Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(7)2023 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-37047271

RESUMO

Oligomeric ion channels are abundant in nature. However, the recombinant expression in cell culture-based systems remains tedious and challenging due to negative side effects, limiting the understanding of their role in health and disease. Accordingly, in this work, we demonstrate the cell-free synthesis (CFS) as an alternative platform to study the assembly of two-pore domain potassium channels (K2P) within endogenous endoplasmic reticulum-derived microsomes. Exploiting the open nature of CFS, we investigate the cotranslational translocation of TREK-2 into the microsomes and suggest a cotranslational assembly with typical single-channel behavior in planar lipid-bilayer electrophysiology. The heteromeric assembly of K2P channels is a contentious matter, accordingly we prove the successful assembly of TREK-2 with TWIK-1 using a biomolecular fluorescence complementation assay, Western blot analysis and autoradiography. The results demonstrate that TREK-2 homodimer assembly is the initial step, followed by heterodimer formation with the nascent TWIK-1, providing evidence of the intergroup heterodimerization of TREK-2 and TWIK-1 in eukaryotic CFS. Since K2P channels are involved in various pathophysiological conditions, including pain and nociception, CFS paves the way for in-depth functional studies and related pharmacological interventions. This study highlights the versatility of the eukaryotic CFS platform for investigating ion channel assembly in a native-like environment.


Assuntos
Eucariotos , Canais de Potássio de Domínios Poros em Tandem , Eucariotos/metabolismo , Canais de Potássio de Domínios Poros em Tandem/genética , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Sistema Livre de Células/metabolismo , Dimerização , Bioensaio
2.
Molecules ; 28(19)2023 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-37836658

RESUMO

TWIK1 (K2P1.1/KCNK1) belongs to the potassium channels of the two-pore domain. Its current is very small and difficult to measure. In this work, we used a 100 mM NH4+ extracellular solution to increase TWIK1 current in its stable cell line expressed in HEK293. Then, the inhibition of magnolol on TWIK1 was observed via a whole-cell patch clamp experiment, and it was found that magnolol had a significant inhibitory effect on TWIK1 (IC50 = 6.21 ± 0.13 µM). By molecular docking and alanine scanning mutagenesis, the IC50 of TWIK1 mutants G229A, T225A, I140A, L223A, and S224A was 20.77 ± 3.20, 21.81 ± 7.93, 10.22 ± 1.07, 9.55 ± 1.62, and 7.43 ± 3.20 µM, respectively. Thus, we conclude that the inhibition of the TWIK1 channel by magnolol is related to G229 and T225 on the P2- pore helix.


Assuntos
Compostos de Bifenilo , Canais de Potássio , Humanos , Simulação de Acoplamento Molecular , Células HEK293 , Canais de Potássio/metabolismo , Compostos de Bifenilo/farmacologia
3.
Int J Mol Sci ; 21(24)2020 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-33348878

RESUMO

Astrocytes, the most abundant cell type in the brain, are non-excitable cells and play critical roles in brain function. Mature astrocytes typically exhibit a linear current-voltage relationship termed passive conductance, which is believed to enable astrocytes to maintain potassium homeostasis in the brain. We previously demonstrated that TWIK-1/TREK-1 heterodimeric channels mainly contribute to astrocytic passive conductance. However, the molecular identity of astrocytic passive conductance is still controversial and needs to be elucidated. Here, we report that spadin, an inhibitor of TREK-1, can dramatically reduce astrocytic passive conductance in brain slices. A series of gene silencing experiments demonstrated that spadin-sensitive currents are mediated by TWIK-1/TREK-1 heterodimeric channels in cultured astrocytes and hippocampal astrocytes from brain slices. Our study clearly showed that TWIK-1/TREK-1-heterodimeric channels can act as the main molecular machinery of astrocytic passive conductance, and suggested that spadin can be used as a specific inhibitor to control astrocytic passive conductance.


