Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biophys J ; 2024 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-38678368

RESUMO

Ion channels of the cyclic nucleotide-binding domain (CNBD) family play a crucial role in the regulation of key biological processes, such as photoreception and pacemaking activity in the heart. These channels exhibit high sequence and structural similarity but differ greatly in their functional responses to membrane potential. The CNBD family includes hyperpolarization-activated ion channels and depolarization-activated ether-à-go-go channels. Structural and functional studies show that the differences in the coupling interface between these two subfamilies' voltage-sensing domain and pore domain may underlie their differential response to membrane polarity. However, other structural components may also contribute to defining the polarity differences in activation. Here, we focus on the role of the C-terminal domain, which interacts with elements in both the pore and voltage-sensing domains. By generating a series of chimeras involving the C-terminal domain derived from distant members of the CNBD family, we find that the nature of the C-termini profoundly influences the gating polarity of these ion channels. Scanning mutagenesis of the C-linker region, a helix-turn-helix motif connecting the pore helix to the CNBD, reveals that residues at the intersubunit interface between the C-linkers are crucial for hyperpolarization-dependent activation. These findings highlight the unique and unexpected role of the intersubunit interface of the C-linker region in regulating the gating polarity of voltage-gated ion channels.

2.
Acta Pharmacol Sin ; 41(2): 163-172, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31399700

RESUMO

Cocaine is one of the most abused illicit drugs worldwide. It is well known that the dopamine (DA) transporter is its major target; but cocaine also acts on other targets including nicotinic acetylcholine receptors (nAChRs). In this study, we investigated the effects of cocaine on a special subtype of neuronal nAChR, α3ß4-nAChR expressed in native SH-SY5Y cells. α3ß4-nAChR-mediated currents were recorded using whole-cell recordings. Drugs were applied using a computer-controlled U-tube drug perfusion system. We showed that bath application of nicotine induced inward currents in a concentration-dependent manner with an EC50 value of 20 µM. Pre-treatment with cocaine concentration-dependently inhibited nicotine-induced current with an IC50 of 1.5 µM. Kinetic analysis showed that cocaine accelerated α3ß4-nAChR desensitization, which caused a reduction of the amplitude of nicotine-induced currents. Co-application of nicotine and cocaine (1.5 µM) depressed the maximum response on the nicotine concentration-response curve without changing the EC50 value, suggesting a non-competitive mechanism. The cocaine-induced inhibition of nicotine response exhibited both voltage- and use-dependence, suggesting an open-channel blocking mechanism. Furthermore, intracellular application of GDP-ßS (via recording electrode) did not affect cocaine-induced inhibition, suggesting that cocaine did not alter receptor internalization. Moreover, intracellular application of cocaine (30 µM) failed to alter the nicotine response. Finally, cocaine (1.5 µM) was unable to inhibit the nicotine-induced inward current in heterologous expressed α6/α3ß2ß3-nAChRs and α4ß2-nAChRs expressed in human SH-EP1 cells. Collectively, our results suggest that cocaine is a potent blocker for native α3ß4-nAChRs expressed in SH-SY5Y cells.


Assuntos
Cocaína/farmacologia , Neurônios/efeitos dos fármacos , Receptores Nicotínicos/efeitos dos fármacos , Linhagem Celular Tumoral , Cocaína/administração & dosagem , Relação Dose-Resposta a Droga , Humanos , Concentração Inibidora 50 , Neuroblastoma/metabolismo , Neurônios/metabolismo , Nicotina/farmacologia , Técnicas de Patch-Clamp , Receptores Nicotínicos/metabolismo
3.
Cell Physiol Biochem ; 47(6): 2613-2625, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29996119

RESUMO

BACKGROUND/AIMS: Alpha-synuclein (α-Syn) is a neuronal protein that is highly implicated in Parkinson's disease (PD), and protein phosphatase 2A (PP2A) is an important serine/threonine phosphatase that is associated with neurodegenerative diseases, such as PD. α-Syn can directly upregulate PP2A activity, but the underling mechanism remains unclear. Therefore, we investigated the molecular mechanism of α-Syn regulating PP2A activity. METHODS: α-Syn and its truncations were expressed in E.coli, and purified by affinity chromatography. PP2A Cα and its mutants were expressed in recombinant baculovirus, and purified by affinity chromatography combined with gel filtration chromatography. The interaction between α-Syn and PP2A Cα was detected by GST pull-down assay. PP2A activity was investigated by the colorimetric assay. RESULTS: The hydrophobic non-amyloid component (NAC) domain of α-Syn interacted with PP2A Cα and upregulated its activity. α-Syn aggregates reduced its ability to upregulate PP2A activity, since the hydrophobic domain of α-Syn was blocked during aggregation. Furthermore, in the hydrophobic center of PP2A Cα, the residue of I123 was responsible for PP2A to interact with α-Syn, and its hydrophilic mutation blocked its interaction with α-Syn as well as its activity upregulation by α-Syn. CONCLUSIONS: α-Syn bound to PP2A Cα by the hydrophobic interaction and upregulated its activity. Blocking the hydrophobic domain of α-Syn or hydrophilic mutation on the residue I123 in PP2A Cα all reduced PP2A activity upregulation by α-Syn. Overall, we explored the mechanism of α-Syn regulating PP2A activity, which might offer much insight into the basis underlying PD pathogenesis.


Assuntos
Mutação de Sentido Incorreto , Proteína Fosfatase 2C , Regulação para Cima , alfa-Sinucleína , Substituição de Aminoácidos , Linhagem Celular Tumoral , Humanos , Ligação Proteica , Domínios Proteicos , Proteína Fosfatase 2C/química , Proteína Fosfatase 2C/genética , Proteína Fosfatase 2C/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
4.
Acta Pharmacol Sin ; 39(10): 1571-1581, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29795357

RESUMO

Neuronal nicotinic acetylcholine receptors containing α6 subunits (α6*-nAChRs) show highly restricted distribution in midbrain neurons associated with pleasure, reward, and mood control, suggesting an important impact of α6*-nAChRs in modulating mesolimbic functions. However, the function and pharmacology of α6*-nAChRs remain poorly understood because of the lack of selective agonists for α6*-nAChRs and the challenging heterologous expression of functional α6*-nAChRs in mammalian cell lines. In particular, the α6 subunit is commonly co-expressed with α4*-nAChRs in the midbrain, which masks α6*-nAChR (without α4) function and pharmacology. In this study, we systematically profiled the pharmacology and function of α6*-nAChRs and compared these properties with those of α4ß2 nAChRs expressed in the same cell line. Heterologously expressed human α6/α3 chimeric subunits (α6 N-terminal domain joined with α3 trans-membrane domains and intracellular loops) with ß2 and ß3 subunits in the human SH-EP1 cell line (α6*-nAChRs) were used. Patch-clamp whole-cell recordings were performed to measure these receptor-mediated currents. Functionally, the heterologously expressed α6*-nAChRs exhibited excellent function and showed distinct nicotine-induced current responses, such as kinetics, inward rectification and recovery from desensitization, compared with α4ß2-nAChRs. Pharmacologically, α6*-nAChR was highly sensitive to the α6 subunit-selective antagonist α-conotoxin MII but had lower sensitivity to mecamylamine and dihydro-ß-erythroidine. Nicotine and acetylcholine were found to be full agonists for α6*-nAChRs, whereas epibatidine and cytisine were determined to be partial agonists. Heterologously expressed α6*-nAChRs exhibited pharmacology and function distinct from those of α4ß2-nAChRs, suggesting that α6*-nAChRs may mediate different cholinergic signals. Our α6*-nAChR expression system can be used as an excellent cell model for future investigations of α6*-nAChR function and pharmacology.


Assuntos
Agonistas Nicotínicos/farmacologia , Antagonistas Nicotínicos/farmacologia , Receptores Nicotínicos/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Humanos , Cinética , Técnicas de Patch-Clamp/métodos , Receptores Nicotínicos/química
5.
J Biol Chem ; 290(9): 5241-55, 2015 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-25555911

RESUMO

Plasminogen activator inhibitor 1 (PAI-1) level is extremely elevated in the edematous fluid of acutely injured lungs and pleurae. Elevated PAI-1 specifically inactivates pulmonary urokinase-type (uPA) and tissue-type plasminogen activators (tPA). We hypothesized that plasminogen activation and fibrinolysis may alter epithelial sodium channel (ENaC) activity, a key player in clearing edematous fluid. Two-chain urokinase (tcuPA) has been found to strongly stimulate heterologous human αßγ ENaC activity in a dose- and time-dependent manner. This activity of tcuPA was completely ablated by PAI-1. Furthermore, a mutation (S195A) of the active site of the enzyme also prevented ENaC activation. By comparison, three truncation mutants of the amino-terminal fragment of tcuPA still activated ENaC. uPA enzymatic activity was positively correlated with ENaC current amplitude prior to reaching the maximal level. In sharp contrast to uPA, neither single-chain tPA nor derivatives, including two-chain tPA and tenecteplase, affected ENaC activity. Furthermore, γ but not α subunit of ENaC was proteolytically cleaved at ((177)GR↓KR(180)) by tcuPA. In summary, the underlying mechanisms of urokinase-mediated activation of ENaC include release of self-inhibition, proteolysis of γ ENaC, incremental increase in opening rate, and activation of closed (electrically "silent") channels. This study for the first time demonstrates multifaceted mechanisms for uPA-mediated up-regulation of ENaC, which form the cellular and molecular rationale for the beneficial effects of urokinase in mitigating mortal pulmonary edema and pleural effusions.


Assuntos
Domínio Catalítico , Canais Epiteliais de Sódio/química , Estrutura Terciária de Proteína , Ativador de Plasminogênio Tipo Uroquinase/química , Animais , Sítios de Ligação/genética , Western Blotting , Canais Epiteliais de Sódio/genética , Canais Epiteliais de Sódio/metabolismo , Feminino , Humanos , Ativação do Canal Iônico/genética , Ativação do Canal Iônico/fisiologia , Cinética , Modelos Moleculares , Mutação , Oócitos/metabolismo , Oócitos/fisiologia , Técnicas de Patch-Clamp , Inibidor 1 de Ativador de Plasminogênio/química , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Ligação Proteica , Proteólise , Sódio/metabolismo , Regulação para Cima , Ativador de Plasminogênio Tipo Uroquinase/genética , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Xenopus laevis
6.
Biochem Biophys Res Commun ; 473(4): 795-800, 2016 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-27049309

RESUMO

Avermectins are a group of compounds isolated from a soil-dwelling bacterium. They have been widely used as parasiticides and insecticides, acting by relatively irreversible activation of invertebrate chloride channels. Emamectin is a soluble derivative of an avermectin. It is an insecticide, which persistently activates glutamate-gated chloride channels. However, its effects on mammalian ligand-gated ion channels are unknown. To this end, we tested the effect of emamectin on two cation selective nicotinic receptors and two GABA-gated chloride channels expressed in Xenopus oocytes using two-electrode voltage clamp. Our results demonstrate that emamectin could directly activate α7 nAChR, α4ß2 nAChR, α1ß2γ2 GABAA receptor and ρ1 GABAC receptor concentration dependently, with similar potencies for each channel. However, the potencies for it to activate these channels were at least two orders of magnitude lower than its potency of activating invertebrate glutamate-gated chloride channel. In contrast, ivermectin only activated the α1ß2γ2 GABAA receptor.


Assuntos
Dissacarídeos/farmacologia , Ativação do Canal Iônico/fisiologia , Ivermectina/análogos & derivados , Receptores de GABA/efeitos dos fármacos , Receptores Nicotínicos/efeitos dos fármacos , Regulação Alostérica/efeitos dos fármacos , Regulação Alostérica/fisiologia , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Inseticidas/farmacologia , Ivermectina/farmacologia , Agonistas Nicotínicos , Oócitos/efeitos dos fármacos , Oócitos/fisiologia , Receptores de GABA/metabolismo , Receptores de GABA-A , Receptores Nicotínicos/metabolismo , Xenopus laevis
7.
J Neurooncol ; 130(1): 43-52, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27422127

RESUMO

The growth factor receptor/PI3K/AKT pathway is an important drug target in many cancers including Glioblastoma. AKT, a key node in the pathway, has 3 isoforms, AKT1, AKT2 and AKT3. Here we investigate their role in GBM. We find each activated, ser473 phosphorylated isoform is present in some GBMs but expression patterns vary. There is a direct relationship between human GBM patient outcome and both AKT1 and AKT2 mRNA levels, but an inverse relationship with AKT3 mRNA. Furthermore, AKT3 mRNA levels were high in a less aggressive GBM subtype. Overexpressing AKT3 improves survival in a rodent model of GBM and decreases colony forming efficiency, but not growth rate, in glioma cells. Silencing AKT3 slows cell cycle progression in one cell line and increases apoptosis in another. Our studies of AKT3 substrates indicate (1) silencing both AKT2 and AKT3 reduces GSK3 phosphorylation (2) only AKT2 silencing reduces S6 phosphorylation. Since S6 phosphorylation is a marker of mTORC1 activity this indicates that AKT2 activates mTORC1, but AKT3 does not. Our results indicate AKT isoforms have different roles and downstream substrates in GBM. Unexpectedly, they indicate AKT3 delays tumor progression. Therefore strategies that inhibit AKT3 may be unhelpful in some GBM patients.


Assuntos
Glioblastoma/enzimologia , Glioblastoma/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ciclo Celular/genética , Linhagem Celular Tumoral , Sobrevivência Celular , Feminino , Regulação Neoplásica da Expressão Gênica/fisiologia , Glioblastoma/patologia , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Estimativa de Kaplan-Meier , Masculino , Fosforilação , Isoformas de Proteínas/genética , Proteínas Proto-Oncogênicas c-akt/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Serina/metabolismo , Transdução de Sinais/genética , Fatores de Tempo , Transfecção
8.
Acta Pharmacol Sin ; 37(8): 1020-30, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27374488

RESUMO

AIM: The continuous presence of an agonist drives its receptor into a refractory state, termed desensitization. In this study, we tested the hypothesis that a competitive antagonist, SR95531, could facilitate the recovery of α1ß2γ2 GABAA receptor from functional desensitization. METHODS: α1ß2γ2 GABAA receptors were expressed in Xenopus oocytes. GABA-evoked currents were recorded using two-electrode voltage-clamp technique. Drugs were applied through perfusion. RESULTS: Long application of GABA (100 µmol/L) evoked a large peak current followed by a small amplitude steady-state current (desensitization). Co-application of SR95531 during the desensitization caused a larger rebound of GABA current after removal of SR95531. Furthermore, application of SR95531 after removal of GABA increased the rate of receptor recovery from desensitization, and the recovery time constant was decreased from 59±3.2 s to 33±1.6 s. SR95531-facilitated receptor recovery from desensitization was dependent on the perfusion duration of SR95531. It was also dependent on the concentration of SR95531, and the curve fitting with Hill equation revealed two potency components, which were similar to the two potency components in inhibition of the steady-state current by SR95531. Bicuculline caused similar facilitation of desensitization recovery. CONCLUSION: SR95531 facilitates α1ß2γ2 GABAA receptor recovery from desensitization, possibly through two mechanisms: binding to the desensitized receptor and converting it to the non-desensitized state, and binding to the resting state receptor and preventing re-desensitization.


Assuntos
Antagonistas de Receptores de GABA-A/farmacologia , Oócitos/metabolismo , Piridazinas/farmacologia , Receptores de GABA-A/metabolismo , Animais , Bicuculina/farmacologia , Células Cultivadas , Interações Medicamentosas , Agonistas de Receptores de GABA-A/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Oócitos/efeitos dos fármacos , Receptores de GABA-A/genética , Recuperação de Função Fisiológica/efeitos dos fármacos , Xenopus laevis , Ácido gama-Aminobutírico/farmacologia
9.
BMC Genomics ; 16: 269, 2015 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-25887537

RESUMO

BACKGROUND: The study of olfaction is key to understanding the interaction of insects with their environment and provides opportunities to develop novel tactics for control of pest species. Recent developments in transcriptomic approaches enable the molecular basis of olfaction to be studied even in species with limited genomic information. Here we use transcriptome and expression profiling analysis to characterize the antennal transcriptome of the noctuid moth and polyphagous pest Spodoptera litura. RESULTS: We identify 74 candidate genes involved in odor detection and recognition, encoding 26 ORs, 21 OBPs, 18 CSPs and 9 IRs. We examine their expression levels in both sexes and seek evidence for their function by relating their expression with levels of EAG response in male and female antennae to 58 host and non-host plant volatiles and sex pheromone components. The majority of olfactory genes showed sex-biased expression, usually male-biased in ORs. A link between OR gene expression and antennal responses to odors was evident, a third of the compounds tested evoking a sex-biased response, in every case also male-biased. Two candidate pheromone receptors, OR14 and OR23 were especially strongly expressed and male-biased and we suggest that these may respond to the two female sex pheromone components of S. litura, Z9E11-14:OAc and Z9E12-14:OAc, which evoked strongly male-biased EAG responses. CONCLUSIONS: Our results provide the molecular basis for elucidating the olfactory profile of moths and the sexual divergence of their behavior and could enable the targeting of particular genes, and behaviors for pest management.


Assuntos
Spodoptera/genética , Transcriptoma , Animais , Antenas de Artrópodes/efeitos dos fármacos , Antenas de Artrópodes/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Proteínas de Insetos/genética , Masculino , RNA/análise , RNA/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real , Receptores Odorantes/genética , Análise de Sequência de RNA , Atrativos Sexuais/química , Atrativos Sexuais/farmacologia
10.
J Pharmacol Exp Ther ; 348(1): 46-58, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24190916

RESUMO

Selected nicotinic agonists were used to activate and desensitize high-sensitivity (HS) (α4)2(ß2)3) or low-sensitivity (LS) (α4)3(ß2)2) isoforms of human α4ß2-nicotinic acetylcholine receptors (nAChRs). Function was assessed using (86)Rb(+) efflux in a stably transfected SH-EP1-hα4ß2 human epithelial cell line, and two-electrode voltage-clamp electrophysiology in Xenopus laevis oocytes expressing concatenated pentameric HS or LS α4ß2-nAChR constructs (HSP and LSP). Unlike previously studied agonists, desensitization by the highly selective agonists A-85380 [3-(2(S)-azetidinylmethoxy)pyridine] and sazetidine-A (Saz-A) preferentially reduced α4ß2-nAChR HS-phase versus LS-phase responses. The concatenated-nAChR experiments confirmed that approximately 20% of LS-isoform acetylcholine-induced function occurs in an HS-like phase, which is abolished by Saz-A preincubation. Six mutant LSPs were generated, each targeting a conserved agonist binding residue within the LS-isoform-only α4(+)/(-)α4 interface agonist binding site. Every mutation reduced the percentage of LS-phase function, demonstrating that this site underpins LS-phase function. Oocyte-surface expression of the HSP and each of the LSP constructs was statistically indistinguishable, as measured using ß2-subunit-specific [(125)I]mAb295 labeling. However, maximum function is approximately five times greater on a "per-receptor" basis for unmodified LSP versus HSP α4ß2-nAChRs. Thus, recruitment of the α4(+)/(-)α4 site at higher agonist concentrations appears to augment otherwise-similar function mediated by the pair of α4(+)/(-)ß2 sites shared by both isoforms. These studies elucidate the receptor-level differences underlying the differential pharmacology of the two α4ß2-nAChR isoforms, and demonstrate that HS versus LS α4ß2-nAChR activity can be selectively manipulated using pharmacological approaches. Since α4ß2 nAChRs are the predominant neuronal subtype, these discoveries likely have significant functional implications, and may provide important insights for drug discovery and development.


Assuntos
Agonistas Nicotínicos/metabolismo , Subunidades Proteicas/metabolismo , Receptores Nicotínicos/química , Receptores Nicotínicos/classificação , Animais , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/genética , Sítios de Ligação/fisiologia , Linhagem Celular , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Feminino , Humanos , Agonistas Nicotínicos/química , Oócitos/química , Oócitos/metabolismo , Subunidades Proteicas/química , Subunidades Proteicas/genética , Receptores Nicotínicos/genética , Xenopus laevis
11.
Nat Commun ; 15(1): 5216, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38890331

RESUMO

Hyperpolarization and cyclic nucleotide (HCN) activated ion channels are critical for the automaticity of action potentials in pacemaking and rhythmic electrical circuits in the human body. Unlike most voltage-gated ion channels, the HCN and related plant ion channels activate upon membrane hyperpolarization. Although functional studies have identified residues in the interface between the voltage-sensing and pore domain as crucial for inverted electromechanical coupling, the structural mechanisms for this unusual voltage-dependence remain unclear. Here, we present cryo-electron microscopy structures of human HCN1 corresponding to Closed, Open, and a putative Intermediate state. Our structures reveal that the downward motion of the gating charges past the charge transfer center is accompanied by concomitant unwinding of the inner end of the S4 and S5 helices, disrupting the tight gating interface observed in the Closed state structure. This helix-coil transition at the intracellular gating interface accompanies a concerted iris-like dilation of the pore helices and underlies the reversed voltage dependence of HCN channels.


Assuntos
Microscopia Crioeletrônica , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Ativação do Canal Iônico , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/química , Humanos , Canais de Potássio/química , Canais de Potássio/metabolismo , Modelos Moleculares , Potenciais da Membrana/fisiologia
12.
bioRxiv ; 2023 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-37645882

RESUMO

Hyperpolarization and cyclic-nucleotide (HCN) activated ion channels play a critical role in generating self-propagating action potentials in pacemaking and rhythmic electrical circuits in the human body. Unlike most voltage-gated ion channels, the HCN channels activate upon membrane hyperpolarization, but the structural mechanisms underlying this gating behavior remain unclear. Here, we present cryo-electron microscopy structures of human HCN1 in Closed, Intermediate, and Open states. Our structures reveal that the inward motion of two gating charges past the charge transfer center (CTC) and concomitant tilting of the S5 helix drives the opening of the central pore. In the intermediate state structure, a single gating charge is positioned below the CTC and the pore appears closed, whereas in the open state structure, both charges move past CTC and the pore is fully open. Remarkably, the downward motion of the voltage sensor is accompanied by progressive unwinding of the inner end of S4 and S5 helices disrupting the tight gating interface that stabilizes the Closed state structure. This "melting" transition at the intracellular gating interface leads to a concerted iris-like displacement of S5 and S6 helices, resulting in pore opening. These findings reveal key structural features that are likely to underlie reversed voltage-dependence of HCN channels.

13.
J Biol Chem ; 286(44): 37905-37918, 2011 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-21873428

RESUMO

To further the understanding of functional α6α5*-nicotinic acetylcholine receptors (nAChR; the asterisk (*) indicates known or possible presence of other subunits), we have heterologously expressed in oocytes different, mouse or human, nAChR subunit combinations. Coexpression with wild-type α5 subunits or chimeric α5/ß3 subunits (in which the human α5 subunit N-terminal, extracellular domain is linked to the remaining domains of the human ß3 subunit) almost completely abolishes the very small amount of function seen for α6ß4*-nAChR and does not induce function of α6ß2*-nAChR. Coexpression with human α5(V9)'(S) subunits bearing a valine 290 to serine mutation in the 9' position of the second transmembrane domain does not rescue the function of α6ß4*-nAChR or induce function of α6ß2*-nAChR. However, coexpression with mutant chimeric α5/ß3(V9)'(S) subunits has a gain-of-function effect (higher functional expression and agonist sensitivity and spontaneous opening inhibited by mecamylamine) on α6ß4*-nAChR. Moreover, N143D + M145V mutations in the α6 subunit N-terminal domain enable α5/ß3(V9)'(S) subunits to have a gain-of-function effect on α6ß2*-nAChR. nAChR containing chimeric α6/α3 subunits plus either ß2 or ß4 subunits have some function that is modulated in the presence of α5 or α5/ß3 subunits. Coexpression with α5/ß3(V9)'(S) subunits has a gain-of-function effect more pronounced than that in the presence of α5(V9)'(S) subunits. Gain-of-function effects are dependent, sometimes subtly, on the nature and apparently the extracellular, cytoplasmic, and/or transmembrane domain topology of partner subunits. These studies yield insight into assembly of functional α6α5*-nAChR and provide tools for development of α6*-nAChR-selective ligands that could be important in the treatment of nicotine dependence, and perhaps other neurological diseases.


Assuntos
Mutação , Proteínas do Tecido Nervoso/genética , Receptores Nicotínicos/genética , Animais , Citoplasma/metabolismo , DNA Complementar/metabolismo , Eletrofisiologia/métodos , Feminino , Humanos , Ligantes , Proteínas do Tecido Nervoso/metabolismo , Nicotina/metabolismo , Oócitos/metabolismo , Mutação Puntual , Estrutura Terciária de Proteína , Receptores Nicotínicos/metabolismo , Proteínas Recombinantes de Fusão/química , Xenopus laevis
14.
J Biol Chem ; 286(44): 37976-37989, 2011 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-21832048

RESUMO

Despite the apparent function of naturally expressed mammalian α6*-nicotinic acetylcholine receptors (α6*-nAChR; where * indicates the known or possible presence of additional subunits), their functional and heterologous expression has been difficult. Here, we report that coexpression with wild-type ß3 subunits abolishes the small amount of function typically seen for all-human or all-mouse α6ß4*-nAChR expressed in Xenopus oocytes. However, levels of function and agonist potencies are markedly increased, and there is atropine-sensitive blockade of spontaneous channel opening upon coexpression of α6 and ß4 subunits with mutant ß3 subunits harboring valine-to-serine mutations at 9'- or 13'-positions. There is no function when α6 and ß2 subunits are expressed alone or in the presence of wild-type or mutant ß3 subunits. Interestingly, hybrid nAChR containing mouse α6 and human (h) ß4 subunits have function potentiated rather than suppressed by coexpression with wild-type hß3 subunits and potentiated further upon coexpression with hß3(V9'S) subunits. Studies using nAChR chimeric mouse/human α6 subunits indicated that residues involved in effects seen with hybrid nAChR are located in the α6 subunit N-terminal domain. More specifically, nAChR hα6 subunit residues Asn-143 and Met-145 are important for dominant-negative effects of nAChR hß3 subunits on hα6hß4-nAChR function. Asn-143 and additional residues in the N-terminal domain of nAChR hα6 subunits are involved in the gain-of-function effects of nAChR hß3(V9'S) subunits on α6ß2*-nAChR function. These studies illuminate the structural bases for effects of ß3 subunits on α6*-nAChR function and suggest that unique subunit interfaces involving the complementary rather than the primary face of α6 subunits are involved.


Assuntos
Receptores Nicotínicos/química , Receptores Nicotínicos/genética , Sequência de Aminoácidos , Animais , Antidepressivos/uso terapêutico , Relação Dose-Resposta a Droga , Humanos , Canais Iônicos/química , Ligantes , Camundongos , Dados de Sequência Molecular , Neurotransmissores/metabolismo , Mutação Puntual , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Abandono do Hábito de Fumar
15.
J Biol Chem ; 286(28): 25331-40, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21610071

RESUMO

Binding of a neurotransmitter to its membrane receptor opens an integral ion conducting pore. However, prolonged exposure to the neurotransmitter drives the receptor to a refractory state termed desensitization, which plays an important role in shaping synaptic transmission. Despite intensive research in the past, the structural mechanism of desensitization is still elusive. Using mutagenesis and voltage clamp in an oocyte expression system, we provide several lines of evidence supporting a novel hypothesis that uncoupling between binding and gating machinery is the underlying mechanism for α7 nicotinic receptor (nAChR) desensitization. First, the decrease in gate tightness was highly correlated to the reduced desensitization. Second, nonfunctional mutants in three important coupling loops (loop 2, loop 7, and the M2-M3 linker) could be rescued by a gating mutant. Furthermore, the decrease in coupling strength in these rescued coupling loop mutants reversed the gating effect on desensitization. Finally, coupling between M1 and hinge region of the M2-M3 linker also influenced the receptor desensitization. Thus, the uncoupling between N-terminal domain and transmembrane domain, governed by the balance of coupling strength and gate tightness, underlies the mechanism of desensitization for the α7 nAChR.


Assuntos
Ativação do Canal Iônico/fisiologia , Receptores Nicotínicos/metabolismo , Animais , Expressão Gênica , Humanos , Mutagênese , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Receptores Nicotínicos/genética , Relação Estrutura-Atividade , Xenopus laevis , Receptor Nicotínico de Acetilcolina alfa7
16.
Biochim Biophys Acta ; 1808(7): 1818-26, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21419751

RESUMO

External Na(+) self-inhibition is an intrinsic feature of epithelial sodium channels (ENaC). Cpt-cAMP regulates heterologous guinea pig but not rat αßγ ENaC in a ligand-gated manner. We hypothesized that cpt-cAMP may eliminate the self-inhibition of human ENaC thereby open channels. Regulation of self-inhibition by this compound in oocytes was analyzed using the two-electrode voltage clamp and Ussing chamber setups. External cpt-cAMP stimulated human but not rat and murine αßγ ENaC in a dose- and external Na(+) concentration-dependent fashion. Intriguingly, cpt-cAMP activated human δßγ more potently than αßγ channels, suggesting that structural diversity in ectoloop between human α, δ, and those ENaC of other species determines the stimulating effects of cpt-cAMP. Cpt-cAMP increased the ratio of stationary and maximal currents. Mutants having abolished self-inhibition (ß(ΔV348) and γ(H233R)) almost completely eliminated cpt-cAMP mediated activation of ENaC. On the other hand, mutants both enhancing self-inhibition and elevating cpt-cAMP sensitivity increased the stimulating effects of the compound. This compound, however, could not activate already fully opened channels, e.g., degenerin mutation (αß(S520C)γ) and the proteolytically cleaved ENaC by plasmin. Cpt-cAMP activated native ENaC to the same extent as that for heterologous ENaC in human lung epithelial cells. Our data demonstrate that cpt-cAMP, a broadly used PKA activator, stimulates human αßγ and δßγ ENaC channels by relieving self-inhibition.


Assuntos
AMP Cíclico/análogos & derivados , Agonistas do Canal de Sódio Epitelial , Tionucleotídeos/farmacologia , Animais , Células Cultivadas , AMP Cíclico/farmacologia , Eletroquímica , Bloqueadores do Canal de Sódio Epitelial , Canais Epiteliais de Sódio/genética , Feminino , Humanos , Camundongos , Mutagênese Sítio-Dirigida , Ratos , Xenopus laevis
17.
Am J Physiol Lung Cell Mol Physiol ; 302(12): L1262-72, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22505667

RESUMO

Salt absorption via apical epithelial sodium channels (ENaC) is a critical rate-limiting process in maintaining airway and lung lining fluid at the physiological level. δ ENaC (termed δ1 in this article) has been detected in human lung epithelial cells in addition to α, ß, and γ subunits (Ji HL, Su XF, Kedar S, Li J, Barbry P, Smith PR, Matalon S, Benos DJ. J Biol Chem 281: 8233-8241, 2006; Nie HG, Chen L, Han DY, Li J, Song WF, Wei SP, Fang XH, Gu X, Matalon S, Ji HL, J Physiol 587: 2663-2676, 2009) and may contribute to the differences in the biophysical properties of amiloride-inhibitable cation channels in pulmonary epithelial cells. Here we cloned a splicing variant of the δ1 ENaC, namely, δ2 ENaC in human bronchoalveolar epithelial cells (16HBEo). δ2 ENaC possesses 66 extra amino acids attached to the distal amino terminal tail of the δ1 ENaC. δ2 ENaC was expressed in both alveolar type I and II cells of human lungs as revealed by in situ hybridization and real-time RT-PCR. To characterize the biophysical and pharmacological features of the splicing variant, we injected Xenopus oocytes with human ENaC cRNAs and measured whole cell and single channel currents of δ1ßγ, δ2ßγ, and αßγ channels. Oocytes injected with δ2ßγ cRNAs exhibited whole cell currents significantly greater than those expressing δ1ßγ and αßγ channels. Single channel activity, unitary conductance, and open probability of δ2ßγ channels were significantly greater compared with δ1ßγ and αßγ channels. In addition, δ2ßγ and δ1ßγ channels displayed significant differences in apparent Na(+) affinity, dissociation constant for amiloride (K(i)(amil)), the EC(50) for capsazepine activation, and gating kinetics by protons. Channels comprising of this novel splice variant may contribute to the diversities of native epithelial Na(+) channels.


Assuntos
Células Epiteliais Alveolares/fisiologia , Canais Epiteliais de Sódio/genética , Canais Epiteliais de Sódio/fisiologia , Ativação do Canal Iônico , Mucosa Respiratória/fisiologia , Sódio/metabolismo , Células Epiteliais Alveolares/efeitos dos fármacos , Amilorida/metabolismo , Amilorida/farmacologia , Sequência de Aminoácidos , Animais , Transporte Biológico , Capsaicina/análogos & derivados , Capsaicina/metabolismo , Clonagem Molecular , Condutividade Elétrica , Exocitose , Humanos , Concentração de Íons de Hidrogênio , Ativação do Canal Iônico/efeitos dos fármacos , Pulmão , Oócitos/citologia , Oócitos/metabolismo , Técnicas de Patch-Clamp , Isoformas de Proteínas/fisiologia , Splicing de RNA , Mucosa Respiratória/citologia , Mucosa Respiratória/efeitos dos fármacos , Xenopus
18.
J Neurochem ; 121(3): 349-61, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22309577

RESUMO

The nicotinic acetylcholine receptor (nAChR) ß3 subunit is thought to serve an accessory role in nAChR subtypes expressed in dopaminergic regions implicated in drug dependence and reward. When ß3 subunits are expressed in excess, they have a dominant-negative effect on function of selected nAChR subtypes. In this study, we show, in Xenopus oocytes expressing α2, α3 or α4 plus either ß2 or ß4 subunits, that in the presumed presence of similar amounts of each nAChR subunit, co-expression with wild-type ß3 subunits generally (except for α3*-nAChR) lowers amplitudes of agonist-evoked, inward peak currents by 20-50% without having dramatic effects (≤ 2-fold) on agonist potencies. By contrast, co-expression with mutant ß3(V9'S) subunits generally (except for α4ß2*-nAChR) increases agonist potencies, consistent with an expected gain-of-function effect. This most dramatically demonstrates formation of complexes containing three kinds of subunit. Moreover, for oocytes expressing nAChR containing any α subunit plus ß4 and ß3(V9'S) subunits, there is spontaneous channel opening sensitive to blockade by the open channel blocker, atropine. Collectively, the results indicate that ß3 subunits integrate into all of the studied receptor assemblies and suggest that natural co-expression with ß3 subunits can influence levels of expression and agonist sensitivities of several nAChR subtypes.


Assuntos
Receptores Nicotínicos/fisiologia , Acetilcolina/farmacologia , Animais , Atropina/farmacologia , Clonagem Molecular , Interpretação Estatística de Dados , Feminino , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Antagonistas Muscarínicos/farmacologia , Mutagênese , Mutação/genética , Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Oócitos/metabolismo , Técnicas de Patch-Clamp , Plasmídeos , RNA Complementar/administração & dosagem , RNA Complementar/genética , Receptores Nicotínicos/efeitos dos fármacos , Transcrição Gênica , Xenopus , Proteínas de Xenopus
19.
Ann Neurol ; 69(4): 664-72, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21391233

RESUMO

OBJECTIVE: Human hypothalamic hamartomas (HHs) are highly associated with treatment-resistant gelastic seizures. HHs are intrinsically epileptogenic, although the basic cellular mechanisms responsible for seizure activity are unknown. Altered gamma-aminobutyric acid (GABA) function can contribute to epileptogenesis in humans and animal models. Recently, functional GABA(A) receptor (GABA(A) R) rundown has been described in surgically resected human temporal lobe epilepsy tissue. We asked whether functional GABA(A) R rundown also occurs in human HH neurons. METHODS: GABA(A) R-mediated currents were measured using perforated patch-clamp recordings in single neurons acutely dissociated from surgically resected HH tissue. In addition, functional GABA(A) Rs were expressed in Xenopus oocytes after microinjection with membrane fractions from either HH or control hypothalamus, and were studied with 2-electrode voltage-clamp recordings. RESULTS: Perforated patch-clamp recordings in dissociated HH neurons showed that repetitive exposure to GABA (5 consecutive exposures to 0.1 mM GABA with 1-second duration and at 20-second intervals) induced a time-dependent rundown of whole-cell currents in small HH neurons, whereas large HH neurons showed much less rundown using the same protocol. Functional rundown was not observed in HH neurons with repetitive exposure to glycine or glutamate. Two-electrode voltage-clamp recordings (6 consecutive exposures to 1 mM GABA with 10-second duration and at 40-second intervals) induced GABA current rundown in Xenopus oocytes microinjected with HH membrane proteins, but not in the oocytes expressing hypothalamic membrane proteins derived from human autopsy controls. Functional rundown of GABA currents was significantly attenuated by intracellular application of adenosine triphosphate or the nonspecific phosphatase inhibitor, okadaic acid. INTERPRETATION: Neurons from surgically resected human HH demonstrate functional rundown of GABA(A) R-mediated transmembrane currents in response to GABA agonist exposure. Rundown may be a marker for impaired GABAergic function and a contributing mechanism for seizure genesis within HH tissue.


Assuntos
Epilepsia/metabolismo , Neurônios/metabolismo , Receptores de GABA-A/metabolismo , Animais , Epilepsia/etiologia , Hamartoma/complicações , Hamartoma/metabolismo , Hamartoma/patologia , Humanos , Doenças Hipotalâmicas/complicações , Doenças Hipotalâmicas/metabolismo , Doenças Hipotalâmicas/patologia , Hipotálamo/metabolismo , Oócitos , Técnicas de Patch-Clamp/instrumentação , Técnicas de Patch-Clamp/métodos , Xenopus
20.
Int J Mol Sci ; 13(4): 4920-4936, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22606020

RESUMO

Endothelial cells (ECs) that line the lumen of blood vessels are important players in blood vessel formation, and EC migration is a key component of the angiogenic process. Thus, identification of genes that are specifically or preferentially expressed in vascular ECs and in-depth understanding of their biological functions may lead to discovery of new therapeutic targets. We have previously reported molecular characterization of human endothelial cell-specific molecule 2 (ECSM2)/endothelial cell-specific chemotaxis regulator (ECSCR). In the present study, we cloned two mouse full-length cDNAs by RT-PCR, which encode two putative ECSCR isoform precursors with considerable homology to the human ECSCR. Nucleotide sequence and exon-intron junction analyses suggested that they are alternative splicing variants (ECSCR isoform-1 and -2), differing from each other in the first and second exons. Quantitative RT-PCR results revealed that isoform-2 is the predominant form, which was most abundant in heart, lung, and muscles, and moderately abundant in uterus and testis. In contrast, the expression of isoform-1 seemed to be more enriched in testis. To further explore their potential cellular functions, we expressed GFP- and FLAG-tagged ECSCR isoforms, respectively, in an ECSCR deficient cell line (HEK293). Interestingly, the actual sizes of either ECSCR-GFP or -FLAG fusion proteins detected by immunoblotting are much larger than their predicted sizes, suggesting that both isoforms are glycoproteins. Fluorescence microscopy revealed that both ECSCR isoforms are localized at the cell surface, which is consistent with the structural prediction. Finally, we performed cell migration assays using mouse endothelial MS1 cells overexpressing GFP alone, isoform-1-GFP, and isoform-2-GFP, respectively. Our results showed that both isoforms significantly inhibited vascular epidermal growth factor (VEGF)-induced cell migration. Taken together, we have provided several lines of experimental evidence that two mouse ECSCR splicing variants/isoform precursors exist. They are differentially expressed in a variety of tissue types and likely involved in modulation of vascular EC migration. We have also defined the gene structure of mouse ECSCR using bioinformatics tools, which provides new information towards a better understanding of alternative splicing of ECSCR.


Assuntos
Processamento Alternativo/genética , Proteínas Reguladoras de Apoptose/genética , Movimento Celular/genética , Células Endoteliais/citologia , Neovascularização Fisiológica/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Clonagem Molecular , Proteínas de Fluorescência Verde/genética , Células HEK293 , Humanos , Proteínas de Membrana , Camundongos , Dados de Sequência Molecular , Isoformas de Proteínas/genética , Alinhamento de Sequência , Fator A de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA