Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Molecules ; 29(13)2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38999185

RESUMO

The growing interest in Kv7.2/7.3 agonists originates from the involvement of these channels in several brain hyperexcitability disorders. In particular, Kv7.2/7.3 mutants have been clearly associated with epileptic encephalopathies (DEEs) as well as with a spectrum of focal epilepsy disorders, often associated with developmental plateauing or regression. Nevertheless, there is a lack of available therapeutic options, considering that retigabine, the only molecule used in clinic as a broad-spectrum Kv7 agonist, has been withdrawn from the market in late 2016. This is why several efforts have been made both by both academia and industry in the search for suitable chemotypes acting as Kv7.2/7.3 agonists. In this context, in silico methods have played a major role, since the precise structures of different Kv7 homotetramers have been only recently disclosed. In the present review, the computational methods used for the design of Kv.7.2/7.3 small molecule agonists and the underlying medicinal chemistry are discussed in the context of their biological and structure-function properties.


Assuntos
Canal de Potássio KCNQ2 , Canal de Potássio KCNQ3 , Humanos , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ2/genética , Canal de Potássio KCNQ2/química , Canal de Potássio KCNQ3/metabolismo , Canal de Potássio KCNQ3/genética , Canal de Potássio KCNQ3/química , Canal de Potássio KCNQ3/antagonistas & inibidores , Simulação por Computador , Relação Estrutura-Atividade , Descoberta de Drogas/métodos , Animais
2.
J Biol Chem ; 299(2): 102819, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36549648

RESUMO

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.


Assuntos
Cálcio , Calmodulina , Espaço Intracelular , Canais de Potássio KCNQ , Zinco , Sinalização do Cálcio , Calmodulina/metabolismo , Canais de Potássio KCNQ/antagonistas & inibidores , Canais de Potássio KCNQ/química , Canais de Potássio KCNQ/genética , Canais de Potássio KCNQ/metabolismo , Mutação , Ligação Proteica/genética , Zinco/farmacologia , Zinco/metabolismo , Espaço Intracelular/metabolismo , Cálcio/metabolismo , Canal de Potássio KCNQ2/antagonistas & inibidores , Canal de Potássio KCNQ2/química , Canal de Potássio KCNQ2/genética , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ3/antagonistas & inibidores , Canal de Potássio KCNQ3/química , Canal de Potássio KCNQ3/genética , Canal de Potássio KCNQ3/metabolismo
3.
Bioorg Med Chem Lett ; 29(23): 126681, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31668424

RESUMO

A set of novel Kv7.2/7.3 (KCNQ2/3) channel blockers was synthesized to address several liabilities of the known compounds XE991 (metabolic instability and CYP inhibition) and the clinical compound DMP 543 (acid instability, insolubility, and lipophilicity). Using the anthrone scaffold of the prior channel blockers, alternative heteroarylmethyl substituents were installed via enolate alkylation reactions. Incorporation of a pyridazine and a fluorinated pyridine gave an analog (compound 18, JDP-107) with a promising combination of potency (IC50 = 0.16 µM in a Kv7.2 thallium flux assay), efficacy in a Kv7.2/7.3 patch clamp assay, and drug-like properties.


Assuntos
Antracenos/farmacologia , Canal de Potássio KCNQ2/antagonistas & inibidores , Canal de Potássio KCNQ3/antagonistas & inibidores , Transtornos Mentais/tratamento farmacológico , Doenças Neurodegenerativas/tratamento farmacológico , Bloqueadores dos Canais de Potássio/farmacologia , Antracenos/síntese química , Antracenos/química , Relação Dose-Resposta a Droga , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ3/metabolismo , Estrutura Molecular , Bloqueadores dos Canais de Potássio/síntese química , Bloqueadores dos Canais de Potássio/química , Relação Estrutura-Atividade
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.
Neuron ; 98(5): 918-925.e3, 2018 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-29754751

RESUMO

Cholinergic interneurons (ChIs) of the striatum pause their firing in response to salient stimuli and conditioned stimuli after learning. Several different mechanisms for pause generation have been proposed, but a unifying basis has not previously emerged. Here, using in vivo and ex vivo recordings in rat and mouse brain and a computational model, we show that ChI pauses are driven by withdrawal of excitatory inputs to striatum and result from a delayed rectifier potassium current (IKr) in concert with local neuromodulation. The IKr is sensitive to Kv7.2/7.3 blocker XE-991 and enables ChIs to report changes in input, to pause on excitatory input recession, and to scale pauses with input strength, in keeping with pause acquisition during learning. We also show that although dopamine can hyperpolarize ChIs directly, its augmentation of pauses is best explained by strengthening excitatory inputs. These findings provide a basis to understand pause generation in striatal ChIs. VIDEO ABSTRACT.


Assuntos
Neurônios Colinérgicos/metabolismo , Corpo Estriado/metabolismo , Dopamina/metabolismo , Interneurônios/metabolismo , Aprendizagem , Animais , Antracenos/farmacologia , Neurônios Colinérgicos/efeitos dos fármacos , Simulação por Computador , Corpo Estriado/citologia , Corpo Estriado/efeitos dos fármacos , Interneurônios/efeitos dos fármacos , Canal de Potássio KCNQ2/antagonistas & inibidores , Canal de Potássio KCNQ3/antagonistas & inibidores , Camundongos , Modelos Neurológicos , Bloqueadores dos Canais de Potássio/farmacologia , Ratos
6.
Epilepsia ; 58(12): 2073-2084, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29067685

RESUMO

OBJECTIVE: Kv7 channels mediate the voltage-gated M-type potassium current. Reduction of M current due to KCNQ2 mutations causes early onset epileptic encephalopathies (EOEEs). Mutations in STXBP1 encoding the syntaxin binding protein 1 can produce a phenotype similar to that of KCNQ2 mutations, suggesting a possible link between STXBP1 and Kv7 channels. These channels are known to be modulated by syntaxin-1A (Syn-1A) that binds to the C-terminal domain of the Kv7.2 subunit and strongly inhibits M current. Here, we investigated whether STXBP1could prevent this inhibitory effect of Syn-1A and analyzed the consequences of two mutations in STXBP1 associated with EOEEs. METHODS: Electrophysiologic analysis of M currents mediated by homomeric Kv7.2 or heteromeric Kv7.2/Kv7.3 channels in Chinese hamster ovary (CHO) cells coexpressing Syn-1A and/or STXBP1 or mutants STXBP1 p.W28* and p.P480L. Expression and interaction of these different proteins have been investigated using biochemical and co-immunoprecipitation experiments. RESULTS: Syn-1A decreased M currents mediated by Kv7.2 or Kv7.2/Kv7.3 channels. STXBP1 had no direct effects on M current but dampened the inhibition produced by Syn-1A by abrogating Syn-1A binding to Kv7 channels. The mutation p.W28*, but not p.P480L, failed to rescue M current from Syn-1A inhibition. Biochemical analysis showed that unlike the mutation p.W28*, the mutation p.P480L did not affect STXBP1 expression and reduced the interaction of Syn-1A with Kv7 channels. SIGNIFICANCE: These data indicate that there is a functional link between STXBP1 and Kv7 channels via Syn-1A, which may be important for regulating M-channel activity and neuronal excitability. They suggest also that a defect in Kv7 channel activity or regulation could be one of the consequences of some STXBP1 mutations associated with EOEEs. Furthermore, our data reveal that STXBP1 mutations associated with the Ohtahara syndrome do not necessarily result in protein haploinsufficiency.


Assuntos
Canal de Potássio KCNQ2/genética , Proteínas Munc18/genética , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Espasmos Infantis/genética , Sintaxina 1/farmacologia , Animais , Biotinilação , Células CHO , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Eletroencefalografia , Humanos , Canal de Potássio KCNQ1/antagonistas & inibidores , Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ3/antagonistas & inibidores , Canal de Potássio KCNQ3/genética
7.
Mol Pain ; 13: 1744806917709371, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28566000

RESUMO

Background Chronic visceral pain is a defining symptom of many gastrointestinal disorders. The KV7 family (KV7.1-KV7.5) of voltage-gated potassium channels mediates the M current that regulates excitability in peripheral sensory nociceptors and central pain pathways. Here, we use a combination of immunohistochemistry, gut-nerve electrophysiological recordings in both mouse and human tissues, and single-cell qualitative real-time polymerase chain reaction of gut-projecting sensory neurons, to investigate the contribution of peripheral KV7 channels to visceral nociception. Results Immunohistochemical staining of mouse colon revealed labelling of KV7 subtypes (KV7.3 and KV7.5) with CGRP around intrinsic enteric neurons of the myenteric plexuses and within extrinsic sensory fibres along mesenteric blood vessels. Treatment with the KV7 opener retigabine almost completely abolished visceral afferent firing evoked by the algogen bradykinin, in agreement with significant co-expression of mRNA transcripts by single-cell qualitative real-time polymerase chain reaction for KCNQ subtypes and the B2 bradykinin receptor in retrogradely labelled extrinsic sensory neurons from the colon. Retigabine also attenuated responses to mechanical stimulation of the bowel following noxious distension (0-80 mmHg) in a concentration-dependent manner, whereas the KV7 blocker XE991 potentiated such responses. In human bowel tissues, KV7.3 and KV7.5 were expressed in neuronal varicosities co-labelled with synaptophysin and CGRP, and retigabine inhibited bradykinin-induced afferent activation in afferent recordings from human colon. Conclusions We show that KV7 channels contribute to the sensitivity of visceral sensory neurons to noxious chemical and mechanical stimuli in both mouse and human gut tissues. As such, peripherally restricted KV7 openers may represent a viable therapeutic modality for the treatment of gastrointestinal pathologies.


Assuntos
Colo/metabolismo , Canais de Potássio KCNQ/metabolismo , Canal de Potássio KCNQ3/metabolismo , Receptores da Bradicinina/metabolismo , Células Receptoras Sensoriais/metabolismo , Animais , Antracenos/farmacologia , Eletrofisiologia , Humanos , Imuno-Histoquímica , Canais de Potássio KCNQ/antagonistas & inibidores , Canal de Potássio KCNQ3/antagonistas & inibidores , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plexo Mientérico/metabolismo , Sinaptofisina/metabolismo
8.
J Recept Signal Transduct Res ; 37(3): 259-266, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27607834

RESUMO

Biological mechanism attributing mutations in KCNQ2/Q3 results in benign familial neonatal epilepsy (BFNE), a rare form of epilepsy and thus neglected. It offers a potential target for antiepileptic drug discovery. In the present work, a pharmacophore-based 3D-QSAR model was generated for a series of N-pyridyl and pyrimidine benzamides possessing KCNQ2/Q3 opening activity. The pharmacophore model generated contains one hydrogen bond donor (D), one hydrophobic (H), and two aromatic rings (R). They are the crucial molecular write-up detailing predicted binding efficacy of high affinity and low affinity ligands for KCNQ2/Q3 opening activity. Furthermore, it has been validated by using a biological correlation between pharmacophore hypothesis-based 3D-QSAR variables and functional fingerprints of openers responsible for the receptor binding and also by docking of these benzamides into the validated homology model. Excellent statistical computational tools of QSAR model such as good correlation coefficient (R2 > 0.80), higher F value (F > 39), and excellent predictive power (Q2 > 0.7) with low standard deviation (SD <0.3) strongly suggest that the developed model could be used for prediction of antiepileptic activity of newer analogs. A preliminary pharmacokinetic profile of these derivatives was also performed on the basis of QikProp predictions.


Assuntos
Benzamidas/química , Descoberta de Drogas , Epilepsia Neonatal Benigna/tratamento farmacológico , Canal de Potássio KCNQ2/química , Canal de Potássio KCNQ3/química , Anticonvulsivantes/química , Anticonvulsivantes/uso terapêutico , Benzamidas/uso terapêutico , Sítios de Ligação , Simulação por Computador , Epilepsia Neonatal Benigna/genética , Epilepsia Neonatal Benigna/patologia , Humanos , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Canal de Potássio KCNQ2/antagonistas & inibidores , Canal de Potássio KCNQ2/genética , Canal de Potássio KCNQ3/antagonistas & inibidores , Canal de Potássio KCNQ3/genética , Modelos Moleculares , Simulação de Acoplamento Molecular , Mutação , Pirimidinas/química , Relação Quantitativa Estrutura-Atividade
9.
Eur J Pharmacol ; 791: 221-228, 2016 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-27590358

RESUMO

Inhibition of KCNQ (Kv7) potassium channels by activation of muscarinic acetylcholine receptors has been well established, and the ion currents through these channels have been long known as M-currents. We found that this cross-talk can be reconstituted in Xenopus oocytes by co-transfection of human recombinant muscarinic M1 receptors and KCNQ2/3 potassium channels. Application of the muscarinic acetylcholine receptor agonist Oxotremorine-methiodide (Oxo-M) between voltage pulses to activate KCNQ2/3 channels caused inhibition of the subsequent KCNQ2/3 responses. This effect of Oxo-M was blocked by the muscarinic acetylcholine receptor antagonist atropine. We also found that KCNQ2/3 currents were inhibited when Oxo-M was applied during an ongoing KCNQ2/3 response, an effect that was not blocked by atropine, suggesting that Oxo-M inhibits KCNQ2/3 channels directly. Indeed, also in oocytes that were transfected with only KCNQ2/3 channels, but not with muscarinic M1 receptors, Oxo-M inhibited the KCNQ2/3 response. These results show that besides the usual muscarinic acetylcholine receptor-mediated inhibition, Oxo-M also inhibits KCNQ2/3 channels by a direct mechanism. We subsequently tested xanomeline, which is a chemically distinct muscarinic acetylcholine receptor agonist, and oxotremorine, which is a close analogue of Oxo-M. Both compounds inhibited KCNQ2/3 currents via activation of M1 muscarinic acetylcholine receptors but, in contrast to Oxo-M, they did not directly inhibit KCNQ2/3 channels. Xanomeline and oxotremorine do not contain a positively charged trimethylammonium moiety that is present in Oxo-M, suggesting that such a charged moiety could be a crucial component mediating this newly described direct inhibition of KCNQ2/3 channels.


Assuntos
Canal de Potássio KCNQ2/antagonistas & inibidores , Canal de Potássio KCNQ3/antagonistas & inibidores , Oxotremorina/análogos & derivados , Bloqueadores dos Canais de Potássio/farmacologia , Animais , Humanos , Oxotremorina/farmacologia , Piridinas/farmacologia , Receptor Muscarínico M1/metabolismo , Tiadiazóis/farmacologia , Xenopus
10.
Int J Mol Sci ; 17(3): 407, 2016 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-26999128

RESUMO

KCNQ (KV7) channels are voltage-gated potassium (KV) channels, and the function of KV7 channels in muscles, neurons, and sensory cells is well established. We confirmed that overall blockade of KV channels with tetraethylammonium augmented the mineralization of bone-marrow-derived human mesenchymal stem cells during osteogenic differentiation, and we determined that KV7.3 was expressed in MG-63 and Saos-2 cells at the mRNA and protein levels. In addition, functional KV7 currents were detected in MG-63 cells. Inhibition of KV7.3 by linopirdine or XE991 increased the matrix mineralization during osteoblast differentiation. This was confirmed by alkaline phosphatase, osteocalcin, and osterix in MG-63 cells, whereas the expression of Runx2 showed no significant change. The extracellular glutamate secreted by osteoblasts was also measured to investigate its effect on MG-63 osteoblast differentiation. Blockade of KV7.3 promoted the release of glutamate via the phosphorylation of extracellular signal-regulated kinase 1/2-mediated upregulation of synapsin, and induced the deposition of type 1 collagen. However, activation of KV7.3 by flupirtine did not produce notable changes in matrix mineralization during osteoblast differentiation. These results suggest that KV7.3 could be a novel regulator in osteoblast differentiation.


Assuntos
Calcificação Fisiológica , Canal de Potássio KCNQ3/metabolismo , Osteoblastos/metabolismo , Osteogênese , Fosfatase Alcalina/metabolismo , Células Cultivadas , Colágeno Tipo I/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Ácido Glutâmico/metabolismo , Humanos , Canal de Potássio KCNQ3/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/citologia , Osteocalcina/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Fator de Transcrição Sp7 , Sinapsinas/metabolismo , Fatores de Transcrição/metabolismo
11.
Neuroscience ; 280: 19-30, 2014 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-25234320

RESUMO

Spatial memory retrieval and hippocampal long-term potentiation (LTP) are impaired by stress. KCNQ/Kv7 channels are closely associated with memory and the KCNQ/Kv7 channel activator flupirtine represents neuroprotective effects. This study aims to test whether KCNQ/Kv7 channel activation prevents acute stress-induced impairments of spatial memory retrieval and hippocampal LTP. Rats were placed on an elevated platform in the middle of a bright room for 30 min to evoke acute stress. The expression of KCNQ/Kv7 subunits was analyzed at 1, 3 and 12 h after stress by Western blotting. Spatial memory was examined by the Morris water maze (MWM) and the field excitatory postsynaptic potential (fEPSP) in the hippocampal CA1 area was recorded in vivo. Acute stress transiently decreased the expression of KCNQ2 and KCNQ3 in the hippocampus. Acute stress impaired the spatial memory retrieval and hippocampal LTP, the KCNQ/Kv7 channel activator flupirtine prevented the impairments, and the protective effects of flupirtine were blocked by XE-991 (10,10-bis(4-Pyridinylmethyl)-9(10H)-anthracenone), a selective KCNQ channel blocker. Furthermore, acute stress decreased the phosphorylation of glycogen synthase kinase-3ß (GSK-3ß) at Ser9 in the hippocampus, and flupirtine inhibited the reduction. These results suggest that the KCNQ/Kv7 channels may be a potential target for protecting both hippocampal synaptic plasticity and spatial memory retrieval from acute stress influences.


Assuntos
Aminopiridinas/farmacologia , Hipocampo/efeitos dos fármacos , Potenciação de Longa Duração/efeitos dos fármacos , Transtornos da Memória/prevenção & controle , Nootrópicos/farmacologia , Estresse Psicológico/tratamento farmacológico , Animais , Antracenos/farmacologia , Carbamatos/farmacologia , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Hipocampo/fisiopatologia , Canal de Potássio KCNQ2/antagonistas & inibidores , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ3/antagonistas & inibidores , Canal de Potássio KCNQ3/metabolismo , Potenciação de Longa Duração/fisiologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/fisiopatologia , Fenilenodiaminas/farmacologia , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Bloqueadores dos Canais de Potássio/farmacologia , Ratos Sprague-Dawley , Memória Espacial/efeitos dos fármacos , Memória Espacial/fisiologia , Estresse Psicológico/fisiopatologia
12.
PLoS One ; 8(9): e76085, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24086693

RESUMO

KCNQ genes encode five Kv7 K(+) channel subunits (Kv7.1-Kv7.5). Four of these (Kv7.2-Kv7.5) are expressed in the nervous system. Kv7.2 and Kv7.3 are the principal molecular components of the slow voltage-gated M-channel, which regulates neuronal excitability. In this study, we demonstrate that tamoxifen, an estrogen receptor antagonist used in the treatment of breast cancer, inhibits Kv7.2/Kv7.3 currents heterologously expressed in human embryonic kidney HEK-293 cells. Current inhibition by tamoxifen was voltage independent but concentration-dependent. The IC50 for current inhibition was 1.68 ± 0.44 µM. The voltage-dependent activation of the channel was not modified. Tamoxifen inhibited Kv7.2 homomeric channels with a higher potency (IC50 = 0.74 ± 0.16 µM). The mutation Kv7.2 R463E increases phosphatidylinositol- 4,5-bisphosphate (PIP2) - channel interaction and diminished dramatically the inhibitory effect of tamoxifen compared with that for wild type Kv7.2. Conversely, the mutation Kv7.2 R463Q, which decreases PIP2 -channel interaction, increased tamoxifen potency. Similar results were obtained on the heteromeric Kv7.2 R463Q/Kv7.3 and Kv7.2 R463E/Kv7.3 channels, compared to Kv7.2/Kv7.3 WT. Overexpression of type 2A PI(4)P5-kinase (PIP5K 2A) significantly reduced tamoxifen inhibition of Kv7.2/Kv7.3 and Kv7.2 R463Q channels. Our results suggest that tamoxifen inhibited Kv7.2/Kv7.3 channels by interfering with PIP2-channel interaction because of its documented interaction with PIP2 and the similar effect of tamoxifen on various PIP2 sensitive channels.


Assuntos
Canal de Potássio KCNQ2/antagonistas & inibidores , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ3/antagonistas & inibidores , Canal de Potássio KCNQ3/metabolismo , Tamoxifeno/farmacologia , Células HEK293 , Humanos , Concentração Inibidora 50 , Canal de Potássio KCNQ2/genética , Mutação de Sentido Incorreto/genética , Técnicas de Patch-Clamp , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo
13.
PLoS One ; 8(3): e58901, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23520542

RESUMO

The parasubiculum (PaS) is a component of the hippocampal formation that sends its major output to layer II of the entorhinal cortex. The PaS receives strong cholinergic innervation from the basal forebrain that is likely to modulate neuronal excitability and contribute to theta-frequency network activity. The present study used whole cell current- and voltage-clamp recordings to determine the effects of cholinergic receptor activation on layer II PaS neurons. Bath application of carbachol (CCh; 10-50 µM) resulted in a dose-dependent depolarization of morphologically-identified layer II stellate and pyramidal cells that was not prevented by blockade of excitatory and inhibitory synaptic inputs. Bath application of the M1 receptor antagonist pirenzepine (1 µM), but not the M2-preferring antagonist methoctramine (1 µM), blocked the depolarization, suggesting that it is dependent on M1 receptors. Voltage-clamp experiments using ramped voltage commands showed that CCh resulted in the gradual development of an inward current that was partially blocked by concurrent application of the selective Kv7.2/3 channel antagonist XE-991, which inhibits the muscarine-dependent K(+) current I M. The remaining inward current also reversed near EK and was inhibited by the K(+) channel blocker Ba(2+), suggesting that M1 receptor activation attenuates both I M as well as an additional K(+) current. The additional K(+) current showed rectification at depolarized voltages, similar to K(+) conductances mediated by Kir 2.3 channels. The cholinergic depolarization of layer II PaS neurons therefore appears to occur through M1-mediated effects on I M as well as an additional K(+) conductance.


Assuntos
Potenciais da Membrana/fisiologia , Neurônios/metabolismo , Células Piramidais/metabolismo , Receptor Muscarínico M1/metabolismo , Animais , Antracenos/farmacologia , Carbacol/farmacologia , Agonistas Colinérgicos/farmacologia , Diaminas/farmacologia , Canal de Potássio KCNQ2/antagonistas & inibidores , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ3/antagonistas & inibidores , Canal de Potássio KCNQ3/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Antagonistas Muscarínicos/farmacologia , Neurônios/citologia , Parassimpatolíticos/farmacologia , Pirenzepina/farmacologia , Células Piramidais/citologia , Ratos , Ratos Long-Evans , Receptor Muscarínico M1/agonistas , Receptor Muscarínico M1/antagonistas & inibidores
14.
Pain ; 154(3): 434-448, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23352759

RESUMO

Bone cancer pain has a strong impact on the quality of life of patients, but is difficult to treat. Better understanding of the pathogenic mechanisms underlying bone cancer pain will likely lead to the development of more effective treatments. In the present study, we investigated whether inhibition of KCNQ/M channels contributed to the hyperexcitability of primary sensory neurons and to the pathogenesis of bone cancer pain. By using a rat model of bone cancer pain based on intratibial injection of MRMT-1 tumour cells, we documented a prominent decrease in expression of KCNQ2 and KCNQ3 proteins and a reduction of M-current density in small-sized dorsal root ganglia (DRG) neurons, which were associated with enhanced excitability of these DRG neurons and the hyperalgesic behaviours in bone cancer rats. Coincidently, we found that inhibition of KCNQ/M channels with XE-991 caused a robust increase in the excitability of small-sized DRG neurons and produced an obvious mechanical allodynia in normal rats. On the contrary, activation of the KCNQ/M channels with retigabine not only inhibited the hyperexcitability of these small DRG neurons, but also alleviated mechanical allodynia and thermal hyperalgesia in bone cancer rats, and all of these effects of retigabine could be blocked by KCNQ/M-channel antagonist XE-991. These results suggest that repression of KCNQ/M channels leads to the hyperexcitability of primary sensory neurons, which in turn causes bone cancer pain. Thus, suppression of KCNQ/M channels in primary DRG neurons plays a crucial role in the development of bone cancer pain.


Assuntos
Neoplasias Ósseas/fisiopatologia , Carcinoma/fisiopatologia , Gânglios Espinais/fisiopatologia , Hiperalgesia/etiologia , Canal de Potássio KCNQ2/fisiologia , Canal de Potássio KCNQ3/fisiologia , Nociceptividade/fisiologia , Dor/etiologia , Células Receptoras Sensoriais/fisiologia , Animais , Antracenos/farmacologia , Neoplasias Ósseas/patologia , Neoplasias Ósseas/secundário , Carbamatos/farmacologia , Carbamatos/uso terapêutico , Carcinoma/patologia , Carcinoma/secundário , Regulação para Baixo , Feminino , Temperatura Alta/efeitos adversos , Hiperalgesia/fisiopatologia , Canal de Potássio KCNQ2/antagonistas & inibidores , Canal de Potássio KCNQ2/biossíntese , Canal de Potássio KCNQ2/genética , Canal de Potássio KCNQ3/antagonistas & inibidores , Canal de Potássio KCNQ3/biossíntese , Canal de Potássio KCNQ3/genética , Neoplasias Mamárias Experimentais/patologia , Transplante de Neoplasias , Técnicas de Patch-Clamp , Fenilenodiaminas/farmacologia , Fenilenodiaminas/uso terapêutico , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , Células Receptoras Sensoriais/metabolismo , Estresse Mecânico , Transmissão Sináptica , Tíbia/patologia
16.
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
17.
J Gen Physiol ; 138(3): 341-52, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21844219

RESUMO

Transient receptor potential vanilloid 1 (TRPV1) is a ligand-gated nonselective cation channel expressed predominantly in peripheral nociceptors. By detecting and integrating diverse noxious thermal and chemical stimuli, and as a result of its sensitization by inflammatory mediators, the TRPV1 receptor plays a key role in inflammation-induced pain. Activation of TRPV1 leads to a cascade of pro-nociceptive mechanisms, many of which still remain to be identified. Here, we report a novel effect of TRPV1 on the activity of the potassium channel KCNQ2/3, a negative regulator of neuronal excitability. Using ion influx assays, we revealed that TRPV1 activation can abolish KCNQ2/3 activity, but not vice versa, in human embryonic kidney (HEK)293 cells. Electrophysiological studies showed that coexpression of TRPV1 caused a 7.5-mV depolarizing shift in the voltage dependence of KCNQ2/3 activation compared with control expressing KCNQ2/3 alone. Furthermore, activation of TRPV1 by capsaicin led to a 54% reduction of KCNQ2/3-mediated current amplitude and attenuation of KCNQ2/3 activation. The inhibitory effect of TRPV1 appears to depend on Ca(2+) influx through the activated channel followed by Ca(2+)-sensitive depletion of phosphatidylinositol 4,5-bisphosphate and activation of protein phosphatase calcineurin. We also identified physical interactions between TRPV1 and KCNQ2/3 coexpressed in HEK293 cells and in rat dorsal root ganglia neurons. Mutation studies established that this interaction is mediated predominantly by the membrane-spanning regions of the respective proteins and correlates with the shift of KCNQ2/3 activation. Collectively, these data reveal that TRPV1 activation may deprive neurons from inhibitory control mediated by KCNQ2/3. Such neurons may thus have a lower threshold for activation, which may indirectly facilitate TRPV1 in integrating multiple noxious signals and/or in the establishment or maintenance of chronic pain.


Assuntos
Canal de Potássio KCNQ2/antagonistas & inibidores , Canal de Potássio KCNQ3/antagonistas & inibidores , Canais de Cátion TRPV/metabolismo , Animais , Células Cultivadas , Células HEK293 , Humanos , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ3/metabolismo , Masculino , Neurônios/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Ratos , Ratos Sprague-Dawley , Transfecção
18.
J Neurosci ; 30(40): 13235-45, 2010 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-20926649

RESUMO

Regulation of the resting membrane potential and the repolarization of neurons are important in regulating neuronal excitability. The potassium channel subunits Kv7.2 and Kv7.3 play a key role in stabilizing neuronal activity. Mutations in KCNQ2 and KCNQ3, the genes encoding Kv7.2 and Kv7.3, cause a neonatal form of epilepsy, and activators of these channels have been identified as novel antiepileptics and analgesics. Despite the observations that regulation of these subunits has profound effects on neuronal function, almost nothing is known about the mechanisms responsible for controlling appropriate expression levels. Here we identify two mechanisms responsible for regulating KCNQ2 and KCNQ3 mRNA levels. We show that the transcription factor Sp1 activates expression of both KCNQ2 and KCNQ3, whereas the transcriptional repressor REST (repressor element 1-silencing transcription factor) represses expression of both of these genes. Furthermore, we show that transcriptional regulation of KCNQ genes is mirrored by the correlated changes in M-current density and excitability of native sensory neurons. We propose that these mechanisms are important in the control of excitability of neurons and may have implications in seizure activity and pain.


Assuntos
Regulação da Expressão Gênica/fisiologia , Canal de Potássio KCNQ2/genética , Canal de Potássio KCNQ3/genética , Proteínas Repressoras/fisiologia , Células Receptoras Sensoriais/fisiologia , Fator de Transcrição Sp1/fisiologia , Ativação Transcricional/genética , Animais , Linhagem Celular , Linhagem Celular Tumoral , Doença Crônica , Epilepsia/genética , Epilepsia/fisiopatologia , Humanos , Canal de Potássio KCNQ2/antagonistas & inibidores , Canal de Potássio KCNQ2/biossíntese , Canal de Potássio KCNQ3/antagonistas & inibidores , Canal de Potássio KCNQ3/biossíntese , Inibição Neural/genética , Vias Neurais/fisiopatologia , Dor/genética , Dor/fisiopatologia , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/biossíntese , Ratos , Proteínas Repressoras/biossíntese , Proteínas Repressoras/genética , Fator de Transcrição Sp1/genética , Regulação para Cima/fisiologia
19.
J Pharmacol Exp Ther ; 332(3): 811-20, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20040580

RESUMO

Changes in the expression of potassium channels regulate skeletal muscle development. The purpose of this study was to investigate the expression profile and pharmacological role of K(v)7 voltage-gated potassium channels in skeletal muscle differentiation, proliferation, and survival after myotoxic insults. Transcripts for all K(v)7 genes (K(v)7.1-K(v)7.5) were detected by polymerase chain reaction (PCR) and/or real-time PCR in murine C(2)C(12) myoblasts; K(v)7.1, K(v)7.3, and K(v)7.4 transcripts were up-regulated after myotube formation. Western blot experiments confirmed K(v)7.2, K(v)7.3, and K(v)7.4 subunit expression, and the up-regulation of K(v)7.3 and K(v)7.4 subunits during in vitro differentiation. In adult skeletal muscles from mice and humans, K(v)7.2 and K(v)7.3 immunoreactivity was mainly localized at the level of intracellular striations positioned between ankyrinG-positive triads, whereas that of K(v)7.4 subunits was largely restricted to the sarcolemmal membrane. In C(2)C(12) cells, retigabine (10 microM), a specific activator of neuronally expressed K(v)7.2 to K(v)7.5 subunits, reduced proliferation, accelerated myogenin expression, and inhibited the myotoxic effect of mevastatin (IC(50) approximately 7 microM); all these effects of retigabine were prevented by the K(v)7 channel blocker 10,10-bis(4-pyridinylmethyl)-9(10H)-anthracenone (XE-991) (10 muM). These data collectively highlight neural K(v)7 channels as significant pharmacological targets to regulate skeletal muscle proliferation, differentiation, and myotoxic effects of drugs.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases/toxicidade , Canais de Potássio KCNQ/biossíntese , Lovastatina/análogos & derivados , Músculo Esquelético/citologia , Músculo Esquelético/efeitos dos fármacos , Adulto , Animais , Antracenos/farmacologia , Carbamatos/farmacologia , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , Cricetinae , Cricetulus , Humanos , Técnicas In Vitro , Canais de Potássio KCNQ/antagonistas & inibidores , Canais de Potássio KCNQ/genética , Canal de Potássio KCNQ1/antagonistas & inibidores , Canal de Potássio KCNQ1/biossíntese , Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ2/antagonistas & inibidores , Canal de Potássio KCNQ2/biossíntese , Canal de Potássio KCNQ2/genética , Canal de Potássio KCNQ3/antagonistas & inibidores , Canal de Potássio KCNQ3/biossíntese , Canal de Potássio KCNQ3/genética , Lovastatina/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/efeitos dos fármacos , Mioblastos Esqueléticos/metabolismo , Fenilenodiaminas/farmacologia , Subunidades Proteicas/biossíntese , RNA Mensageiro/biossíntese , Regulação para Cima
20.
Neurosci Lett ; 461(1): 25-9, 2009 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-19463897

RESUMO

A large amount of in vitro studies demonstrate suppression of M-current in hippocampal neurons by Kv7/M channel blocker results in depolarization of membrane potential and release of neurotransmitters, such as acetylcholine and glutamate, suggesting that Kv7/M channel may play important roles in regulating synaptic plasticity. In the present study, we examined the in vivo effect of Kv7/M channel inhibition on the long-term potentiation (LTP) induction at basal dendrites in hippocampal CA1 area of urethane-anaesthetized rats. The Kv7/M channel was inhibited by intraperitoneal injection of XE991 (10mg/kg) and the LTP of field excitatory postsynaptic potential (fEPSP) was induced by supra-threshold high frequency stimulation (S1 HFS). A weak protocol which was just below the threshold for evoking LTP was used as sub-threshold high frequency stimulation (S2 HFS). XE991 did not significantly alter the slope of fEPSP and the magnitude of LTP induced by S1 HFS, suggesting that Kv7/M channel inhibition had little or no effect on glutamatergic transmission under basal conditions. However, XE991 could make S2 HFS evoke LTP even after the application of the muscarinic cholinergic (mACh) receptor antagonist scopolamine, suggesting that Kv7/M channel inhibition lowered the threshold for LTP induction and the effect was independent of muscarinic activation. Based on the above findings, we concluded that the facilitating effect of XE991 on LTP induction is not mediated by its ability to enhance the release of acetylcholine; therefore, Kv7/M channel blockers may provide a therapeutic benefit to cholinergic deficiency-related cognitive impairment, e.g., Alzheimer's disease.


Assuntos
Antracenos/farmacologia , Hipocampo/efeitos dos fármacos , Canal de Potássio KCNQ2/antagonistas & inibidores , Canal de Potássio KCNQ3/antagonistas & inibidores , Potenciação de Longa Duração , Bloqueadores dos Canais de Potássio/farmacologia , Receptores Muscarínicos/fisiologia , Animais , Dendritos/fisiologia , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores , Hipocampo/fisiologia , Masculino , Antagonistas Muscarínicos/farmacologia , Ratos , Ratos Wistar , Escopolamina/farmacologia , Sinapses/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...