Assuntos
Astrócitos/fisiologia , Encéfalo/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Peptídeos/farmacologia , Canais de Potássio de Domínios Poros em Tandem/antagonistas & inibidores , Multimerização Proteica , Animais , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL
4.
Int J Mol Sci ; 19(1)2017 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-29295556

RESUMO

Two-pore domain K⁺ (K2P) channels play essential roles in regulating resting membrane potential and cellular excitability. Although TWIK-1 (TWIK-tandem of pore domains in a weak inward rectifying K⁺ channel) was the first identified member of the K2P channel family, it is only in recent years that the physiological roles of TWIK-1 have been studied in depth. A series of reports suggest that TWIK-1 may underlie diverse functions, such as intrinsic excitability of neurons, astrocytic passive conductance, and astrocytic glutamate release, as a homodimer or heterodimer with other K2P isotypes. Here, we summarize expression patterns and newly identified functions of TWIK-1 in the brain.


Assuntos
Encéfalo/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Multimerização Proteica , Animais , Astrócitos/metabolismo , Encefalopatias/metabolismo , Encefalopatias/patologia , Humanos , Neurônios/metabolismo
5.
J Mol Cell Cardiol ; 97: 24-35, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27103460

RESUMO

The two-pore domain potassium (K(+)) channel TWIK-1 (or K2P1.1) contributes to background K(+) conductance in diverse cell types. TWIK-1, encoded by the KCNK1 gene, is present in the human heart with robust expression in the atria, however its physiological significance is unknown. To evaluate the cardiac effects of TWIK-1 deficiency, we studied zebrafish embryos after knockdown of the two KCNK1 orthologues, kcnk1a and kcnk1b. Knockdown of kcnk1a or kcnk1b individually caused bradycardia and atrial dilation (p<0.001 vs. controls), while ventricular stroke volume was preserved. Combined knockdown of both kcnk1a and kcnk1b resulted in a more severe phenotype, which was partially reversed by co-injection of wild-type human KCNK1 mRNA, but not by a dominant negative variant of human KCNK1 mRNA. To determine whether genetic variants in KCNK1 might cause atrial fibrillation (AF), we sequenced protein-coding regions in two independent cohorts of patients (373 subjects) and identified three non-synonymous variants, p.R171H, p.I198M and p.G236S, that were all located in highly conserved amino acid residues. In transfected mammalian cells, zebrafish and wild-type human TWIK-1 channels had a similar cellular distribution with predominant localization in the endosomal compartment. Two-electrode voltage-clamp experiments using Xenopus oocytes showed that both zebrafish and wild-type human TWIK-1 channels produced K(+) currents that are sensitive to external K(+) concentration as well as acidic pH. There were no effects of the three KCNK1 variants on cellular localization, current amplitude or reversal potential at pH7.4 or pH6. Our data indicate that TWIK-1 has a highly conserved role in cardiac function and is required for normal heart rate and atrial morphology. Despite the functional importance of TWIK-1 in the atrium, genetic variation in KCNK1 is not a common primary cause of human AF.


Assuntos
Remodelamento Atrial/genética , Estudos de Associação Genética , Átrios do Coração/metabolismo , Frequência Cardíaca/genética , Canais de Potássio de Domínios Poros em Tandem/genética , Adulto , Idoso , Animais , Fibrilação Atrial/genética , Fibrilação Atrial/metabolismo , Fibrilação Atrial/patologia , Fibrilação Atrial/fisiopatologia , Feminino , Expressão Gênica , Técnicas de Inativação de Genes , Variação Genética , Átrios do Coração/anatomia & histologia , Átrios do Coração/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Linhagem , Canais de Potássio de Domínios Poros em Tandem/deficiência , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Transporte Proteico , Fatores de Risco , Peixe-Zebra
6.
Mol Cell Neurosci ; 57: 1-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23994814

RESUMO

Neuropathic pain is associated with hyperexcitability of DRG neurons. Despite the importance of leakage potassium channels for neuronal excitability, little is known about their cell-specific expression in DRGs and possible modulation in neuropathic pain. Multiple leakage channels are expressed in DRG neurons, including TASK1, TASK3, TRESK, TRAAK, TWIK1, TREK1 and TREK2 but little is known about their distribution among different cell types. Our immunohistochemical studies show robust TWIK1 expression in large and medium size neurons, without overlap with TRPV1 or IB4 staining. TASK1 and TASK3, on the contrary, are selectively expressed in small cells; TASK1 expression closely overlaps TRPV1-positive cells, while TASK3 is expressed in TRPV1- and IB4-negative cells. We also studied mRNA expression of these channels in L4-L5 DRGs in control conditions and up to 4 weeks after spared nerve injury lesion. We found that TWIK1 expression is much higher than TASK1 and TASK3 and is strongly decreased 1, 2 and 4 weeks after neuropathic injury. TASK3 expression, on the other hand, decreases 1 week after surgery but reverts to baseline by 2weeks; TASK1 shows no significant change at any time point. These data suggest an involvement of TWIK1 in the maintenance of the pain condition.


Assuntos
Regulação para Baixo , Gânglios Espinais/metabolismo , Neuralgia/metabolismo , Neurônios/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Animais , Gânglios Espinais/citologia , Masculino , Neurônios/classificação , Especificidade de Órgãos , Canais de Potássio de Domínios Poros em Tandem/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
7.
Methods Mol Biol ; 2762: 293-308, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38315373

RESUMO

Although membrane proteins are abundant in nature, their investigation is limited due to bottlenecks in heterologous overexpression and consequently restricted accessibility for downstream applications. In this chapter, we address these challenges by presenting a fast and straightforward synthesis platform based on eukaryotic cell-free protein synthesis (CFPS) and an efficient solubilization strategy using styrene-maleic acid (SMA) copolymers. We demonstrate CFPS of TWIK-1, a dimeric ion channel, based on Sf21 (Spodoptera frugiperda) insect lysate showing homooligomerization and N-glycosylation enabled by endoplasmic reticulum-derived microsomes. Furthermore, we employ SMA copolymers for protein solubilization, which preserves the native-like microsomal environment. This approach not only retains the solubilized protein's suitability for downstream applications but also maintains the oligomerization and glycosylation of TWIK-1 post-solubilization. We validate the solubilization procedure using autoradiography, particle size analysis, and biomolecular fluorescence assay and confirm the very efficient, structurally intact solubilization of cell-free synthesized TWIK-1.


Assuntos
Maleatos , Poliestirenos , Proteínas de Membrana
8.
Front Biosci (Landmark Ed) ; 28(3): 51, 2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-37005754

RESUMO

BACKGROOUND: At low extracellular potassium ([K+]e) conditions, human cardiomyocytes can depolarize to -40 mV. This is closely related to hypokalemia-induced fatal cardiac arrhythmia. The underlying mechanism, however, is still not well understood. TWIK-1 channels are background K+ channels that are highly expressed in human cardiomyocytes. We previously reported that TWIK-1 channels changed ion selectivity and conducted leak Na+ currents at low [K+]e. Moreover, a specific threonine residue (Thr118) within the ion selectivity filter was responsible for this altered ion selectivity. METHODS: Patch clamp were used to investigate the effects of TWIK-1 channels on the membrane potentials of cardiomyocytes in response to low [K+]e. RESULTS: At 2.7 mM [K+]e and 1 mM [K+]e, both Chinese hamster ovary (CHO) cells and HL-1 cells ectopically expressed human TWIK-1 channels displayed inward leak Na+ currents and reconstitute depolarization of membrane potential. In contrast, cells ectopically expressed human TWIK-1-T118I mutant channels that remain high selectivity to K+ exhibited hyperpolarization of membrane potential. Furthermore, human iPSC-derived cardiomyocytes showed depolarization of membrane potential in response to 1 mM [K+]e, while the knockdown of TWIK-1 expression eliminated this phenomenon. CONCLUSIONS: These results demonstrate that leak Na+ currents conducted by TWIK-1 channels contribute to the depolarization of membrane potential induced by low [K+]e in human cardiomyocytes.


Assuntos
Canais de Potássio de Domínios Poros em Tandem , Cricetinae , Animais , Humanos , Células CHO , Canais de Potássio de Domínios Poros em Tandem/genética , Canais de Potássio de Domínios Poros em Tandem/química , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Cricetulus , Miócitos Cardíacos/metabolismo , Potássio/metabolismo
9.
Biomed Pharmacother ; 165: 115139, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37454597

RESUMO

TREK-1 (TWIK-related potassium channel-1) is a subunit of the two-pore domain potassium (K2p) channel and is widely expressed in the brain. TREK-1 knockout mice were shown to have antidepressant-like effects, providing evidence for the channel's potential as a therapeutic target. However, currently there is no good pharmacological inhibitor specifically targeting TREK-1 containing K2p channels that also displays similar antidepressant-like effects. Here, we sought to find selective and potent inhibitors for TREK-1 related dimers both in vitro and in vivo. We synthesized and evaluated 2-hydroxy-3-phenoxypropyl piperidine derivatives yielding a library from which many TREK-1 targeting candidates emerged. Among these, hydroxyl-phenyl- (2a), piperidino- (2g), and pyrrolidino- (2h) piperidinyl substituted compounds showed high potencies to TREK-1 homodimers with significant antidepressant-like effects in forced swim test and tail suspension test. Interestingly, these compounds were found to have high potencies to TWIK-1/TREK-1 heterodimers. Contrastingly, difluoropiperidinyl-4-fluorophenoxy (3e) and 4-hydroxyphenyl-piperidinyl-4-fluorophenoxy (3j) compounds had high potencies to TREK-1 homodimer but lower potency to TWIK-1/TREK-1 heterodimers without significant antidepressant-like effects. We observed positive correlation between inhibition potency to TWIK-1/TREK-1 and immobility time, and no correlation between inhibition potency to TREK-1 homodimer and immobility time. This was consistent with molecular docking simulations of selected compounds to TREK-1 homodimeric and TWIK-1/TREK-1 heterodimeric models. Existing antidepressant fluoxetine was also found to potently inhibit TWIK-1/TREK-1 heterodimers. Our study reveals novel potent TWIK-1/TREK-1 inhibitors 2a, 2g, and 2h as potential antidepressants and suggest that the TWIK-1/TREK-1 heterodimer could be a potential novel molecular therapeutic target for antidepressants.


Assuntos
Canais de Potássio de Domínios Poros em Tandem , Camundongos , Animais , Simulação de Acoplamento Molecular , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Encéfalo/metabolismo , Antidepressivos/farmacologia , Camundongos Knockout
10.
Cells ; 11(20)2022 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-36291187

RESUMO

Mature astrocytes are characterized by a K+ conductance (passive conductance) that changes with a constant slope with voltage, which is involved in K+ homeostasis in the brain. Recently, we reported that the tandem of pore domains in a weak inward rectifying K+ channel (TWIK1 or KCNK1) and TWIK-related K+ channel 1 (TREK1 or KCNK2) form heterodimeric channels that mediate passive conductance in astrocytes. However, little is known about the binding proteins that regulate the function of the TWIK1/TREK1 heterodimeric channels. Here, we found that ß-coat protein (COP) regulated the surface expression and activity of the TWIK1/TREK1 heterodimeric channels in astrocytes. ß-COP binds directly to TREK1 but not TWIK1 in a heterologous expression system. However, ß-COP also interacts with the TWIK1/TREK1 heterodimeric channel in a TREK1 dependent manner and enhances the surface expression of the heterodimeric channel in astrocytes. Consequently, it regulates TWIK1/TREK1 heterodimeric channel-mediated passive conductance in astrocytes in the mouse brain. Taken together, these results suggest that ß-COP is a potential regulator of astrocytic passive conductance in the brain.


Assuntos
Astrócitos , Canais de Potássio de Domínios Poros em Tandem , Animais , Camundongos , Astrócitos/metabolismo , Encéfalo/metabolismo , Membrana Celular/metabolismo , Proteína Coatomer/metabolismo
11.
Brain Sci ; 11(1)2021 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-33440655

RESUMO

AEG-1, also called MTDH, has oncogenic potential in numerous cancers and is considered a multifunctional modulator because of its involvement in developmental processes and inflammatory and degenerative brain diseases. However, the role of AEG-1 in astrocytes remains unknown. This study aimed to investigate proteins directly regulated by AEG-1 by analyzing their RNA expression patterns in astrocytes transfected with scramble shRNA and AEG-1 shRNA. AEG-1 knockdown down-regulated TWIK-1 mRNA. Real-time quantitative PCR (qPCR) and immunocytochemistry assays confirmed that AEG-1 modulates TWIK-1 mRNA and protein expression. Electrophysiological experiments further revealed that AEG-1 further regulates TWIK-1-mediated potassium currents in normal astrocytes. An RNA immunoprecipitation assay to determine how AEG-1 regulates the expression of TWIK-1 revealed that AEG-1 binds directly to TWIK-1 mRNA. Furthermore, TWIK-1 mRNA stability was significantly increased upon overexpression of AEG-1 in cultured astrocytes (p < 0.01). Our findings show that AEG-1 serves as an RNA-binding protein to regulate TWIK-1 expression in normal astrocytes.

12.
Cells ; 10(10)2021 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-34685731

RESUMO

TWIK-1 is the first identified member of the two-pore domain potassium (K2P) channels that are involved in neuronal excitability and astrocytic passive conductance in the brain. Despite the physiological roles of TWIK-1, there is still a lack of information on the basic expression patterns of TWIK-1 proteins in the brain. Here, using a modified bacterial artificial chromosome (BAC), we generated a transgenic mouse (Tg mouse) line expressing green fluorescent protein (GFP) under the control of the TWIK-1 promoter (TWIK-1 BAC-GFP Tg mice). We confirmed that nearly all GFP-producing cells co-expressed endogenous TWIK-1 in the brain of TWIK-1 BAC-GFP Tg mice. GFP signals were highly expressed in various brain areas, including the dentate gyrus (DG), lateral entorhinal cortex (LEC), and cerebellum (Cb). In addition, we found that GFP signals were highly expressed in immature granule cells in the DG. Finally, our TWIK-1 BAC-GFP Tg mice mimic the upregulation of TWIK-1 mRNA expression in the hippocampus following the injection of kainic acid (KA). Our data clearly showed that TWIK-1 BAC-GFP Tg mice are a useful animal model for studying the mechanisms regulating TWIK-1 gene expression and the physiological roles of TWIK-1 channels in the brain.


Assuntos
Cromossomos Artificiais Bacterianos/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Canais de Potássio de Domínios Poros em Tandem/genética , Animais , Cerebelo/metabolismo , Giro Denteado/metabolismo , Córtex Entorrinal/metabolismo , Ácido Caínico , Camundongos Transgênicos , Modelos Animais , Neuroglia/metabolismo , Neurônios/metabolismo , Reprodutibilidade dos Testes , Regulação para Cima
13.
Mol Neurobiol ; 53(9): 6169-6182, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-26553349

RESUMO

TWIK-1 two-pore domain K+ channels are highly expressed in mature hippocampal astrocytes. While the TWIK-1 activity is readily detectable on astrocyte membrane, the majority of channels are retained in the intracellular compartments, which raises an intriguing question of whether the membrane TWIK-1 channels could be dynamically regulated for functions yet unknown. Here, the regulation of TWIK-1 membrane expression by Gi/Go-coupled metabotropic glutamate receptor 3 (mGluR3) and its functional impact on ammonium uptake has been studied. Activation of mGluR3 induced a marked translocation of TWIK-1 channels from the cytoplasm to the membrane surface. Consistent with our early observation that membrane TWIK-1 behaves as nonselective monovalent cation channel, mGluR3-mediated TWIK-1 membrane expression was associated with a depolarizing membrane potential (V M). As TWIK-1 exhibits a discernibly high permeability to ammonium (NH4+), a critical substrate in glutamate-glutamine cycle for neurotransmitter replenishment, regulation of NH4+ uptake capacity by TWIK-1 membrane expression was determined by response of astrocyte V M to bath application of 5 mM NH4Cl. Stimulation of mGluR3 potentiated NH4+-induced V M depolarization by ∼30 % in wild type, but not in TWIK-1 knockout astrocytes. Furthermore, activation of mGluR3 mediated a coordinated translocation of TWIK-1 channels with recycling endosomes toward astrocyte membrane and the mGluR3-mediated potentiation of NH4+ uptake required a functional Rab-mediated trafficking pathway. Altogether, we demonstrate that the activation of mGluR3 up-regulates the membrane expression of TWIK-1 that in turn enhances NH4+ uptake in astrocytes, a mechanism potentially important for functional coupling of astrocyte glutamate-glutamine cycle with the replenishment of neurotransmitters in neurons.


Assuntos
Compostos de Amônio/metabolismo , Astrócitos/metabolismo , Membrana Celular/metabolismo , Citoplasma/metabolismo , Hipocampo/citologia , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Animais , Endocitose , Endossomos/metabolismo , Exocitose , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Glutamato Metabotrópico/genética , ATPase Trocadora de Sódio-Potássio/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo
14.
Adv Protein Chem Struct Biol ; 103: 263-94, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26920692

RESUMO

Based on their intimate spatial association with synapses and the capillary, astrocytes are critically involved in the control of ion, transmitter, and energy homeostasis as well as regulation of the cerebral blood flow. Under pathophysiological conditions, dysfunctional astrocytes can no longer assure homeostatic control although the underlying mechanisms are poorly understood. Specifically, neurological diseases are often accompanied by acidification of the extracellular space, but the properties of astrocytes in such an acidic environment are still a matter of debate. To meet the homeostatic requirements, astrocytes are equipped with intercellular gap junctions, inwardly rectifying K(+) (Kir) channels, and two-pore domain K(+) (K2P) channels. One goal of the present study was to overview current knowledge about astrocyte K(+) channel function during acidosis. In addition, we combined functional and molecular analyses to clarify how low pH affects K(+) channel function in astrocytes freshly isolated from the developing mouse hippocampus. Extracellular acidification led to a decrease of K(+) currents in astrocytes, probably due to modulation of Kir4.1 channels. After blocking Kir4.1 channels, low pH enhanced K(+) current amplitudes. This current activation was mimicked by modulators of TREK-1 channels, which belong to the K2P channels family. We found no evidence for the presence of acid-sensitive ion channels and transient receptor potential vanilloid receptors in hippocampal astrocytes. In conclusion, the assembly of astrocytic K(+) channels allows tolerating short, transient acidification, and glial Kir4.1 and K2P channels can be considered promising new targets in brain diseases accompanied by pH shifts.


Assuntos
Astrócitos/metabolismo , Circulação Cerebrovascular/genética , Hipocampo/metabolismo , Canais de Potássio/metabolismo , Animais , Astrócitos/patologia , Metabolismo Energético/genética , Hipocampo/patologia , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Canais de Potássio/química
15.
Channels (Austin) ; 9(1): 44-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25487004

RESUMO

Several recent ion channel structures have revealed large side portals, or 'fenestrations' at the interface between their transmembrane helices that potentially expose the ion conduction pathway to the lipid core of the bilayer. In a recent study we demonstrated that functional activity of the TWIK-1 K2P channel is influenced by the presence of hydrophobic residues deep within the inner pore. These residues are located near the fenestrations in the TWIK-1 structure and promote dewetting of the pore by forming a hydrophobic barrier to ion conduction. During our previous MD simulations, lipid tails were observed to enter these fenestrations. In this addendum to that study, we investigate lipid contribution to the dewetting process. Our results demonstrate that lipid tails from both the upper and lower leaflets can occupy the fenestrations and partially penetrate into the pore. The lipid tails do not sterically occlude the pore, but there is an inverse correlation between the presence of water within the hydrophobic barrier and the number of lipids tails within the lining of the pore. However, dewetting still occurs in the absence of lipids tails, and pore hydration appears to be determined primarily by those side-chains lining the narrowest part of the pore cavity.


Assuntos
Lipídeos/química , Canais de Potássio de Domínios Poros em Tandem/química , Interações Hidrofóbicas e Hidrofílicas , Simulação de Dinâmica Molecular , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Conformação Proteica
16.
Front Cell Neurosci ; 7: 246, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24368895

RESUMO

TWIK-1 two-pore domain K(+) channels are expressed abundantly in astrocytes. In the present study, we examined the extent to which TWIK-1 contributes to the linear current-voltage (I-V) relationship (passive) K(+) membrane conductance, a dominant electrophysiological feature of mature hippocampal astrocytes. Astrocytes from TWIK-1 knockout mice have a more negative resting potential than those from wild type animals and a reduction in both inward rectification and Cs(+) permeability. Nevertheless, the overall whole-cell passive conductance is not altered significantly in TWIK-1 knockout astrocytes. The expression of Kir4.1 and TREK-1, two other major astrocytic K(+) channels, or of other two-pore K(+) channels is not altered in TWIK-1 knockout mice, suggesting that the mild effect of TWIK-1 knockout does not result from compensation by these channels. Fractionation experiments showed that TWIK-1 is primarily localized in intracellular cytoplasmic fractions (55%) and mildly hydrophobic internal compartment fractions (41%), with only 5% in fractions containing plasma membranes. Our study revealed that TWIK-1 proteins are mainly located in the intracellular compartments of hippocampal astrocyte under physiological condition, therefore a minimal contribution of TWIK-1 channels to whole-cell currents is likely attributable to a relatively low level presence of channels in the plasma membrane.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA