17): 3004-3008, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30061030

ABSTRACT

KCNQ (Kv7) has emerged as a validated target for the development of novel anti-epileptic drugs. In this paper, a series of novel N-phenylbutanamide derivatives were designed, synthesized and evaluated as KCNQ openers for the treatment of epilepsy. These compounds were evaluated for their KCNQ opening activity in vitro and in vivo. Several compounds were found to be potent KCNQ openers. Compound 1 with favorable in vitro activity was submitted to evaluation in vivo. Results showed that compound 1 owned significant anti-convulsant activity with no adverse effects. It was also found to posses favorable pharmacokinetic profiles in rat. This research may provide novel potent compounds for the discovery of KCNQ openers in treating epilepsy.


Subject(s)
Drug Design , Epilepsy/drug therapy , KCNQ Potassium Channels/antagonists & inhibitors , Phenylbutyrates/pharmacology , Potassium Channel Blockers/pharmacology , Animals , Dose-Response Relationship, Drug , Epilepsy/metabolism , Exercise Test , KCNQ Potassium Channels/metabolism , Mice , Molecular Structure , Phenylbutyrates/chemical synthesis , Phenylbutyrates/chemistry , Potassium Channel Blockers/chemical synthesis , Potassium Channel Blockers/chemistry , Rats , Stereoisomerism , Structure-Activity Relationship , Tissue Distribution
13.
Bioorg Med Chem Lett ; 28(10): 1731-1735, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29706422

ABSTRACT

Epilepsy is a kind of disease with complicated pathogenesis. KCNQ (Kv7) is a voltage dependent potassium channel that is mostly associated with epilepsy and thus becomes an important target in the treatment of epilepsy. In this paper, a series of substituted piperidine derivatives targeting KCNQ were designed and synthesized by using scaffold hopping and active substructure hybridization. Compounds were evaluated by fluorescence-based thallium influx assay, Rb+ flow assay and electrophysiological patch-clamp assay. Results showed that some compounds possessed more potent potassium channel opening activity than Retigabine. More significantly, compound 11 was found to have good pharmacokinetic profiles in vivo.


Subject(s)
Anticonvulsants/pharmacology , Drug Design , Epilepsy/drug therapy , KCNQ Potassium Channels/antagonists & inhibitors , Piperidines/pharmacology , Anticonvulsants/chemical synthesis , Anticonvulsants/chemistry , Dose-Response Relationship, Drug , Epilepsy/metabolism , Humans , KCNQ Potassium Channels/metabolism , Molecular Structure , Piperidines/chemical synthesis , Piperidines/chemistry , Structure-Activity Relationship
14.
Hear Res ; 361: 152-156, 2018 04.
Article in English | MEDLINE | ID: mdl-29459166

ABSTRACT

Specific pharmacological blockade of KCNQ (Kv7) channels with XE991 rapidly (within 20 min) and profoundly alters inner ear gravity receptor responses to head motion (Lee et al., 2017). We hypothesized that these effects were attributable to the suppression of K+ secretion following blockade of KCNQ1-KCNE1 channels in vestibular dark cells and marginal cells. To test this hypothesis, K+ secretion was independently inhibited by blocking the Na+-K+-2Cl- cotransporter (NKCC1, Slc12a2) rather than KCNQ1-KCNE1 channels. Acute blockade of NKCC1 with ethacrynic acid (40 mg/kg) eliminated auditory responses (ABRs) within approximately 70 min of injection, but had no effect on vestibular gravity receptor function (VsEPs) over a period of 2 h in the same animals. These findings show that, vestibular gravity receptors are highly resistant to acute disruption of endolymph secretion unlike the auditory system. Based on this we argue that acute suppression of K+ secretion alone does not likely account for the rapid profound effects of XE991 on gravity receptors. Instead the effects of XE991 likely require additional action at KCNQ channels located within the sensory epithelium itself.


Subject(s)
Ethacrynic Acid/pharmacology , Gravitation , Head Movements , KCNQ Potassium Channels/metabolism , Potassium/metabolism , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Solute Carrier Family 12, Member 2/drug effects , Vestibule, Labyrinth/drug effects , Animals , Anthracenes/pharmacology , Endolymph/metabolism , Evoked Potentials, Auditory, Brain Stem/drug effects , KCNQ Potassium Channels/antagonists & inhibitors , Mice, Inbred C57BL , Potassium Channel Blockers/pharmacology , Secretory Pathway , Solute Carrier Family 12, Member 2/metabolism , Time Factors , Vestibule, Labyrinth/cytology , Vestibule, Labyrinth/metabolism
15.
Proc Natl Acad Sci U S A ; 115(7): 1646-1651, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29358396

ABSTRACT

Centipedes can subdue giant prey by using venom, which is metabolically expensive to synthesize and thus used frugally through efficiently disrupting essential physiological systems. Here, we show that a centipede (Scolopendra subspinipes mutilans, ∼3 g) can subdue a mouse (∼45 g) within 30 seconds. We found that this observation is largely due to a peptide toxin in the venom, SsTx, and further established that SsTx blocks KCNQ potassium channels to exert the lethal toxicity. We also demonstrated that a KCNQ opener, retigabine, neutralizes the toxicity of a centipede's venom. The study indicates that centipedes' venom has evolved to simultaneously disrupt cardiovascular, respiratory, muscular, and nervous systems by targeting the broadly distributed KCNQ channels, thus providing a therapeutic strategy for centipede envenomation.


Subject(s)
Arthropod Venoms/toxicity , Arthropods/physiology , KCNQ Potassium Channels/antagonists & inhibitors , Nervous System Diseases/chemically induced , Predatory Behavior/drug effects , Respiratory System Abnormalities/chemically induced , Animals , Anticonvulsants/pharmacology , Carbamates/pharmacology , Mice , Nervous System Diseases/drug therapy , Nervous System Diseases/metabolism , Phenylenediamines/pharmacology , Respiratory System Abnormalities/drug therapy , Respiratory System Abnormalities/metabolism
16.
Br J Pharmacol ; 175(12): 2158-2172, 2018 06.
Article in English | MEDLINE | ID: mdl-28800673

ABSTRACT

Pathological pain is a hyperexcitability disorder. Since the excitability of a neuron is set and controlled by a complement of ion channels it expresses, in order to understand and treat pain, we need to develop a mechanistic insight into the key ion channels controlling excitability within the mammalian pain pathways and how these ion channels are regulated and modulated in various physiological and pathophysiological settings. In this review, we will discuss the emerging data on the expression in pain pathways, functional role and modulation of a family of voltage-gated K+ channels called 'M channels' (KCNQ, Kv 7). M channels are increasingly recognized as important players in controlling pain signalling, especially within the peripheral somatosensory system. We will also discuss the therapeutic potential of M channels as analgesic drug targets. LINKED ARTICLES: This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc/.


Subject(s)
KCNQ Potassium Channels/antagonists & inhibitors , KCNQ Potassium Channels/metabolism , Nociceptive Pain/drug therapy , Nociceptive Pain/metabolism , Animals , Humans
17.
Br J Pharmacol ; 175(3): 501-516, 2018 02.
Article in English | MEDLINE | ID: mdl-29156097

ABSTRACT

BACKGROUND AND PURPOSE: Kv 7.4 and Kv 7.5 channels are regulators of vascular tone. 4-Aminopyridine (4-AP) is considered a broad inhibitor of voltage-gated potassium (KV ) channels, with little inhibitory effect on Kv 7 family members at mmol concentrations. However, the effect of 4-AP on Kv 7 channels has not been systematically studied. The aim of this study was to investigate the pharmacological activity of 4-AP on Kv 7.4 and Kv 7.5 channels and characterize the effect of 4-AP on rat resistance arteries. EXPERIMENTAL APPROACH: Voltage clamp experiments were performed on Xenopus laevis oocytes injected with cRNA encoding KCNQ4 or KCNQ5, HEK cells expressing Kv 7.4 channels and on rat, freshly isolated mesenteric artery smooth muscle cells. The effect of 4-AP on tension, membrane potential, intracellular calcium and pH was assessed in rat mesenteric artery segments. KEY RESULTS: 4-AP increased the Kv 7.4-mediated current in oocytes and HEK cells but did not affect Kv 7.5 current. 4-AP also enhanced native mesenteric artery myocyte K+ current at sub-mmol concentrations. When applied to NA-preconstricted mesenteric artery segments, 4-AP hyperpolarized the membrane, decreased [Ca2+ ]i and caused concentration-dependent relaxations that were independent of 4-AP-mediated changes in intracellular pH. Application of the Kv 7 channel blocker XE991 and BKCa channel blocker iberiotoxin attenuated 4-AP-mediated relaxation. 4-AP also inhibited the NA-mediated signal transduction to elicit a relaxation. CONCLUSIONS AND IMPLICATIONS: These data show that 4-AP is able to relax NA-preconstricted rat mesenteric arteries by enhancing the activity of Kv 7.4 and BKCa channels and attenuating NA-mediated signalling.


Subject(s)
4-Aminopyridine/pharmacology , KCNQ Potassium Channels/physiology , Mesenteric Arteries/physiology , Norepinephrine/pharmacology , Potassium Channel Blockers/pharmacology , Vasoconstriction/physiology , Animals , Dose-Response Relationship, Drug , Female , HEK293 Cells , Humans , KCNQ Potassium Channels/antagonists & inhibitors , Male , Mesenteric Arteries/drug effects , Norepinephrine/antagonists & inhibitors , Organ Culture Techniques , Rats , Rats, Wistar , Vasoconstriction/drug effects , Xenopus laevis
18.
Brain Res Bull ; 137: 132-139, 2018 03.
Article in English | MEDLINE | ID: mdl-29174294

ABSTRACT

The excitability of dopaminergic neurons in the substantia nigra pars compacta (SNc) that supply the striatum with dopamine (DA) determines the function of the nigrostriatal system for motor coordination. We previously showed that 4-pyridinylmethyl-9(10H)-anthracenone (XE991), a specific blocker of Kv7/KCNQ channels, enhanced the excitability of nigral DA neurons and resulted in attenuation of haloperidol-induced catalepsy in a Parkinson's disease (PD) rat model. However, whether XE991 exhibits neuroprotective effects towards DA neuron degeneration remains unknown. The aim of this study was to investigate the effects of Kv7/KCNQ channel blocker, XE991, on 6-hydroxydopamine (6-OHDA)-induced nigral DA neuron degeneration and motor dysfunction. Using immunofluorescence staining and western blotting, we showed that intracerebroventricular administration of XE991 prevented the 6-OHDA-induced decrease in tyrosine hydroxylase (TH)-positive neurons and TH protein expression in the SNc. High-performance liquid chromatography with electrochemical detection (HPLC-ECD) also revealed that XE991 partly restored the levels of DA and its metabolites in the striatum. Moreover, XE991 decreased apomorphine (APO)-induced contralateral rotations, enhanced balance and coordination, and attenuated muscle rigidity in 6-OHDA-treated rats. Importantly, all neuroprotective effects by XE991 were abolished by co-application of Kv7/KCNQ channel opener retigabine and XE991. Thus, Kv7/KCNQ channel inhibition by XE991 can exert neuroprotective effects against 6-OHDA-induced degeneration of the nigrostriatal DA system and motor dysfunction.


Subject(s)
Anthracenes/pharmacology , Dopaminergic Neurons/drug effects , KCNQ Potassium Channels/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/drug therapy , Substantia Nigra/drug effects , Animals , Apomorphine/pharmacology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Dopamine/metabolism , Dopamine Agonists/pharmacology , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Infusions, Intraventricular , KCNQ Potassium Channels/metabolism , Male , Motor Activity/drug effects , Muscle Rigidity/drug therapy , Muscle Rigidity/physiopathology , Oxidopamine , Parkinsonian Disorders/pathology , Parkinsonian Disorders/physiopathology , Potassium Channel Blockers/pharmacology , Rats, Wistar , Substantia Nigra/metabolism , Substantia Nigra/pathology
19.
J Neurophysiol ; 118(6): 2991-3006, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28855291

ABSTRACT

The precise role and mechanisms underlying efferent modulation of peripheral vestibular afferent function are not well understood in mammals. Clarifying the details of efferent action may lead to new strategies for clinical management of debilitating disturbances in vestibular and balance function. Recent evidence in turtle indicates that efferent modulation of M-currents is likely one mechanism for modifying afferent discharge. M-currents depend in part on KCNQ potassium conductances (Kv7), which can be adjusted through efferent activation of M1, M3, and/or M5 muscarinic acetylcholine receptors (mAChRs). How KCNQ channels and altered M-currents affect vestibular afferent function in vivo is unclear, and whether such a mechanism operates in mammals is unknown. In this study we used the KCNQ antagonist XE991 and the KCNQ activator retigabine in anesthetized mice to evaluate the effects of M-current modulation on peripheral vestibular responses to transient head motion. At low doses of XE991, responses were modestly enhanced, becoming larger in amplitude and shorter in latency. Higher doses of XE991 produced transient response enhancement, followed by steady-state suppression where latencies and thresholds increased and amplitudes decreased. Retigabine produced opposite effects. Auditory function was also impacted, based on results of companion auditory brain stem response testing. We propose that closure of KCNQ channels transforms vestibular afferent behavior by suppressing responses to transient high-frequency stimuli while simultaneously enhancing responses to sustained low-frequency stimulation. Our results clearly demonstrate that KCNQ channels are critical for normal mammalian vestibular function and suggest that efferent action may utilize these mechanisms to modulate the dynamic characteristics and gain of vestibular afferent responses.NEW & NOTEWORTHY The role of calyceal KCNQ channels and associated M-current in normal mammalian vestibular function is unknown. Our results show that calyceal KCNQ channels are critical for normal vestibular function in the intact mammal. The findings provide evidence that efferent modulation of M-currents may act normally to differentially adjust the sensitivity of vestibular neurons to transient and tonic stimulation and that such mechanisms may be targeted to achieve effective clinical management of vestibular disorders.


Subject(s)
Head Movements , Motor Neurons/physiology , Vestibule, Labyrinth/physiology , Animals , Anthracenes/pharmacology , Carbamates/pharmacology , Evoked Potentials , Female , KCNQ Potassium Channels/agonists , KCNQ Potassium Channels/antagonists & inhibitors , KCNQ Potassium Channels/metabolism , Membrane Transport Modulators/pharmacology , Mice , Mice, Inbred C57BL , Motor Neurons/metabolism , Phenylenediamines/pharmacology
20.
Biophys J ; 113(3): 613-626, 2017 Aug 08.
Article in English | MEDLINE | ID: mdl-28793216

ABSTRACT

Voltage-gated potassium channels of the KCNQ (Kv7) subfamily are essential for control of cellular excitability and repolarization in a wide range of cell types. Recently, we and others found that some KCNQ channels functionally and physically interact with sodium-dependent solute transporters, including myo-inositol transporters SMIT1 and SMIT2, potentially facilitating various modes of channel-transporter signal integration. In contrast to indirect effects such as channel regulation by SMIT-transported, myo-inositol-derived phosphatidylinositol 4,5-bisphosphate (PIP2), the mechanisms and functional consequences of the physical interaction of channels with transporters have been little studied. Here, using co-immunoprecipitation with different channel domains, we found that SMIT1 binds to the KCNQ2 pore module. We next tested the effects of SMIT1 co-expression, in the absence of extracellular myo-inositol or other SMIT1 substrates, on fundamental functional attributes of KCNQ2, KCNQ2/3, KCNQ1, and KCNQ1-KCNE1 channels. Without exception, SMIT1 altered KCNQ ion selectivity, sensitivity to extracellular K+, and pharmacology, consistent with an impact on conformation of the KCNQ pore. SMIT1 also altered the gating kinetics and/or voltage dependence of KCNQ2, KCNQ2/3, and KCNQ1-KCNE1. In contrast, SMIT1 had no effect on Kv1.1 (KCNA1) gating, ion selectivity, or pharmacology. We conclude that, independent of its transport activity and indirect regulatory mechanisms involving inositol-derived increases in PIP2, SMIT1, and likely other related sodium-dependent solute transporters, regulates KCNQ channel ion selectivity, gating, and pharmacology by direct physical interaction with the pore module.


Subject(s)
Heat-Shock Proteins/metabolism , KCNQ Potassium Channels/chemistry , KCNQ Potassium Channels/metabolism , Symporters/metabolism , Extracellular Space/metabolism , Humans , Ion Channel Gating/drug effects , KCNQ Potassium Channels/antagonists & inhibitors , Models, Molecular , Porosity , Potassium/metabolism , Potassium Channels, Voltage-Gated/metabolism , Protein Binding , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...