Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
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
2.
Neurosci Lett ; 662: 29-35, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-28987817

ABSTRACT

Mental disorders such as schizophrenia are associated with impaired firing properties of fast spiking inhibitory interneurons (FSINs) causing reduced task-evoked gamma-oscillation in prefrontal cortex. The voltage-gated sodium channel NaV1.1 is highly expressed in PV-positive interneurons, but only at low levels in principal cells. Positive modulators of Nav1.1 channels are for this reason considered potential candidates for the treatment of cognitive disorders. Here we examined the effect of the novel positive modulator of voltage-gated sodium channels Lu AE98134. We found that Lu AE98134 facilitated the sodium current mediated by NaV1.1 expressed in HEK cells by shifting its activation to more negative values, decreasing its inactivation kinetics and promoting a persistent inward current. In a slice preparation from the brain of adult mice, Lu AE98134 promoted the excitability of fast spiking interneurons by decreasing the threshold for action potentials. We then tested if Lu AE98134 could normalize the altered firing properties of FSINs in Dlx5/6+/- mutant mice. FSINs of this model for schizophrenia are characterized by broader action potentials and higher spike threshold. We found that in the presence of Lu AE98134, the firing frequency was increased while the spike duration and the threshold were decreased. Compounds with similar mode of action appear as promising candidates for restoring cognitive deficits present in schizophrenia.


Subject(s)
Heterocyclic Compounds, 3-Ring/pharmacology , NAV1.1 Voltage-Gated Sodium Channel/metabolism , Sulfonamides/pharmacology , Animals , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Mice, Mutant Strains
3.
Eur J Neurosci ; 46(3): 1887-1896, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28635024

ABSTRACT

Nav 1.1 (SCN1A) channels primarily located in gamma-aminobutyric acid (GABA)ergic fast-spiking interneurons are pivotal for action potential generation and propagation in these neurons. Inappropriate function of fast-spiking interneurons, leading to disinhibition of pyramidal cells and network desynchronization, correlates with decreased cognitive capability. Further, reduced functionality of Nav 1.1 channels is linked to various diseases in the central nervous system. There is, at present, however no subtype selective pharmacological activators of Nav 1.1 channels available for studying pharmacological modulation of interneuron function. In the current study, we identified a small molecule Nav 1.1 activator, 3-amino-5-(4-methoxyphenyl)thiophene-2-carboxamide, named AA43279, and provided an in vitro to in vivo characterization of the compound. In HEK-293 cells expressing human Nav 1.1 channels, AA43279 increased the Nav 1.1-mediated current in a concentration-dependent manner mainly by impairing the fast inactivation kinetics of the channels. In rat hippocampal brain slices, AA43279 increased the firing activity of parvalbumin-expressing, fast-spiking GABAergic interneurons and increased the spontaneous inhibitory post-synaptic currents (sIPSCs) recorded from pyramidal neurons. When tested in vivo, AA43279 had anti-convulsive properties in the maximal electroshock seizure threshold test. AA43279 was tested for off-target effects on 72 different proteins, including Nav 1.2, Nav 1.4, Nav 1.5, Nav 1.6 and Nav 1.7 and exhibited reasonable selectivity. Taken together, AA43279 might constitute a valuable tool compound for revealing biological functions of Nav 1.1 channels.


Subject(s)
Anticonvulsants/pharmacology , GABAergic Neurons/drug effects , Interneurons/drug effects , NAV1.1 Voltage-Gated Sodium Channel/metabolism , Seizures/drug therapy , Sodium Channel Blockers/pharmacology , Thiophenes/pharmacology , Action Potentials , Animals , Anticonvulsants/chemical synthesis , Anticonvulsants/therapeutic use , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiology , Excitatory Postsynaptic Potentials , GABAergic Neurons/metabolism , GABAergic Neurons/physiology , HEK293 Cells , Humans , Interneurons/metabolism , Interneurons/physiology , Male , Mice , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/chemical synthesis , Sodium Channel Blockers/therapeutic use
4.
Neuropharmacology ; 118: 102-112, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28242439

ABSTRACT

Due to their fast kinetic properties, Kv3.1 voltage gated potassium channels are important in setting and controlling firing frequency in neurons and pivotal in generating high frequency firing of interneurons. Pharmacological activation of Kv3.1 channels may possess therapeutic potential for treatment of epilepsy, hearing disorders, schizophrenia and cognitive impairments. Here we thoroughly investigate the selectivity and positive modulation of the two small molecules, EX15 and RE01, on Kv3 channels. Selectivity studies, conducted in Xenopus laevis oocytes confirmed a positive modulatory effect of the two compounds on Kv3.1 and to a minor extent on Kv3.2 channels. RE01 had no effect on the Kv3.3 and Kv3.4 channels, whereas EX15 had an inhibitory impact on the Kv3.4 mediated current. Voltage-clamp experiments in monoclonal hKv3.1b/HEK293 cells (34 °C) revealed that the two compounds indeed induced larger currents and faster activation kinetics. They also decrease the speed of deactivation and shifted the voltage dependence of activation, to a more negative activation threshold. Application of action potential clamping and repetitive stimulation protocols of hKv3.1b expressing HEK293 cells revealed that EX15 and RE01 significantly increased peak amplitude, half width and decay time of Kv3.1 mediated currents, even during high-frequency action potential clamping (250 Hz). In rat hippocampal slices, EX15 and RE01 increased neuronal excitability in fast-spiking interneurons in dentate gyrus. Action potential frequency was prominently increased at minor depolarizing steps, whereas more marginal effects of EX15 and RE01 were observed after stronger depolarizations. In conclusion, our results suggest that EX15 and RE01 positive modulation of Kv3.1 and Kv3.2 currents facilitate increased firing frequency in fast-spiking GABAergic interneurons.


Subject(s)
Action Potentials/physiology , Biophysical Phenomena/physiology , GABAergic Neurons/physiology , Hydantoins/pharmacology , Pyridines/pharmacology , Shaw Potassium Channels/metabolism , 2-Amino-5-phosphonovalerate/pharmacology , Action Potentials/drug effects , Animals , Biophysical Phenomena/drug effects , Brain/cytology , Excitatory Amino Acid Antagonists/pharmacology , GABA Antagonists/pharmacology , GABAergic Neurons/drug effects , HEK293 Cells , Humans , Kinetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oocytes , Pyridazines/pharmacology , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Repressor Proteins/pharmacology , Saccharomyces cerevisiae Proteins/pharmacology , Shaw Potassium Channels/genetics , Xenopus laevis
5.
PLoS One ; 10(3): e0120239, 2015.
Article in English | MEDLINE | ID: mdl-25798598

ABSTRACT

The 1,5-benzodiazepine clobazam is indicated for the adjunctive treatment of seizures associated with Lennox-Gastaut syndrome in patients 2 years of age or older in the United States, and for treatment of anxiety and various forms of epilepsy elsewhere. Clobazam has been reported to exhibit different in vivo adverse effects and addiction liability profile than the classic 1,4-benzodiazepines. In this study, it was investigated whether the in vitro pharmacological properties of clobazam and its major active metabolite N-desmethylclobazam could explain some of these clinical differences. The functional properties of the two 1,5-benzodiazepines were characterized at the human γ-aminobutyric acid type A receptor (GABA(A)R) subtypes α1ß2γ(2S), α2ß2γ(2S), α3ß2γ(2S), α5ß2γ(2S) and α6ß2δ expressed in Xenopus laevis oocytes by use of two-electrode voltage-clamp electrophysiology and compared to those exhibited by the 1,4-benzodiazepine clonazepam. All three compounds potentiated GABA EC20-evoked responses through the α(1,2,3,5)ß2γ(2S) GABA(A)Rs in a reversible and concentration-dependent manner, with each displaying similar EC50 values at the four subtypes. Furthermore, the degrees of potentiation of the GABA EC20 currents through the four receptors mediated by saturating modulator concentrations did not differ substantially for any of the three benzodiazepines. The three compounds were substantially less potent (200-3900 fold) as positive allosteric modulators at the α6ß2δ GABA(A)R than at the α(1,2,3,5)ß2γ(2S) receptors. Interestingly, however, clobazam and especially N-desmethylclobazam were highly efficacious potentiators of α6ß2δ receptor signaling. Although this activity component is unlikely to contribute to the in vivo effects of clobazam/N-desmethylclobazam, the 1,5-benzodiazepine could constitute an interesting lead for novel modulators targeting this low-affinity binding site in GABAARs. In conclusion, the non-selective modulation exerted by clobazam, N-desmethylclobazam and clonazepam at the α1ß2γ(2S), α2ß2γ(2S), α3ß2γ(2S) and α5ß2γ(2S) GABA(A)Rs indicate that the observed clinical differences between clobazam and 1,4-benzodiazepines are likely to arise from factors other than their respective pharmacological properties at the GABA(A)Rs as investigated here.


Subject(s)
Benzodiazepines/metabolism , Benzodiazepines/pharmacology , Oocytes/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Xenopus laevis/genetics , Animals , Clobazam , GABA-A Receptor Agonists/metabolism , GABA-A Receptor Agonists/pharmacology , GABA-A Receptor Antagonists/metabolism , GABA-A Receptor Antagonists/pharmacology , Gene Expression , Humans
6.
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
7.
PLoS One ; 9(2): e88456, 2014.
Article in English | MEDLINE | ID: mdl-24533090

ABSTRACT

Clobazam (CLB), a 1,5-benzodiazepine (BZD), was FDA-approved in October 2011 for the adjunctive treatment of seizures associated with Lennox-Gastaut syndrome (LGS) in patients 2 years and older. BZDs exert various CNS effects through allosteric modulation of GABAA receptors. The structurally distinct, 1,4-BZD clonazepam (CLN) is also approved to treat LGS. The precise mechanisms of action and clinical efficacy of both are unknown. Data show that the GABAA α1-subunit-selective compound zolpidem [ZOL] exhibits hypnotic/sedative effects. Conversely, data from knock-in mice carrying BZD binding site mutations suggest that the α2 subunit mediates anticonvulsant effects, without sedative actions. Hence, the specific pattern of interactions across the GABAA receptor complexes of BZDs might be reflected in their clinical efficacies and adverse effect profiles. In this study, GABAA-receptor binding affinities of CLB, N-desmethylclobazam (N-CLB, the major metabolite of CLB), CLN, and ZOL were characterized with native receptors from rat-brain homogenates and on cloned receptors from HEK293 cells transfected with combinations of α (α1, α2, α3, or α5), ß2, and γ2 subtypes. Our results demonstrate that CLB and N-CLB have significantly greater binding affinities for α2- vs. α1-receptor complexes, a difference not observed for CLN, for which no distinction between α2 and α1 receptors was observed. Our experiments with ZOL confirmed the high preference for α1 receptors. These results provide potential clues to a new understanding of the pharmacologic modes of action of CLB and N-CLB.


Subject(s)
Anticonvulsants/pharmacology , Benzodiazepines/pharmacology , Allosteric Site , Animals , Anticonvulsants/chemistry , Benzodiazepines/chemistry , Binding Sites , Brain/metabolism , Clobazam , Clonazepam/chemistry , Clonazepam/pharmacology , HEK293 Cells , Humans , Kinetics , Male , Mice , Mutation , Protein Binding , Pyridines/chemistry , Pyridines/pharmacology , Rats , Receptors, GABA-A/metabolism , Seizures/drug therapy , Zolpidem
8.
J Alzheimers Dis ; 40(2): 429-42, 2014.
Article in English | MEDLINE | ID: mdl-24448785

ABSTRACT

The formation of neurofibrillary tangles from the assembly of hyperphosphorylated tau leads to dendritic and axonal instability, synaptic degeneration, and neuronal loss. To understand the early physiological consequences of aberrant tau expression, we characterized the physiology of CA1 pyramidal neurons in rTg4510 female mice and non-transgenic (wt) littermate controls. We studied mice at the age of 10-12 weeks where only minimal hyperphosphorylated pretangle tau was present, and 22-24 weeks old mice with significant neurofibrillary tangle pathology. Our electrophysiological analysis included input-output relation, paired-pulse facilitation, and whole cell patch-clamp recordings of neurons to measure action potential threshold and action potential properties, chord-conductance, and characterization of AMPA receptor mediated synaptic transmission. We found that the input-output relation in field (excitatory postsynaptic potentials, EPSP) and whole cell recordings (excitatory postsynaptic currents, EPSC) were impaired in rTg4510 mice compared to wt controls at both ages. We measured a diminished tail current charge after depolarizing voltage input in rTg4510 mice compared to wt in both young and aged mice. Additionally, mini-EPSC properties (peak and decay time) were essentially similar between genotypes and age groups investigated. Surprisingly, in the 22-24 week old group, the mini-EPSC frequency was significantly increased (interevent interval 0.8 ± 0.1 in wt compared to 0.3 ± 0.1 in rTg4510 mice). These data indicate that the developmentally regulated expression of human P301L tau in CA1 pyramidal neurons coincide with changes in neuronal excitability but also that significant presynaptic changes occur late during the progression of tau pathology in this mouse model.


Subject(s)
CA1 Region, Hippocampal/pathology , Excitatory Postsynaptic Potentials/physiology , Pyramidal Cells/physiopathology , Tauopathies/pathology , tau Proteins/metabolism , Age Factors , Animals , Animals, Newborn , Biophysics , Disease Models, Animal , Doxycycline/pharmacology , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Female , Humans , In Vitro Techniques , Mice , Mice, Transgenic , Patch-Clamp Techniques , Tauopathies/genetics , tau Proteins/genetics
9.
Anal Biochem ; 418(1): 66-72, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21782781

ABSTRACT

To develop a real-time thallium flux assay for high-throughput screening (HTS) of human KCNQ4 (Kv7.4) potassium channel openers, we used CHO-K1 cells stably expressing human KCNQ4 channel protein and a thallium-sensitive dye based on the permeability of thallium through potassium channels. The electrophysiological and pharmacological properties of the cell line expressing the KCNQ4 protein were found to be in agreement with that reported elsewhere. The EC(50) values of the positive control compound (retigabine) determined by the thallium and (86)rubidium flux assays were comparable to and consistent with those documented in the literature. Signal-to-background (S/B) ratio and Z factor of the thallium influx assay system were assessed to be 8.82 and 0.63, respectively. In a large-scale screening of 98,960 synthetic and natural compounds using the thallium influx assay, 76 compounds displayed consistent KCNQ4 activation, and of these 6 compounds demonstrated EC(50) values of less than 20 µmol/L and 2 demonstrated EC(50) values of less than 1 µmol/L. Taken together, the fluorescence-based thallium flux assay is a highly efficient, automatable, and robust tool to screen potential KCNQ4 openers. This approach may also be expanded to identify and evaluate potential modulators of other potassium channels.


Subject(s)
KCNQ Potassium Channels/chemistry , Thallium/chemistry , Animals , CHO Cells , Cricetinae , Drug Evaluation, Preclinical/methods , Fluorescence , High-Throughput Screening Assays , Humans , Ion Channel Gating , KCNQ Potassium Channels/metabolism , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Thallium/metabolism
10.
Pharmacol Res ; 64(3): 268-73, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21504793

ABSTRACT

The stability and transcriptional activity of the hypoxia-inducible factors (HIFs) are regulated by oxygen-dependent hydroxylation that is catalyzed by three HIF prolyl 4-hydroxylases (HPHs). Use of HPH inhibition as a mean for HIF-upregulation has recently gained interest as a potential treatment paradigm against neurodegenerative diseases like ischemia and Parkinson's disease. In the present investigation we report the development of a new and robust assay to measure HPH activity. The assay is based on capture of hydroxylated peptide product by the von Hippel-Lindau protein which is directly measured in a scintillation proximity assay. In addition we describe the determination of HPH subtype potencies of HPH inhibitors which either directly or indirectly inhibit the HPH enzyme. The potencies of the HPH inhibitors displayed almost identical IC(50) values toward the HPH1 and HPH2 subtype while the potency against the HPH3 subtype was increased for several of the compounds. For the most potent compound, a hydroxyl thiazole derivative, the potency against HPH2 and HPH3 was 7nM and 0.49nM, respectively corresponding to a 14-fold difference. These results suggest that HPH subtype-selective compounds may be developed. In addition we determined the 2-oxoglutarate concentration in brain tissue and neuronal cell lines as 2-oxoglutarate is an important co-factor used by the HPH enzyme during the hydroxylation reaction. The high intracellular 2-oxoglutarate concentration provides an explanation for the diminished cellular HIF activating potency of a competitive HPH inhibitor compared to its orders of magnitude higher HPH inhibiting potency. The present reported data suggest that in the development of specific Hif prolyl hydroxylase inhibitors the high 2-oxoglutarate tissue level should be taken into account as this might affect the cellular potency. Thus to specifically inhibit the intracellular HPH enzymatic reaction a competitive inhibitor with a low Ki should be developed.


Subject(s)
Dioxygenases/metabolism , Enzyme Inhibitors/pharmacology , Ketoglutaric Acids/metabolism , Nuclear Proteins/metabolism , Procollagen-Proline Dioxygenase/metabolism , Cell Line , Dioxygenases/antagonists & inhibitors , Dioxygenases/genetics , Drug Evaluation, Preclinical/methods , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Procollagen-Proline Dioxygenase/genetics , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
11.
Pharmacology ; 86(3): 174-81, 2010.
Article in English | MEDLINE | ID: mdl-20714208

ABSTRACT

BACKGROUND/AIMS: the neuronal K(V)7 family members (K(V)7.2-5) are important regulators of neuronal excitability. K(V)7 channel openers are therefore attractive drug candidates for the treatment of several hyperexcitability disorders. While most described K(V)7 channel openers discriminate poorly between K(V)7.2-5, Icagen's N-(6-chloropyridin- 3-yl)-3,4-difluorobenzamide (ICA-27243) is more potent at K(V)7.2/3 than at K(V)7.4 and K(V)7.3/5 and offers some progress towards subtype selectivity. We have investigated its mode of action on K(V)7.2 and K(V)7.4, compared its effect to that of retigabine and studied the combinatorial effect of retigabine and ICA-27243, as these two compounds recognize different binding sites in the channels. METHODS: the effects of ICA-27243 and retigabine were studied using voltage-clamp electrophysiology in Xenopus laevis oocytes and rubidium flux in Chinese hamster ovary cells. RESULTS: we found that in contrast to retigabine's voltage-dependent action on K(V)7.2, ICA-27243 induced a voltage-independent current on this subtype, which was not observed on K(V)7.4. Additionally, the combined treatment of K(V)7.2 and K(V)7.4 with retigabine and ICA-27243 revealed that the effect of ICA-27243 on K(V)7.2 dominates that of retigabine, while the compounds act additively and synergistically on K(V)7.4. CONCLUSIONS: these results offer further detailed insight into pharmacological activation of K(V)7 channels and offer evidence of differential functional and subtype-specific effects by activation of different binding sites in the K(V)7 channels.


Subject(s)
Benzamides/pharmacology , KCNQ Potassium Channels/metabolism , KCNQ2 Potassium Channel/metabolism , Pyridines/pharmacology , Animals , Benzamides/metabolism , Binding Sites , CHO Cells , Carbamates/metabolism , Carbamates/pharmacology , Cells, Cultured , Cloning, Molecular , Cricetinae , Cricetulus , Female , Membrane Potentials/drug effects , Oocytes/metabolism , Phenylenediamines/metabolism , Phenylenediamines/pharmacology , Pyridines/metabolism , Rubidium Radioisotopes/metabolism , Xenopus laevis
12.
PLoS One ; 4(12): e8251, 2009 Dec 11.
Article in English | MEDLINE | ID: mdl-20011514

ABSTRACT

BACKGROUND: Activation of voltage-gated potassium channels of the Kv7 (KCNQ) family reduces cellular excitability. These channels are therefore attractive targets for treatment of diseases characterized by hyperexcitability, such as epilepsy, migraine and neuropathic pain. Retigabine, which opens Kv7.2-5, is now in clinical trial phase III for the treatment of partial onset seizures. One of the main obstacles in developing Kv7 channel active drugs has been to identify compounds that can discriminate between the neuronal subtypes, a feature that could help diminish side effects and increase the potential of drugs for particular indications. METHODOLOGY/PRINCIPAL FINDINGS: In the present study we have made a thorough investigation of the Bristol-Myers Squibb compound (S)-N-[1-(4-Cyclopropylmethyl-3,4-dihydro-2H-benzo[1], [4]oxazin-6-yl)-ethyl]-3-(2-fluoro-phenyl)-acrylamide [(S)-2] on human Kv7.1-5 channels expressed in Xenopus laevis oocytes. We found that the compound was a weak inhibitor of Kv7.1. In contrast, (S)-2 efficiently opened Kv7.2-5 by producing hyperpolarizing shifts in the voltage-dependence of activation and enhancing the maximal current amplitude. Further, it reduced inactivation, accelerated activation kinetics and slowed deactivation kinetics. The mechanisms of action varied between the subtypes. The enhancing effects of (S)-2 were critically dependent on a tryptophan residue in S5 also known to be crucial for the effects of retigabine, (S)-1 and BMS-204352. However, while (S)-2 did not at all affect a mutant Kv7.4 with a leucine in this position (Kv7.4-W242L), a Kv7.2 with the same mutation (Kv7.2-W236L) was inhibited by the compound, showing that (S)-2 displays a subtype-selective interaction with in the Kv7 family. CONCLUSIONS/SIGNIFICANCE: These results offer further insight into pharmacological activation of Kv7 channels, add to the understanding of small molecule interactions with the channels and may contribute to the design of subtype selective modulators.


Subject(s)
Acrylamide/pharmacology , Acrylamides/pharmacology , KCNQ Potassium Channels/metabolism , Oocytes/drug effects , Oocytes/metabolism , Oxazines/pharmacology , Acrylamide/chemistry , Acrylamides/chemistry , Animals , Binding Sites , Carbamates/metabolism , Humans , Ion Channel Gating/drug effects , Kinetics , Membrane Potentials/drug effects , Mutant Proteins/metabolism , Oxazines/chemistry , Phenylenediamines/metabolism , Tryptophan/metabolism , Xenopus laevis
13.
Biophys J ; 92(8): 2747-56, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17237198

ABSTRACT

Voltage-gated K(+) channels of the Kv7 (KCNQ) family have important physiological functions in both excitable and nonexcitable tissue. The family encompasses five genes encoding the channel subunits Kv7.1-5. Kv7.1 is found in epithelial and cardiac tissue. Kv7.2-5 channels are predominantly neuronal channels and are important for controlling excitability. Kv7.1 channels have been considered the only Kv7 channels to undergo inactivation upon depolarization. However, here we demonstrate that inactivation is also an intrinsic property of Kv7.4 and Kv7.5 channels, which inactivate to a larger extent than Kv7.1 channels at all potentials. We demonstrate that at least 30% of these channels are inactivated at physiologically relevant potentials. The onset of inactivation is voltage dependent and occurs on the order of seconds. Both time- and voltage-dependent recovery from inactivation was investigated for Kv7.4 channels. A time constant of 1.47 +/- 0.21 s and a voltage constant of 54.9 +/- 3.4 mV were determined. It was further demonstrated that heteromeric Kv7.3/Kv7.4 channels had inactivation properties different from homomeric Kv7.4 channels. Finally, the Kv7 channel activator BMS-204352 was in contrast to retigabine found to abolish inactivation of Kv7.4. In conclusion, this work demonstrates that inactivation is a key regulatory mechanism of Kv7.4 and Kv7.5 channels.


Subject(s)
Ion Channel Gating/physiology , KCNQ Potassium Channels/physiology , Membrane Potentials/physiology , Oocytes/physiology , Animals , Cells, Cultured , Humans , Xenopus laevis
14.
Brain Res Mol Brain Res ; 141(1): 66-73, 2005 Nov 18.
Article in English | MEDLINE | ID: mdl-16154661

ABSTRACT

Potassium channels containing the KCNQ2 subunit play an important role in the regulation of neuronal excitability and therefore have been implicated in epilepsy. This study describes the expression of KCNQ2 subunit immunoreactivity in the basolateral amygdala in two rat models of temporal lobe epilepsy, (1) amygdala kindling and (2) spontaneously epileptic rats after status epilepticus induced by hippocampal electrical stimulation. KCNQ2 subunit immunoreactivity was assessed with a commercial antibody raised against a C-terminal part of the KCNQ2 protein. We show that KCNQ2 subunit immunoreactivity is upregulated in the basolateral amygdala in both models and that generalized seizures are required to induce this upregulation. We hypothesize that the upregulation of potassium channels containing the KCNQ2 subunit might represent a mechanism to counteract seizures in experimental temporal lobe epilepsy.


Subject(s)
Amygdala/metabolism , Epilepsy, Temporal Lobe/metabolism , KCNQ2 Potassium Channel/metabolism , Amygdala/cytology , Animals , Disease Models, Animal , Epilepsy, Temporal Lobe/physiopathology , Humans , Immunohistochemistry , Kindling, Neurologic , Male , Rats , Rats, Wistar , Up-Regulation
15.
Brain Res Mol Brain Res ; 139(1): 52-62, 2005 Sep 13.
Article in English | MEDLINE | ID: mdl-15963599

ABSTRACT

The KCNQ proteins compose a sub-group of the voltage-activated potassium channel family. The family consists of five members (KCNQ1 to 5--also named Kv7.1 to Kv7.5) encoded by single genes, which all give rise to proteins forming slowly activating potassium-selective ion channels. The physiological importance of the KCNQ channel family is emphasized by the fact that mutations in four of the five genes have been linked to human pathologies (KCNQ1 to 4). Here, we present the cloning and characterization of a novel KCNQ5 ortholog from mouse isolated by homology cloning from total mouse brain RNA (GenBank accession number: AY679158). The predicted protein is 95% identical to human KCNQ5. Upon expression in Xenopus oocytes, these proteins form voltage-dependent slowly activating channels with half-maximal activation at -21 mV. Our functional characterization revealed three novel modes of modulation: pH-dependent potentiation by Zn2+ (EC50 = 21.8 microM at pH 7.4), inhibition by acidification (IC50 = 0.75 microM; pKa = 6.1), and regulation by small changes in cell volume. Furthermore, the channels are activated by the anti-convulsant drug retigabine (EC50 = 2.0 microM) and inhibited by the M-current blockers linopiridine and XE-991. Finally, real-time RT-PCR was used to quantify the expression profile in a wide range of mouse tissues. These experiments revealed a relatively broad expression pattern in the nervous system but also expression in other tissues. Highest overall expression levels were observed in cortex and hippocampus. This study shows that murine KCNQ5 channels, in addition to sharing biophysical and pharmacological characteristics with the human ortholog, are tightly regulated by physiological stimuli such as changes in extracellular Zn2+, pH, and tonicity, thus adding to the complex regulation of these channels.


Subject(s)
Cell Size , KCNQ Potassium Channels/metabolism , Zinc/metabolism , Animals , Cloning, Molecular , Humans , Hydrogen-Ion Concentration , KCNQ Potassium Channels/genetics , Mice , Molecular Sequence Data , Oocytes/physiology , Patch-Clamp Techniques , RNA, Messenger/metabolism , Sequence Alignment , Tissue Distribution , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...