Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Assoc Res Otolaryngol ; 22(2): 127-139, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33575914

RESUMO

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.


Assuntos
Perda Auditiva , Canal de Potássio KCNQ2/agonistas , Canal de Potássio KCNQ3/agonistas , Ruído/efeitos adversos , Zumbido , Animais , Perda Auditiva/tratamento farmacológico , Perda Auditiva/etiologia , Camundongos , Zumbido/tratamento farmacológico , Zumbido/etiologia
2.
Commun Biol ; 3(1): 356, 2020 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-32641720

RESUMO

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.


Assuntos
Canais de Potássio KCNQ/agonistas , Quercetina/farmacologia , Animais , Sítios de Ligação , Capparis/química , Canais de Potássio KCNQ/química , Canal de Potássio KCNQ2/agonistas , Canal de Potássio KCNQ3/agonistas , Oócitos , Técnicas de Patch-Clamp , Extratos Vegetais/farmacologia , Estrutura Terciária de Proteína , Rutina/farmacologia , Xenopus laevis
3.
J Med Chem ; 63(1): 163-185, 2020 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-31815462

RESUMO

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.


Assuntos
Indóis/farmacologia , Canal de Potássio KCNQ2/agonistas , Canal de Potássio KCNQ3/agonistas , Animais , Células CHO , Carbamatos/química , Cricetulus , Indóis/síntese química , Indóis/metabolismo , Canal de Potássio KCNQ2/genética , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ3/metabolismo , Microssomos Hepáticos/metabolismo , Modelos Moleculares , Conformação Molecular , Mutação , Fenilenodiaminas/química , Ligação Proteica , Bibliotecas de Moléculas Pequenas/síntese química , Bibliotecas de Moléculas Pequenas/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Relação Estrutura-Atividade , Xenopus laevis
4.
Eur J Pharmacol ; 858: 172474, 2019 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-31238068

RESUMO

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.


Assuntos
Fenômenos Eletrofisiológicos , Células-Tronco Pluripotentes Induzidas/citologia , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ3/metabolismo , Neurônios/citologia , Receptor Muscarínico M1/metabolismo , Animais , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Canal de Potássio KCNQ2/agonistas , Canal de Potássio KCNQ2/antagonistas & inibidores , Canal de Potássio KCNQ2/genética , Canal de Potássio KCNQ3/agonistas , Canal de Potássio KCNQ3/antagonistas & inibidores , Canal de Potássio KCNQ3/genética , Antagonistas Muscarínicos/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Receptor Muscarínico M1/agonistas , Receptor Muscarínico M1/antagonistas & inibidores
5.
FASEB J ; 33(8): 9154-9166, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31063701

RESUMO

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.


Assuntos
Anticonvulsivantes/farmacologia , Canal de Potássio KCNQ2/agonistas , Canal de Potássio KCNQ3/agonistas , Moduladores de Transporte de Membrana/farmacologia , Piridinas/farmacologia , Substituição de Aminoácidos , Animais , Anticonvulsivantes/síntese química , Anticonvulsivantes/química , Células Cultivadas , Canal de Potássio ERG1/antagonistas & inibidores , Epilepsia/tratamento farmacológico , Células HEK293 , Humanos , Canais de Potássio KCNQ/agonistas , Canal de Potássio KCNQ2/genética , Canal de Potássio KCNQ2/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Moduladores de Transporte de Membrana/síntese química , Moduladores de Transporte de Membrana/química , Camundongos , Mutagênese Sítio-Dirigida , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Técnicas de Patch-Clamp , Piridinas/síntese química , Piridinas/química , Ratos , Convulsões/tratamento farmacológico
6.
Behav Pharmacol ; 28(5): 405-407, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28125509

RESUMO

The efficacy of KCNQ2/3 channel agonists against drug reward has not been defined despite their ability to reduce locomotor-stimulant and dopamine-activating effects of psychostimulants. We tested the hypothesis that flupirtine (FLU) (2.5, 10, 20 mg/kg), a KCNQ2/3 agonist, reduces cocaine (15 mg/kg) conditioned place preference. FLU (20 mg/kg), injected concurrently with cocaine during conditioning, reduced the development of cocaine conditioned place preference. FLU (20 mg/kg) also reduced cocaine locomotor activation without affecting baseline activity. The disruption of cocaine place preference by FLU suggests that KCNQ2/3 channels influence cocaine's rewarding effects.


Assuntos
Aminopiridinas/farmacologia , Canal de Potássio KCNQ3/efeitos dos fármacos , Aminopiridinas/metabolismo , Animais , Estimulantes do Sistema Nervoso Central/farmacologia , Cocaína/farmacologia , Condicionamento Operante/efeitos dos fármacos , Dopaminérgicos/farmacologia , Relação Dose-Resposta a Droga , Canal de Potássio KCNQ3/agonistas , Locomoção/efeitos dos fármacos , Masculino , Atividade Motora/efeitos dos fármacos , Ratos , Receptores Dopaminérgicos/efeitos dos fármacos , Recompensa
7.
J Pharm Pharmacol ; 68(4): 494-502, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26969140

RESUMO

OBJECTIVES: This study investigated the effect of tannic acid (TA), a plant-derived hydrolyzable polyphenol, on Kv7.4 and Kv7.5 K(+) channels and rat mesenteric artery. METHODS: Whole-cell patch clamp experiments were used to record the Kv7.4 and Kv7.3/7.5 K(+) currents expressed in HEK293 cells; and the tension changes of mesenteric arteries isolated from rats were recorded using small vessel myography apparatus. KEY FINDINGS: Tannic acid increases the Kv7.4 and Kv7.3/7.5 K(+) currents in a concentration-dependent manner (median effective concentration (EC50 ) = 27.3 ± 3.6 µm and EC50 = 23.1 ± 3.9 µm, respectively). In addition, 30 µm TA shifts the G-V curve of Kv7.4 and Kv7.3/7.5 K(+) currents to the left by 14.18 and 25.24 mV, respectively, and prolongs the deactivation time constants by 184.44 and 154.77 ms, respectively. Moreover, TA relaxes the vascular tension of rat mesenteric arteries in a concentration-dependent manner (half inhibitory concentration (IC50 ) = 148.7 ± 13.4 µm). CONCLUSION: These results confirms the vasodilatory effects of TA on rat mesenteric artery and the activating effects on the Kv7.4 and Kv7.3/7.5 K(+) channels, which may be a mechanism to explain the vasodilatory effect and this mechanism can be used in the research of antihypertension.


Assuntos
Anti-Hipertensivos/farmacologia , Canais de Potássio KCNQ/agonistas , Canal de Potássio KCNQ3/agonistas , Artérias Mesentéricas/efeitos dos fármacos , Taninos/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Animais , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Técnicas In Vitro , Canais de Potássio KCNQ/genética , Canais de Potássio KCNQ/metabolismo , Canal de Potássio KCNQ3/genética , Canal de Potássio KCNQ3/metabolismo , Masculino , Potenciais da Membrana , Miografia , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Ratos Sprague-Dawley , Fatores de Tempo , Transfecção
8.
Acta Physiol (Oxf) ; 218(1): 28-37, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26914447

RESUMO

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.


Assuntos
Ácidos Graxos Insaturados/farmacologia , Canal de Potássio KCNQ2/agonistas , Canal de Potássio KCNQ3/agonistas , Animais , Ácidos Graxos Ômega-3/farmacologia , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Técnicas de Patch-Clamp , Xenopus laevis
9.
J Gen Physiol ; 147(3): 229-41, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26880756

RESUMO

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.


Assuntos
Anticonvulsivantes/farmacologia , Carbamatos/farmacologia , Canal de Potássio KCNQ2/agonistas , Canal de Potássio KCNQ3/agonistas , Potenciais da Membrana , Fenilenodiaminas/farmacologia , Animais , Humanos , Ativação do Canal Iônico , Canal de Potássio KCNQ2/química , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ3/química , Canal de Potássio KCNQ3/metabolismo , Domínios Proteicos , Multimerização Proteica , Xenopus
10.
Eur J Pharmacol ; 764: 633-642, 2015 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-26134502

RESUMO

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.


Assuntos
Analgésicos/farmacologia , Canais de Cloreto/antagonistas & inibidores , Gânglios Espinais/efeitos dos fármacos , Canais de Potássio KCNQ/agonistas , Nociceptividade/efeitos dos fármacos , Dor Nociceptiva/tratamento farmacológico , Células Receptoras Sensoriais/efeitos dos fármacos , Taninos/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Canais de Sódio Disparados por Voltagem/efeitos dos fármacos , Animais , Anoctamina-1 , Comportamento Animal/efeitos dos fármacos , Bradicinina , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Gânglios Espinais/metabolismo , Gânglios Espinais/fisiopatologia , Células HEK293 , Humanos , Canais de Potássio KCNQ/genética , Canais de Potássio KCNQ/metabolismo , Canal de Potássio KCNQ2/agonistas , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ3/agonistas , Canal de Potássio KCNQ3/metabolismo , Potenciais da Membrana , Dor Nociceptiva/induzido quimicamente , Dor Nociceptiva/metabolismo , Dor Nociceptiva/fisiopatologia , Dor Nociceptiva/psicologia , Ratos Sprague-Dawley , Células Receptoras Sensoriais/metabolismo , Transfecção , Canais de Sódio Disparados por Voltagem/metabolismo
11.
Neurosci Lett ; 545: 59-63, 2013 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-23623937

RESUMO

Spontaneous involuntary dystonic and choreatic movements induced by L-DOPA (L-DOPA-induced dyskinesias (LID)) represent a severe complication of long-time pharmacotherapy in Parkinson's disease that deserves novel therapeutics. Previous studies demonstrated antidyskinetics effect of the KV7.2-7.5 channel opener retigabine after acute and chronic treatment in a rat model of LID. We hypothesized that this effect was mainly mediated by KV7.2/3 channels located on striatal projection neurons, as an increased activity of these neurons seems to be involved in the pathophysiology of LID. We therefore examined the acute effects of the KV7.2/3 preferring channel opener ICA 27243 (N-(6-chloro-pyridin-3-yl)-3,4-difluoro-benzamide, 5-15 mg/kg i.p.) on LID in this animal model. Ten and 15 mg ICA 27243 significantly reduced abnormal involuntary movements (AIM) while no negative impact on the antiparkinsonian effect of L-DOPA was observed. However, at the end of the testing session (180 min) AIM scores increased after application of both doses. Further studies have to clarify if this can be avoided by a different application regime. Nevertheless, the present results suggest that selective openers of KV7.2/3 channels might be interesting candidates for the treatment of LID as antidyskinetic effects occurred at well-tolerated doses and did not interfere with the antiparkinsonian effect of L-DOPA.


Assuntos
Benzamidas/administração & dosagem , Discinesia Induzida por Medicamentos/tratamento farmacológico , Discinesia Induzida por Medicamentos/metabolismo , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ3/metabolismo , Transtornos Parkinsonianos/tratamento farmacológico , Transtornos Parkinsonianos/metabolismo , Piridinas/administração & dosagem , Animais , Discinesia Induzida por Medicamentos/etiologia , Feminino , Ativação do Canal Iônico/efeitos dos fármacos , Canal de Potássio KCNQ2/agonistas , Canal de Potássio KCNQ3/agonistas , Levodopa , Transtornos Parkinsonianos/induzido quimicamente , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento
12.
J Gen Physiol ; 140(1): 41-53, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22689829

RESUMO

Low-threshold voltage-gated M-type potassium channels (M channels) are tetraheteromers, commonly of two Kv7.2 and two Kv7.3 subunits. Though gated by voltage, the channels have an absolute requirement for binding of the membrane phospholipid phosphatidylinositol-4,5-bisphosphate (PI(4,5)P(2)) to open. We have investigated the quantitative relation between the concentration of a water-soluble PI(4,5)P(2) analog, dioctanoyl-PI(4,5)P(2) (DiC(8)-PI(4,5)P(2)), and channel open probability (P(open)) by fast application of increasing concentrations of DiC(8)-PI(4,5)P(2) to the inside face of membrane patches excised from Chinese hamster ovary cells expressing M channels as heteromeric Kv7.2/7.3 subunits. The rationale for the experiments is that this will mimic the effect of changes in membrane PI(4,5)P(2) concentration. Single-channel conductances from channel current-voltage relations in cell-attached mode were 9.2 ± 0.1 pS with a 2.5-mM pipette [K(+)]. Plots of P(open) against DiC(8)-PI(4,5)P(2) concentration were best fitted using a two-component concentration-P(open) relationship with high and low affinity, half-maximal effective concentration (EC(50)) values of 1.3 ± 0.14 and 75.5 ± 2.5 µM, respectively, and Hill slopes of 1.4 ± 0.06. In contrast, homomeric channels from cells expressing only Kv7.2 or Kv7.3 constructs yielded single-component curves with EC(50) values of 76.2 ± 19.9 or 3.6 ± 1.0 µM, respectively. When wild-type (WT) Kv7.2 was coexpressed with a mutated Kv7.3 subunit with >100-fold reduced sensitivity to PI(4,5)P(2), the high-affinity component of the activation curve was lost. Fitting the data for WT and mutant channels to an activation mechanism with independent PI(4,5)P(2) binding to two Kv7.2 and two Kv7.3 subunits suggests that the two components of the M-channel activation curve correspond to the interaction of PI(4,5)P(2) with the Kv7.3 and Kv7.2 subunits, respectively, that channels can open when only the two Kv7.3 subunits have bound DiC(8)-PI(4,5)P(2), and that maximum channel opening requires binding to all four subunits.


Assuntos
Canal de Potássio KCNQ2/agonistas , Canal de Potássio KCNQ3/agonistas , Fosfatidilinositol 4,5-Difosfato/análogos & derivados , Animais , Sítios de Ligação , Células CHO , Cricetinae , Cricetulus , Ativação do Canal Iônico/efeitos dos fármacos , Canal de Potássio KCNQ2/química , Canal de Potássio KCNQ2/genética , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ3/química , Canal de Potássio KCNQ3/genética , Canal de Potássio KCNQ3/metabolismo , Mutação de Sentido Incorreto , Técnicas de Patch-Clamp , Fosfatidilinositol 4,5-Difosfato/farmacologia , Subunidades Proteicas/agonistas , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo
13.
PLoS One ; 6(9): e23898, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21915266

RESUMO

Of the five human KCNQ (Kv7) channels, KCNQ1 with auxiliary subunit KCNE1 mediates the native cardiac I(Ks) current with mutations causing short and long QT cardiac arrhythmias. KCNQ4 mutations cause deafness. KCNQ2/3 channels form the native M-current controlling excitability of most neurons, with mutations causing benign neonatal febrile convulsions. Drosophila contains a single KCNQ (dKCNQ) that appears to serve alone the functions of all the duplicated mammalian neuronal and cardiac KCNQ channels sharing roughly 50-60% amino acid identity therefore offering a route to investigate these channels. Current information about the functional properties of dKCNQ is lacking therefore we have investigated these properties here. Using whole cell patch clamp electrophysiology we compare the biophysical and pharmacological properties of dKCNQ with the mammalian neuronal and cardiac KCNQ channels expressed in HEK cells. We show that Drosophila KCNQ (dKCNQ) is a slowly activating and slowly-deactivating K(+) current open at sub-threshold potentials that has similar properties to neuronal KCNQ2/3 with some features of the cardiac KCNQ1/KCNE1 accompanied by conserved sensitivity to a number of clinically relevant KCNQ blockers (chromanol 293B, XE991, linopirdine) and opener (zinc pyrithione). We also investigate the molecular basis of the differential selectivity of KCNQ channels to the opener retigabine and show a single amino acid substitution (M217W) can confer sensitivity to dKCNQ. We show dKCNQ has similar electrophysiological and pharmacological properties as the mammalian KCNQ channels, allowing future study of physiological and pathological roles of KCNQ in Drosophila and whole organism screening for new modulators of KCNQ channelopathies.


Assuntos
Proteínas de Drosophila/metabolismo , Canais de Potássio KCNQ/metabolismo , Animais , Antracenos/farmacologia , Carbamatos/farmacologia , Linhagem Celular , Cromanos/farmacologia , Drosophila , Proteínas de Drosophila/agonistas , Proteínas de Drosophila/antagonistas & inibidores , Eletrofisiologia , Humanos , Indóis/farmacologia , Canais de Potássio KCNQ/agonistas , Canais de Potássio KCNQ/antagonistas & inibidores , Canal de Potássio KCNQ1/agonistas , Canal de Potássio KCNQ1/antagonistas & inibidores , Canal de Potássio KCNQ1/metabolismo , Canal de Potássio KCNQ2/agonistas , Canal de Potássio KCNQ2/antagonistas & inibidores , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ3/agonistas , Canal de Potássio KCNQ3/antagonistas & inibidores , Canal de Potássio KCNQ3/metabolismo , Compostos Organometálicos/farmacologia , Técnicas de Patch-Clamp , Fenilenodiaminas/farmacologia , Piridinas/farmacologia , Sulfonamidas/farmacologia
14.
Pharmacology ; 87(5-6): 297-310, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21577044

RESUMO

The voltage-gated M-type potassium channel, encoded mainly by the KCNQ2/3 genes, plays an important role in the control of neuronal excitability. Mutations in the KCNQ2 gene lead to a form of neonatal epilepsy in humans termed 'benign familial neonatal convulsions', which is characterized by hyperexcitability of neurons. KCNQ openers or activators are expected to decrease the firing of overactive neurons and are thus conducive to the treatment of epilepsy and pain. Here, we report that four novel synthesized derivatives of pyrazolo[1,5-a]pyrimidin-7(4H)-one (PPO) named QO-26, QO-28, QO-40 and QO-41 potently augmented KCNQ2/3 channels expressed in Chinese hamster ovary cells and shifted the half-maximal activation voltage (V(1/2)) in the hyperpolarizing direction. The V(1/2) was negatively shifted in a concentration-dependent manner. The compounds markedly slowed both KCNQ2/3 channel activation and deactivation kinetics. Structure-activity relationship studies suggest that trifluoromethyl at the C-2 position, phenyl or naphthyl at the C-3 position, and trifluoromethyl or chloromethyl at the C-5 position are essential for the activity. These results suggest the four PPO derivatives act as KCNQ2/3 channel openers, providing a new dimension for the design and development of more potent channel openers.


Assuntos
Canal de Potássio KCNQ2/agonistas , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ3/agonistas , Canal de Potássio KCNQ3/metabolismo , Pirimidinonas/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Células CHO , Cricetinae , Cricetulus , Humanos , Canal de Potássio KCNQ2/genética , Canal de Potássio KCNQ3/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Pirazóis/química , Pirazóis/farmacologia , Pirimidinas/química , Pirimidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade , Transfecção
15.
J Med Chem ; 53(2): 887-96, 2010 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-20020710

RESUMO

Current drugs for the treatment of seizure disorders, although effective in many patients, still suffer from a number of failures and are not effective in some forms of resistant epilepsies. Historically, many of these drugs have multiple mechanisms of action including calcium and sodium channel blockade as well as GABAergic activity and thus a number of associated side effects. Modulation of the M-current through opening of KCNQ channels has been proposed as a way to attenuate neuroexcitability and have a therapeutic benefit for the treatment of seizure disorders. Therefore, as part of our program to identify new treatments for epilepsy, we set out to identify agonists of KCNQ channels. High throughput screening of our corporate collection led to the identification of 1, adamantane-1-carboxylic acid (3-methyl-3H-benzothiazol-2-ylidine) hydrazide, a potent KCNQ2/Q3 agonist. Herein, we describe the syntheses and structure-activity relationships of analogues of 1 as well as their in vivo activity in animal models of epilepsy and neuropathic pain.


Assuntos
Epilepsia/tratamento farmacológico , Canal de Potássio KCNQ2/agonistas , Canal de Potássio KCNQ3/agonistas , Neuralgia/tratamento farmacológico , Adamantano/química , Adamantano/farmacologia , Animais , Azidas/química , Azidas/farmacologia , Camundongos , Relação Estrutura-Atividade
16.
J Pharmacol Exp Ther ; 326(3): 818-28, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18577704

RESUMO

Openers or activators of neuronal KCNQ2/Q3 potassium channels decrease neuronal excitability and may provide benefit in the treatment of disorders of neuronal excitability such as epilepsy. In the present study, we evaluate the effects of ICA-27243 [N-(6-chloro-pyridin-3-yl)-3,4-difluoro-benzamide], an orally bioavailable, potent, and selective KCNQ2/Q3 opener, in a broad range of rodent seizure models. ICA-27243 was effective against maximal electroshock (MES) and pentylenetetrazole (PTZ)-induced seizures in both rats (MES, ED(50) = 1.5 mg/kg p.o.; PTZ, ED(50) = 2.2 mg/kg p.o.) and mice (MES, ED(50) = 8.6 mg/kg p.o.; PTZ, ED(50) = 3.9 mg/kg p.o.) in the rat amygdala kindling model of partial seizures (full protection from seizure at 9 mg/kg p.o.) and in the 6-Hz model of psychomotor seizures in mice (active at 10 mg/kg i.p.). Antiseizure efficacy in all models was observed at doses significantly less than those shown to effect open-field locomotor activity (rat ED(50) = 40 mg/kg p.o.) or ability to remain on a Rotorod (no effect in rat at doses up to 100 mg/kg p.o.). There was no evidence of cognition impairment as measured in the Morris water maze in the rat (10 and 30 mg/kg p.o.), nor was there evidence of the development of tolerance after multiple doses of ICA-27243. Our findings suggest that selective KCNQ2/Q3 opening activity in the absence of effects on KCNQ3/Q5 or GABA-activated channels may be sufficient for broad-spectrum antiepileptic activity in rodents.


Assuntos
Anticonvulsivantes/farmacologia , Benzamidas/farmacologia , Modelos Animais de Doenças , Canal de Potássio KCNQ2/agonistas , Canal de Potássio KCNQ3/agonistas , Piridinas/farmacologia , Convulsões/prevenção & controle , Animais , Anticonvulsivantes/uso terapêutico , Benzamidas/uso terapêutico , Relação Dose-Resposta a Droga , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ3/metabolismo , Masculino , Camundongos , Piridinas/uso terapêutico , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Convulsões/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA