Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 38(1): e23374, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38161283

RESUMO

This study was undertaken to identify and characterize the first ligands capable of selectively identifying nicotinic acetylcholine receptors containing α7 and ß2 subunits (α7ß2-nAChR subtype). Basal forebrain cholinergic neurons express α7ß2-nAChR. Here, they appear to mediate neuronal dysfunction induced by the elevated levels of oligomeric amyloid-ß associated with early Alzheimer's disease. Additional work indicates that α7ß2-nAChR are expressed across several further critically important cholinergic and GABAergic neuronal circuits within the central nervous system. Further studies, however, are significantly hindered by the inability of currently available ligands to distinguish heteromeric α7ß2-nAChR from the closely related and more widespread homomeric α7-only-nAChR subtype. Functional screening using two-electrode voltage-clamp electrophysiology identified a family of α7ß2-nAChR-selective analogs of α-conotoxin PnIC (α-CtxPnIC). A combined electrophysiology, functional kinetics, site-directed mutagenesis, and molecular dynamics approach was used to further characterize the α7ß2-nAChR selectivity and site of action of these α-CtxPnIC analogs. We determined that α7ß2-nAChR selectivity of α-CtxPnIC analogs arises from interactions at a site distinct from the orthosteric agonist-binding site shared between α7ß2- and α7-only-nAChR. As numerous previously identified α-Ctx ligands are competitive antagonists of orthosteric agonist-binding sites, this study profoundly expands the scope of use of α-Ctx ligands (which have already provided important nAChR research and translational breakthroughs). More immediately, analogs of α-CtxPnIC promise to enable, for the first time, both comprehensive mapping of the distribution of α7ß2-nAChR and detailed investigations of their physiological roles.


Assuntos
Receptores Nicotínicos , Receptor Nicotínico de Acetilcolina alfa7 , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Colinérgicos , Sítios de Ligação , Neurônios GABAérgicos/metabolismo , Antagonistas Nicotínicos/farmacologia
2.
Pharmacol Res ; 191: 106743, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37084859

RESUMO

Initiated by findings that Alzheimer's disease is associated with a profound loss of cholinergic markers in human brain, decades of studies have examined the interactions between specific subtypes of nicotinic acetylcholine receptors and amyloid-ß [derived from the amyloid precursor protein (APP), which is cleaved to yield variable isoforms of amyloid-ß]. We review the evolving understanding of amyloid-ß's roles in Alzheimer's disease and pioneering studies that highlighted a role of nicotinic acetylcholine receptors in mediating important aspects of amyloid-ß's effects. This review also surveys the current state of research into amyloid-ß / nicotinic acetylcholine receptor interactions. The field has reached an exciting point in which common themes are emerging from the wide range of prior research and a range of accessible, relevant model systems are available to drive further progress. We highlight exciting new areas of inquiry and persistent challenges that need to be considered while conducting this research. Studies of amyloid-ß and the nicotinic acetylcholine receptor populations that it interacts with provide opportunities for innovative basic and translational scientific breakthroughs related to nicotinic receptor biology, Alzheimer's disease, and cholinergic contributions to cognition more broadly.


Assuntos
Doença de Alzheimer , Receptores Nicotínicos , Animais , Humanos , Receptores Nicotínicos/metabolismo , Doença de Alzheimer/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Peptídeos beta-Amiloides/metabolismo , Colinérgicos , Modelos Animais de Doenças
3.
J Neurosci ; 41(3): 555-575, 2021 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-33239400

RESUMO

Neuronal and network-level hyperexcitability is commonly associated with increased levels of amyloid-ß (Aß) and contribute to cognitive deficits associated with Alzheimer's disease (AD). However, the mechanistic complexity underlying the selective loss of basal forebrain cholinergic neurons (BFCNs), a well-recognized characteristic of AD, remains poorly understood. In this study, we tested the hypothesis that the oligomeric form of amyloid-ß (oAß42), interacting with α7-containing nicotinic acetylcholine receptor (nAChR) subtypes, leads to subnucleus-specific alterations in BFCN excitability and impaired cognition. We used single-channel electrophysiology to show that oAß42 activates both homomeric α7- and heteromeric α7ß2-nAChR subtypes while preferentially enhancing α7ß2-nAChR open-dwell times. Organotypic slice cultures were prepared from male and female ChAT-EGFP mice, and current-clamp recordings obtained from BFCNs chronically exposed to pathophysiologically relevant level of oAß42 showed enhanced neuronal intrinsic excitability and action potential firing rates. These resulted from a reduction in action potential afterhyperpolarization and alterations in the maximal rates of voltage change during spike depolarization and repolarization. These effects were observed in BFCNs from the medial septum diagonal band and horizontal diagonal band, but not the nucleus basalis. Last, aged male and female APP/PS1 transgenic mice, genetically null for the ß2 nAChR subunit gene, showed improved spatial reference memory compared with APP/PS1 aged-matched littermates. Combined, these data provide a molecular mechanism supporting a role for α7ß2-nAChR in mediating the effects of oAß42 on excitability of specific populations of cholinergic neurons and provide a framework for understanding the role of α7ß2-nAChR in oAß42-induced cognitive decline.


Assuntos
Peptídeos beta-Amiloides/genética , Prosencéfalo Basal/fisiopatologia , Disfunção Cognitiva/genética , Disfunção Cognitiva/fisiopatologia , Sistema Nervoso Parassimpático/fisiopatologia , Fragmentos de Peptídeos/genética , Transdução de Sinais/genética , Receptor Nicotínico de Acetilcolina alfa7/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Linhagem Celular , Fenômenos Eletrofisiológicos , Feminino , Genótipo , Humanos , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Transgênicos , Neurônios/patologia
4.
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
5.
FASEB J ; 31(4): 1398-1420, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28100642

RESUMO

This study investigates-for the first time to our knowledge-the existence and mechanisms of functional interactions between the endogenous mammalian prototoxin, lynx1, and α3- and ß4-subunit-containing human nicotinic acetylcholine receptors (α3ß4*-nAChRs). Concatenated gene constructs were used to express precisely defined α3ß4*-nAChR isoforms (α3ß4)2ß4-, (α3ß4)2α3-, (α3ß4)2α5(398D)-, and (α3ß4)2α5(398N)-nAChR in Xenopus oocytes. In the presence or absence of lynx1, α3ß4*-nAChR agonist responses were recorded by using 2-electrode voltage clamp and single-channel electrophysiology, whereas radioimmunolabeling measured cell-surface expression. Lynx1 reduced (α3ß4)2ß4-nAChR function principally by lowering cell-surface expression, whereas single-channel effects were primarily responsible for reducing (α3ß4)2α3-nAChR function [decreased unitary conductance (≥50%), altered burst proportions (3-fold reduction in the proportion of long bursts), and enhanced closed dwell times (3- to 6-fold increase)]. Alterations in both cell-surface expression and single-channel properties accounted for the reduction in (α3ß4)2α5-nAChR function that was mediated by lynx1. No effects were observed when α3ß4*-nAChRs were coexpressed with mutated lynx1 (control). Lynx1 is expressed in the habenulopeduncular tract, where α3ß4*-α5*-nAChR subtypes are critical contributors to the balance between nicotine aversion and reward. This gives our findings a high likelihood of physiologic significance. The exquisite isoform selectivity of lynx1 interactions provides new insights into the mechanisms and allosteric sites [α(-)-interface containing] by which prototoxins can modulate nAChR function.-George, A. A., Bloy, A., Miwa, J. M., Lindstrom, J. M., Lukas, R. J., Whiteaker, P. Isoform-specific mechanisms of α3ß4*-nicotinic acetylcholine receptor modulation by the prototoxin lynx1.


Assuntos
Proteínas Ligadas por GPI/metabolismo , Receptores Nicotínicos/metabolismo , Potenciais de Ação , Proteínas Adaptadoras de Transdução de Sinal , Animais , Membrana Celular/metabolismo , Membrana Celular/fisiologia , Proteínas Ligadas por GPI/genética , Humanos , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Transporte Proteico , Receptores Nicotínicos/química , Receptores Nicotínicos/genética , Xenopus
6.
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
7.
J Biol Chem ; 291(5): 2444-59, 2016 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-26644472

RESUMO

Two α4ß2 nicotinic acetylcholine receptor (α4ß2-nAChR) isoforms exist with (α4)2(ß2)3 and (α4)3(ß2)2 subunit stoichiometries and high versus low agonist sensitivities (HS and LS), respectively. Both isoforms contain a pair of α4(+)/(-)ß2 agonist-binding sites. The LS isoform also contains a unique α4(+)/(-)α4 site with lower agonist affinity than the α4(+)/(-)ß2 sites. However, the relative roles of the conserved α4(+)/(-)ß2 agonist-binding sites in and between the isoforms have not been studied. We used a fully linked subunit concatemeric nAChR approach to express pure populations of HS or LS isoform α4ß2*-nAChR. This approach also allowed us to mutate individual subunit interfaces, or combinations thereof, on each isoform background. We used this approach to systematically mutate a triplet of ß2 subunit (-)-face E-loop residues to their non-conserved α4 subunit counterparts or vice versa (ß2HQT and α4VFL, respectively). Mutant-nAChR constructs (and unmodified controls) were expressed in Xenopus oocytes. Acetylcholine concentration-response curves and maximum function were measured using two-electrode voltage clamp electrophysiology. Surface expression was measured with (125)I-mAb 295 binding and was used to define function/nAChR. If the α4(+)/(-)ß2 sites contribute equally to function, making identical ß2HQT substitutions at either site should produce similar functional outcomes. Instead, highly differential outcomes within the HS isoform, and between the two isoforms, were observed. In contrast, α4VFL mutation effects were very similar in all positions of both isoforms. Our results indicate that the identity of subunits neighboring the otherwise equivalent α4(+)/(-)ß2 agonist sites modifies their contributions to nAChR activation and that E-loop residues are an important contributor to this neighbor effect.


Assuntos
Regulação da Expressão Gênica , Receptores Nicotínicos/química , Acetilcolina/química , Sítio Alostérico , Animais , Azetidinas/química , Sítios de Ligação/genética , DNA Complementar/metabolismo , Eletrofisiologia , Humanos , Mutagênese Sítio-Dirigida , Mutação , Nicotina/química , Agonistas Nicotínicos/química , Oócitos/metabolismo , Técnicas de Patch-Clamp , Ligação Proteica , Isoformas de Proteínas , RNA/metabolismo , Xenopus laevis
8.
FASEB J ; 30(3): 1109-19, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26586467

RESUMO

Prototoxins are a diverse family of membrane-tethered molecules expressed in the nervous system that modulate nicotinic cholinergic signaling, but their functions and specificity have yet to be completely explored. We tested the selectivity and efficacy of leukocyte antigen, PLAUR (plasminogen activator, urokinase receptor) domain-containing (LYPD)-6B on α3ß4-, α3α5ß4-, and α7-containing nicotinic acetylcholine receptors (nAChRs). To constrain stoichiometry, fusion proteins encoding concatemers of human α3, ß4, and α5 (D and N variants) subunits were expressed in Xenopus laevis oocytes and tested with or without LYPD6B. We used the 2-electrode voltage-clamp method to quantify responses to acetylcholine (ACh): agonist sensitivity (EC50), maximal agonist-induced current (Imax), and time constant (τ) of desensitization. For ß4-α3-α3-ß4-α3 and ß4-α3-ß4-α3-α3, LYPD6B decreased EC50 from 631 to 79 µM, reduced Imax by at least 59%, and decreased τ. For ß4-α3-α5D-ß4-α3 and ß4-α3-ß4-α-α5D, LYPD6B decreased Imax by 63 and 32%, respectively. Thus, LYPD6B acted only on (α3)3(ß4)2 and (α3)2(α5D)(ß4)2 and did not affect the properties of (α3)2(ß4)3, α7, or (α3)2(α5N)(ß4)2 nAChRs. Therefore, LYPD6B acts as a mixed modulator that enhances the sensitivity of (α3)3(ß4)2 nAChRs to ACh while reducing ACh-induced whole-cell currents. LYPD6B also negatively modulates α3ß4 nAChRs that include the α5D common human variant, but not the N variant associated with nicotine dependence.


Assuntos
Receptores Nicotínicos/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Acetilcolina/farmacologia , Animais , Humanos , Nicotina/farmacologia , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Subunidades Proteicas/metabolismo , Xenopus laevis/metabolismo
9.
Mol Pharmacol ; 88(5): 881-93, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26330550

RESUMO

Ligands that selectively inhibit human α3ß2 and α6ß2 nicotinic acetylcholine receptor (nAChRs) and not the closely related α3ß4 and α6ß4 subtypes are lacking. Current α-conotoxins (α-Ctxs) that discriminate among these nAChR subtypes in rat fail to discriminate among the human receptor homologs. In this study, we describe the development of α-Ctx LvIA(N9R,V10A) that is 3000-fold more potent on oocyte-expressed human α3ß2 than α3ß4 and 165-fold more potent on human α6/α3ß2ß3 than α6/α3ß4 nAChRs. This analog was used in conjuction with three other α-Ctx analogs and patch-clamp electrophysiology to characterize the nAChR subtypes expressed by human adrenal chromaffin cells. LvIA(N9R,V10A) showed little effect on the acetylcholine-evoked currents in these cells at concentrations expected to inhibit nAChRs with ß2 ligand-binding sites. In contrast, the ß4-selective α-Ctx BuIA(T5A,P6O) inhibited >98% of the acetylcholine-evoked current, indicating that most of the heteromeric receptors contained ß4 ligand-binding sites. Additional studies using the α6-selective α-Ctx PeIA(A7V,S9H,V10A,N11R,E14A) indicated that the predominant heteromeric nAChR expressed by human adrenal chromaffin cells is the α3ß4* subtype (asterisk indicates the possible presence of additional subunits). This conclusion was supported by polymerase chain reaction experiments of human adrenal medulla gland and of cultured human adrenal chromaffin cells that demonstrated prominent expression of RNAs for α3, α5, α7, ß2, and ß4 subunits and a low abundance of RNAs for α2, α4, α6, and α10 subunits.


Assuntos
Medula Suprarrenal/metabolismo , Células Cromafins/metabolismo , Conotoxinas/farmacologia , Antagonistas Nicotínicos/farmacologia , Receptores Nicotínicos/metabolismo , Animais , Sítios de Ligação , Células Cultivadas , Humanos , Técnicas de Patch-Clamp , Isoformas de Proteínas , Ratos , Receptores Nicotínicos/classificação , Xenopus laevis
10.
FASEB J ; 28(4): 1842-53, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24398291

RESUMO

This study was performed to discover and characterize the first potent α3ß2-subtype-selective nicotinic acetylcholine receptor (nAChR) ligand. A novel α4/7-conotoxin, α-CTxLvIA, was cloned from Conus lividus. Its pharmacological profile at Xenopus laevis oocyte-expressed rat nAChR subtypes was determined by 2-electrode voltage-clamp electrophysiology, and its 3-dimensional (3D) structure was determined by NMR spectroscopy. α-CTx LvIA is a 16-aa C-terminally-amidated peptide with 2-disulfide bridges. Using rat subunits expressed in Xenopus oocytes, we found the highest affinity of α-CTxLvIA was for α3ß2 nAChRs (IC50 8.7 nM), where blockade was reversible within 2 min. IC50 values were >100 nM at α6/α3ß2ß3, α6/α3ß4, and α3ß4 nAChRs, and ≥3 µM at all other subtypes tested. α3ß2 vs. α6ß2 subtype selectivity was confirmed for human-subunit nAChRs with much greater preference (300-fold) for α3ß2 over α6ß2 nAChRs. This is the first α-CTx reported to show high selectivity for human α3ß2 vs. α6ß2 nAChRs. α-CTxLvIA adopts two similarly populated conformations water: one (assumed to be bioactive) is highly structured, whereas the other is mostly random coil in nature. Selectivity differences with the similarly potent, but less selective, α3ß2 nAChR antagonist α-CTx PeIA probably reside within the three residues, which differ in loop 2, given their otherwise similar 3D structures. α4/7-CTx LvIA is a new, potent, selective α3ß2 nAChR antagonist, which will enable detailed studies of α3ß2 nAChR structure, function, and physiological roles.


Assuntos
Conotoxinas/metabolismo , Caramujo Conus/metabolismo , Antagonistas Nicotínicos/metabolismo , Receptores Nicotínicos/metabolismo , Acetilcolina/farmacologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Ligação Competitiva , Agonistas Colinérgicos/farmacologia , Conotoxinas/genética , Conotoxinas/farmacologia , Caramujo Conus/genética , Feminino , Humanos , Potenciais da Membrana/efeitos dos fármacos , Modelos Moleculares , Dados de Sequência Molecular , Antagonistas Nicotínicos/química , Antagonistas Nicotínicos/farmacologia , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Oócitos/fisiologia , Técnicas de Patch-Clamp/instrumentação , Técnicas de Patch-Clamp/métodos , Ligação Proteica , Estrutura Terciária de Proteína , Ratos , Receptores Nicotínicos/química , Receptores Nicotínicos/genética , Xenopus laevis
11.
Mol Pharmacol ; 86(3): 306-17, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25002271

RESUMO

We examined α7ß2-nicotinic acetylcholine receptor (α7ß2-nAChR) expression in mammalian brain and compared pharmacological profiles of homomeric α7-nAChRs and α7ß2-nAChRs. α-Bungarotoxin affinity purification or immunoprecipitation with anti-α7 subunit antibodies (Abs) was used to isolate nAChRs containing α7 subunits from mouse or human brain samples. α7ß2-nAChRs were detected in forebrain, but not other tested regions, from both species, based on Western blot analysis of isolates using ß2 subunit-specific Abs. Ab specificity was confirmed in control studies using subunit-null mutant mice or cell lines heterologously expressing specific human nAChR subtypes and subunits. Functional expression in Xenopus oocytes of concatenated pentameric (α7)5-, (α7)4(ß2)1-, and (α7)3(ß2)2-nAChRs was confirmed using two-electrode voltage clamp recording of responses to nicotinic ligands. Importantly, pharmacological profiles were indistinguishable for concatenated (α7)5-nAChRs or for homomeric α7-nAChRs constituted from unlinked α7 subunits. Pharmacological profiles were similar for (α7)5-, (α7)4(ß2)1-, and (α7)3(ß2)2-nAChRs except for diminished efficacy of nicotine (normalized to acetylcholine efficacy) at α7ß2- versus α7-nAChRs. This study represents the first direct confirmation of α7ß2-nAChR expression in human and mouse forebrain, supporting previous mouse studies that suggested relevance of α7ß2-nAChRs in Alzheimer disease etiopathogenesis. These data also indicate that α7ß2-nAChR subunit isoforms with different α7/ß2 subunit ratios have similar pharmacological profiles to each other and to α7 homopentameric nAChRs. This supports the hypothesis that α7ß2-nAChR agonist activation predominantly or entirely reflects binding to α7/α7 subunit interface sites.


Assuntos
Prosencéfalo/metabolismo , Receptores Nicotínicos/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Animais , Sítios de Ligação , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Bungarotoxinas/farmacologia , Cerebelo/metabolismo , Feminino , Hipocampo/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Agonistas Nicotínicos/farmacologia , Antagonistas Nicotínicos/farmacologia , Oócitos/metabolismo , Multimerização Proteica , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Piridinas/farmacologia , Ensaio Radioligante , Receptores Nicotínicos/genética , Xenopus laevis , Receptor Nicotínico de Acetilcolina alfa7/genética
12.
J Neurochem ; 130(2): 185-98, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24661093

RESUMO

Nicotinic acetylcholine receptors (nAChR) of the α6ß2* subtype (where *indicates the possible presence of additional subunits) are prominently expressed on dopaminergic neurons. Because of this, their role in tobacco use and nicotine dependence has received much attention. Previous studies have demonstrated that α6ß2*-nAChR are down-regulated following chronic nicotine exposure (unlike other subtypes that have been investigated - most prominently α4ß2* nAChR). This study examines, for the first time, effects across a comprehensive chronic nicotine dose range. Chronic nicotine dose-responses and quantitative ligand-binding autoradiography were used to define nicotine sensitivity of changes in α4ß2*-nAChR and α6ß2*-nAChR expression. α6ß2*-nAChR down-regulation by chronic nicotine exposure in dopaminergic and optic-tract nuclei was ≈three-fold more sensitive than up-regulation of α4ß2*-nAChR. In contrast, nAChR-mediated [(3) H]-dopamine release from dopamine-terminal region synaptosomal preparations changed only in response to chronic treatment with high nicotine doses, whereas dopaminergic parameters (transporter expression and activity, dopamine receptor expression) were largely unchanged. Functional measures in olfactory tubercle preparations were made for the first time; both nAChR expression levels and nAChR-mediated functional measures changed differently between striatum and olfactory tubercles. These results show that functional changes measured using synaptosomal [(3) H]-DA release are primarily owing to changes in nAChR, rather than in dopaminergic, function. This study examined dose-response relationships for murine α6ß2*-nicotinic acetylcholine receptor (nAChR) down-regulation by chronic nicotine treatment. The ID50 value for α6ß2* down-regulation (35 nM) is ≈ 3x lower than the ED50 value for α4ß2* nAChR up-regulation (95 nM), both well within the range reached by human smokers. Chronic nicotine treatment altered α6ß2*- and α4ß2*-nAChR-mediated [(3) H]-dopamine release from striatal and olfactory tubercle synaptosomes, but dopaminergic parameters were largely unaffected. We conclude that functional changes are primarily driven by altered nAChR activity.


Assuntos
Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Receptores Nicotínicos/efeitos dos fármacos , Alcaloides/farmacologia , Animais , Autorradiografia , Azocinas/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/metabolismo , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Dopamina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Técnicas In Vitro , Camundongos , Camundongos Knockout , Neostriado/citologia , Neostriado/efeitos dos fármacos , Neostriado/metabolismo , Nicotina/administração & dosagem , Agonistas Nicotínicos/administração & dosagem , Bulbo Olfatório/citologia , Bulbo Olfatório/efeitos dos fármacos , Bulbo Olfatório/metabolismo , Piridinas/metabolismo , Piridinas/farmacologia , Quinolizinas/farmacologia , Ensaio Radioligante , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo
13.
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
14.
J Biol Chem ; 287(30): 25151-62, 2012 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-22665477

RESUMO

Genome-wide studies have strongly associated a non-synonymous polymorphism (rs16969968) that changes the 398th amino acid in the nAChR α5 subunit from aspartic acid to asparagine (D398N), with greater risk for increased nicotine consumption. We have used a pentameric concatemer approach to express defined and consistent populations of α3ß4α5 nAChR in Xenopus oocytes. α5(Asn-398; risk) variant incorporation reduces ACh-evoked function compared with inclusion of the common α5(Asp-398) variant without altering agonist or antagonist potencies. Unlinked α3, ß4, and α5 subunits assemble to form a uniform nAChR population with pharmacological properties matching those of concatemeric α3ß4* nAChRs. α5 subunit incorporation reduces α3ß4* nAChR function after coinjection with unlinked α3 and ß4 subunits but increases that of α3ß4α5 versus α3ß4-only concatemers. α5 subunit incorporation into α3ß4* nAChR also alters the relative efficacies of competitive agonists and changes the potency of the non-competitive antagonist mecamylamine. Additional observations indicated that in the absence of α5 subunits, free α3 and ß4 subunits form at least two further subtypes. The pharmacological profiles of these free subunit α3ß4-only subtypes are dissimilar both to each other and to those of α3ß4α5 nAChR. The α5 variant-induced change in α3ß4α5 nAChR function may underlie some of the phenotypic changes associated with this polymorphism.


Assuntos
Substituição de Aminoácidos , Complexos Multiproteicos/metabolismo , Mutação de Sentido Incorreto , Subunidades Proteicas/metabolismo , Receptores Nicotínicos/metabolismo , Transdução de Sinais/fisiologia , Animais , Estudo de Associação Genômica Ampla , Humanos , Complexos Multiproteicos/genética , Oócitos , Polimorfismo Genético , Subunidades Proteicas/genética , Receptores Nicotínicos/genética , Xenopus laevis
15.
Immunol Cell Biol ; 91(3): 195-200, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23399696

RESUMO

Nicotine is a potent inhibitor of the immune response and is protective against experimental autoimmune encephalomyelitis (EAE). Initial studies suggested that the cholinergic system modulates inflammation via the α7-nicotinic acetylcholine receptor (nAChR) subtype. We recently have shown that effector T cells and myeloid cells constitutively express mRNAs encoding nAChR α9 and ß2 subunits and found evidence for immune system roles for non-α7-nAChRs. In the present study, we assessed the effects of nAChR α9 or ß2 subunit gene deletion on EAE onset and severity, with or without nicotine treatment. We report again that disease onset is delayed and severity is attenuated in nicotine-treated, wild-type mice, an effect that also is observed in α9 subunit knock-out (KO) mice irrespective of nicotine treatment. On the other hand, ß2 KO mice fail to recover from peak measures of disease severity regardless of nicotine treatment, despite retaining sensitivity to nicotine's attenuation of disease severity. Prior to disease onset, we found significantly less reactive oxygen species production in the central nervous system (CNS) of ß2 KO mice, elevated proportions of CNS myeloid cells but decreased ratios of CNS macrophages/microglia in α9 or ß2 KO mice, and some changes in iNOS, TNF-α and IL-1ß mRNA levels in α9 KO and/or ß2 KO mice. Our data thus suggest that ß2*- and α9*-nAChRs, in addition to α7-nAChRs, have different roles in endogenous and nicotine-dependent modulation of immune functions and could be exploited as therapeutic targets to modulate inflammation and autoimmunity.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Macrófagos/imunologia , Microglia/imunologia , Receptores Nicotínicos/imunologia , Animais , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/genética , Deleção de Genes , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Macrófagos/patologia , Camundongos , Camundongos Knockout , Microglia/patologia , Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/imunologia , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Espécies Reativas de Oxigênio/imunologia , Receptores Nicotínicos/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
16.
J Med Chem ; 66(1): 306-332, 2023 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-36526469

RESUMO

Modifications of the cationic head and the ethylene linker of 2-(triethylammonium)ethyl ether of 4-stilbenol (MG624) have been proved to produce selective α9*-nAChR antagonism devoid of any effect on the α7-subtype. Here, single structural changes at the styryl portion of MG624 lead to prevailing α7-nAChR antagonism without abolishing α9*-nAChR antagonism. Nevertheless, rigidification of the styryl into an aromatic bicycle, better if including a H-bond donor NH, such as 5-indolyl (31), resulted in higher and more selective α7-nAChR affinity. Hybridization of this modification with the constraint of the 2-triethylammoniumethyloxy portion into (R)-N,N-dimethyl-3-pyrrolidiniumoxy substructure, previously reported as the best modification for the α7-nAChR affinity of MG624 (2), was a winning strategy. The resulting hybrid 33 had a subnanomolar α7-nAChR affinity and was a potent and selective α7-nAChR antagonist, producing at the α7-, but not at the α9*-nAChR, a profound loss of subsequent ACh function.


Assuntos
Receptores Nicotínicos , Éter , Receptor Nicotínico de Acetilcolina alfa7 , Etil-Éteres , Éteres
17.
J Neurosci ; 31(7): 2537-48, 2011 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-21325521

RESUMO

Diverse nicotinic acetylcholine receptor (nAChR) subtypes containing different subunit combinations can be placed on nerve terminals or soma/dendrites in the ventral tegmental area (VTA). nAChR α6 subunit message is abundant in the VTA, but α6*-nAChR cellular localization, function, pharmacology, and roles in cholinergic modulation of dopaminergic (DA) neurons within the VTA are not well understood. Here, we report evidence for α6ß2*-nAChR expression on GABA neuronal boutons terminating on VTA DA neurons. α-Conotoxin (α-Ctx) MII labeling coupled with immunocytochemical staining localizes putative α6*-nAChRs to presynaptic GABAergic boutons on acutely dissociated, rat VTA DA neurons. Functionally, acetylcholine (ACh) induces increases in the frequency of bicuculline-, picrotoxin-, and 4-aminopyridine-sensitive miniature IPSCs (mIPSCs) mediated by GABA(A) receptors. These increases are abolished by α6*-nAChR-selective α-Ctx MII or α-Ctx PIA (1 nm) but not by α7 (10 nm methyllycaconitine) or α4* (1 µm dihydro-ß-erythroidine)-nAChR-selective antagonists. ACh also fails to increase mIPSC frequency in VTA DA neurons prepared from nAChR ß2 knock-out mice. Moreover, ACh induces an α-Ctx PIA-sensitive elevation in intraterminal Ca(2+) in synaptosomes prepared from the rat VTA. Subchronic exposure to 500 nm nicotine reduces ACh-induced GABA release onto the VTA DA neurons, as does 10 d of systemic nicotine exposure. Collectively, these results indicate that α6ß2*-nAChRs are located on presynaptic GABAergic boutons within the VTA and modulate GABA release onto DA neurons. These presynaptic α6ß2*-nAChRs likely play important roles in nicotinic modulation of DA neuronal activity.


Assuntos
Dopamina/metabolismo , Neurônios/citologia , Terminações Pré-Sinápticas/fisiologia , Receptores Nicotínicos/metabolismo , Área Tegmentar Ventral/citologia , Ácido gama-Aminobutírico/metabolismo , 4-Aminopiridina/farmacologia , Acetilcolina/farmacologia , Aconitina/análogos & derivados , Aconitina/farmacologia , Animais , Animais Recém-Nascidos , Bungarotoxinas/farmacologia , Cálcio/metabolismo , Conotoxinas/metabolismo , Di-Hidro-beta-Eritroidina/farmacologia , Dopamina/farmacologia , Interações Medicamentosas , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Feminino , GABAérgicos/farmacologia , Glutamato Descarboxilase/metabolismo , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Neurônios/efeitos dos fármacos , Nicotina/farmacologia , Antagonistas Nicotínicos/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Terminações Pré-Sinápticas/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores Nicotínicos/deficiência , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo
18.
Mol Pharmacol ; 81(2): 175-88, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22039094

RESUMO

We investigated assembly and function of nicotinic acetylcholine receptors (nAChRs) composed of α7 and ß2 subunits. We measured optical and electrophysiological properties of wild-type and mutant subunits expressed in cell lines and Xenopus laevis oocytes. Laser scanning confocal microscopy indicated that fluorescently tagged α7 and ß2 subunits colocalize. Förster resonance energy transfer between fluorescently tagged subunits strongly suggested that α7 and ß2 subunits coassemble. Total internal reflection fluorescence microscopy revealed that assemblies localized to filopodia-like processes of SH-EP1 cells. Gain-of-function α7 and ß2 subunits confirmed that these subunits coassemble within functional receptors. Moreover, α7ß2 nAChRs composed of wild-type subunits or fluorescently tagged subunits had pharmacological properties similar to those of α7 nAChRs, although amplitudes of α7ß2 nAChR-mediated, agonist-evoked currents were generally ~2-fold lower than those for α7 nAChRs. It is noteworthy that α7ß2 nAChRs displayed sensitivity to low concentrations of the antagonist dihydro-ß-erythroidine that was not observed for α7 nAChRs at comparable concentrations. In addition, cysteine mutants revealed that the α7-ß2 subunit interface does not bind ligand in a functionally productive manner, partly explaining lower α7ß2 nAChR current amplitudes and challenges in identifying the function of native α7ß2 nAChRs. On the basis of our findings, we have constructed a model predicting receptor function that is based on stoichiometry and position of ß2 subunits within the α7ß2 nAChRs.


Assuntos
Fenômenos Eletrofisiológicos , Multimerização Proteica , Receptores Nicotínicos/fisiologia , Proteínas de Xenopus/fisiologia , Animais , Linhagem Celular , Eletrofisiologia , Oócitos , Ligação Proteica , Estrutura Quaternária de Proteína , Subunidades Proteicas , Receptores Nicotínicos/química , Receptores Nicotínicos/metabolismo , Proteínas de Xenopus/química , Proteínas de Xenopus/metabolismo , Xenopus laevis , Receptor Nicotínico de Acetilcolina alfa7
19.
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
20.
J Neurochem ; 122(1): 48-57, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22506481

RESUMO

Mouse superficial superior colliculus (SuSC) contains dense GABAergic innervation and diverse nicotinic acetylcholine receptor subtypes. Pharmacological and genetic approaches were used to investigate the subunit compositions of nicotinic acetylcholine receptors (nAChR) expressed on mouse SuSC GABAergic terminals. [(125) I]-Epibatidine competition-binding studies revealed that the α3ß2* and α6ß2* nicotinic subtype-selective peptide α-conotoxin MII-blocked binding to 40 ± 5% of SuSC nAChRs. Acetylcholine-evoked [(3) H]-GABA release from SuSC crude synaptosomal preparations is calcium dependent, blocked by the voltage-sensitive calcium channel blocker, cadmium, and the nAChR antagonist mecamylamine, but is unaffected by muscarinic, glutamatergic, P2X and 5-HT3 receptor antagonists. Approximately 50% of nAChR-mediated SuSC [(3) H]-GABA release is inhibited by α-conotoxin MII. However, the highly α6ß2*-subtype-selective α-conotoxin PIA did not affect [(3) H]-GABA release. Nicotinic subunit-null mutant mouse experiments revealed that ACh-stimulated SuSC [(3) H]-GABA release is entirely ß2 subunit-dependent. α4 subunit deletion decreased total function by >90%, and eliminated α-conotoxin MII-resistant release. ACh-stimulated SuSC [(3) H]-GABA release was unaffected by ß3, α5 or α6 nicotinic subunit deletions. Together, these data suggest that a significant proportion of mouse SuSC nicotinic agonist-evoked GABA-release is mediated by a novel, α-conotoxin MII-sensitive α3α4ß2 nAChR. The remaining α-conotoxin MII-resistant, nAChR agonist-evoked SuSC GABA release appears to be mediated via α4ß2* subtype nAChRs.


Assuntos
Conotoxinas/farmacologia , Antagonistas Nicotínicos/farmacologia , Receptores Nicotínicos/fisiologia , Colículos Superiores/efeitos dos fármacos , Colículos Superiores/metabolismo , Ácido gama-Aminobutírico/metabolismo , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Acetilcolina/farmacologia , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Animais , Compostos Bicíclicos Heterocíclicos com Pontes/farmacocinética , Bungarotoxinas/farmacologia , Maleato de Dizocilpina/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Antagonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Técnicas In Vitro , Isótopos de Iodo/farmacocinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Agonistas Nicotínicos/farmacocinética , Ligação Proteica/efeitos dos fármacos , Subunidades Proteicas/genética , Subunidades Proteicas/fisiologia , Piridinas/farmacocinética , Receptores Nicotínicos/deficiência , Antagonistas da Serotonina/farmacologia , Colículos Superiores/citologia , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo , Trítio/metabolismo , Tropanos/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA