Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
1.
J Neurosci ; 43(13): 2222-2241, 2023 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-36868853

RESUMO

Selective serotonin reuptake inhibitors (SSRIs) are the most prescribed treatment for individuals experiencing major depressive disorder. The therapeutic mechanisms that take place before, during, or after SSRIs bind the serotonin transporter (SERT) are poorly understood, partially because no studies exist on the cellular and subcellular pharmacokinetic properties of SSRIs in living cells. We studied escitalopram and fluoxetine using new intensity-based, drug-sensing fluorescent reporters targeted to the plasma membrane, cytoplasm, or endoplasmic reticulum (ER) of cultured neurons and mammalian cell lines. We also used chemical detection of drug within cells and phospholipid membranes. The drugs attain equilibrium in neuronal cytoplasm and ER at approximately the same concentration as the externally applied solution, with time constants of a few s (escitalopram) or 200-300 s (fluoxetine). Simultaneously, the drugs accumulate within lipid membranes by ≥18-fold (escitalopram) or 180-fold (fluoxetine), and possibly by much larger factors. Both drugs leave cytoplasm, lumen, and membranes just as quickly during washout. We synthesized membrane-impermeant quaternary amine derivatives of the two SSRIs. The quaternary derivatives are substantially excluded from membrane, cytoplasm, and ER for >2.4 h. They inhibit SERT transport-associated currents sixfold or 11-fold less potently than the SSRIs (escitalopram or fluoxetine derivative, respectively), providing useful probes for distinguishing compartmentalized SSRI effects. Although our measurements are orders of magnitude faster than the therapeutic lag of SSRIs, these data suggest that SSRI-SERT interactions within organelles or membranes may play roles during either the therapeutic effects or the antidepressant discontinuation syndrome.SIGNIFICANCE STATEMENT Selective serotonin reuptake inhibitors stabilize mood in several disorders. In general, these drugs bind to SERT, which clears serotonin from CNS and peripheral tissues. SERT ligands are effective and relatively safe; primary care practitioners often prescribe them. However, they have several side effects and require 2-6 weeks of continuous administration until they act effectively. How they work remains perplexing, contrasting with earlier assumptions that the therapeutic mechanism involves SERT inhibition followed by increased extracellular serotonin levels. This study establishes that two SERT ligands, fluoxetine and escitalopram, enter neurons within minutes, while simultaneously accumulating in many membranes. Such knowledge will motivate future research, hopefully revealing where and how SERT ligands engage their therapeutic target(s).


Assuntos
Transtorno Depressivo Maior , Inibidores Seletivos de Recaptação de Serotonina , Animais , Humanos , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Fluoxetina/farmacologia , Escitalopram , Serotonina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Retículo Endoplasmático/metabolismo , Citalopram/farmacologia , Mamíferos
2.
Mol Pharmacol ; 103(6): 339-347, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37001996

RESUMO

Study of α6ß4 nicotinic acetylcholine receptors (nAChRs) as a pharmacological target has recently gained interest because of their involvement in analgesia, control of catecholamine secretion, dopaminergic pathways, and aversive pathways. However, an extensive characterization of the human α6ß4 nAChRs has been vitiated by technical difficulties resulting in poor receptor expression. In 2020, Knowland and collaborators identified BARP (ß-anchoring and regulatory protein), a previously known voltage-gated calcium channel suppressor, as a novel human α6ß4 chaperone. Here, we establish that co-expression of human BARP with human α6ß4 in Xenopus oocytes, resulted in the functional expression of human α6ß4 receptors with acetylcholine-elicited currents that allow an in-depth characterization of the receptor using two electrode voltage-clamp electrophysiology together with diverse agonists and receptor mutations. We report: 1) an extended pharmacological characterization of the receptor, and 2) key residues for agonist-activity located in or near the first shell of the binding pocket. SIGNIFICANCE STATEMENT: The human α6ß4 nicotinic acetylcholine receptor has attained increased interest because of its involvement in diverse physiological processes and diseases. Although recognized as a pharmacological target, development of specific agonists has been hampered by limited knowledge of its structural characteristics and by challenges in expressing the receptor. By including the chaperone ß-anchoring and regulatory protein for enhanced expression and employing different ligands, we have studied the pharmacology of α6ß4, providing insight into receptor residues and structural requirements for ligands important to consider for agonist-induced activation.


Assuntos
Receptores Nicotínicos , Humanos , Animais , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Ligantes , Sítios de Ligação , Acetilcolina/farmacologia , Acetilcolina/metabolismo , Domínios Proteicos , Oócitos/metabolismo , Xenopus laevis/metabolismo
3.
J Am Chem Soc ; 144(35): 16101-16117, 2022 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-36006801

RESUMO

Nicotinic acetylcholine receptors (nAChRs) play an important role in neurotransmission and are also involved in addiction and several disease states. There is significant interest in therapeutic targeting of nAChRs; however, achieving selectivity for one subtype over others has been a longstanding challenge, given the close structural similarities across the family. Here, we characterize binding interactions in the α3ß4 nAChR subtype via structure-function studies involving noncanonical amino acid mutagenesis and two-electrode voltage clamp electrophysiology. We establish comprehensive binding models for both the endogenous neurotransmitter ACh and the smoking cessation drug cytisine. We also use a panel of C(10)-substituted cytisine derivatives to probe the effects of subtle changes in the ligand structure on binding. By comparing our results to those obtained for the well-studied α4ß2 subtype, we identify several features of both the receptor and agonist structure that can be utilized to enhance selectivity for either α3ß4 or α4ß2. Finally, we characterize binding interactions of the α3ß4-selective partial agonist AT-1001 to determine factors that contribute to its selectivity. These results shed new light on the design of selective nAChR-targeted ligands and can be used to inform the design of improved therapies with minimized off-target effects.


Assuntos
Agonistas Nicotínicos , Receptores Nicotínicos , Sítios de Ligação , Ligantes , Agonistas Nicotínicos/química , Receptores Nicotínicos/química
4.
J Am Chem Soc ; 144(19): 8480-8486, 2022 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-35446570

RESUMO

We report a reagentless, intensity-based S-methadone fluorescent sensor, iS-methadoneSnFR, consisting of a circularly permuted GFP inserted within the sequence of a mutated bacterial periplasmic binding protein (PBP). We evolved a previously reported nicotine-binding PBP to become a selective S-methadone-binding sensor, via three mutations in the PBP's second shell and hinge regions. iS-methadoneSnFR displays the necessary sensitivity, kinetics, and selectivity─notably enantioselectivity against R-methadone─for biological applications. Robust iS-methadoneSnFR responses in human sweat and saliva and mouse serum enable diagnostic uses. Expression and imaging in mammalian cells demonstrate that S-methadone enters at least two organelles and undergoes acid trapping in the Golgi apparatus, where opioid receptors can signal. This work shows a straightforward strategy in adapting existing PBPs to serve real-time applications ranging from subcellular to personal pharmacokinetics.


Assuntos
Agonistas Nicotínicos , Proteínas Periplásmicas de Ligação , Animais , Mamíferos/metabolismo , Metadona , Camundongos , Mutação , Organelas/metabolismo
5.
Mol Pharmacol ; 96(2): 212-218, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31175182

RESUMO

The α7 nicotinic acetylcholine receptor (nAChR) is a potential drug target for the treatment of a number of neurologic and inflammatory disorders. Silent agonists are an emerging class of drugs that bind to the receptor but do not open the channel. Instead they shift the receptor to a desensitized state. Silent agonists may be able to target a subset of α7 nAChR-mediated signaling processes. Here we use noncanonical amino acid mutagenesis to characterize the binding to α7 by the silent agonist 1,4-diazabicyclo[3.2.2]nonan-4-yl(5-(3-(trifluoromethyl)phenyl)furan-2-yl)methanone (NS6740). We find that, like α7 agonists, NS6740 forms a cation-π interaction with Y115 (TyrA). We also showed that NS6740 makes a novel hydrogen bond to TyrA. This interaction is necessary for the silent agonist activity of NS6740; when the hydrogen bond is blocked, silent agonist NS6740 converts to a conventional partial agonist and appreciably opens the channel in the absence of a positive allosteric modulator (EC50 150 nM). SIGNIFICANCE STATEMENT: Noncanonical amino acids were used to show that a hydrogen bond to tyrosine (Y115) is required for silent agonist activity of NS6740 at the α7 nicotinic acetylcholine receptor.


Assuntos
Compostos Azabicíclicos/farmacologia , Proteínas de Bactérias/metabolismo , Furanos/farmacologia , Complexos Multienzimáticos/metabolismo , Mutação , Receptor Nicotínico de Acetilcolina alfa7/química , Regulação Alostérica , Animais , Compostos Azabicíclicos/química , Proteínas de Bactérias/química , Furanos/química , Ligação de Hidrogênio , Complexos Multienzimáticos/química , Ligação Proteica , Ratos , Xenopus laevis , Receptor Nicotínico de Acetilcolina alfa7/agonistas , Receptor Nicotínico de Acetilcolina alfa7/genética
6.
J Biol Chem ; 293(8): 2903-2914, 2018 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-29298898

RESUMO

Cys-loop receptors are pentameric ligand-gated ion channels that facilitate communication within the nervous system. Upon neurotransmitter binding, these receptors undergo an allosteric activation mechanism connecting the binding event to the membrane-spanning channel pore, which expands to conduct ions. Some of the earliest steps in this activation mechanism are carried out by residues proximal to the binding site, the relative positioning of which may reflect functional differences among members of the Cys-loop family of receptors. Herein, we investigated key side-chain interactions near the binding site via mutagenesis and two-electrode voltage-clamp electrophysiology in serotonin-gated 5-HT3A receptors (5-HT3ARs) and nicotinic acetylcholine receptors (nAChRs) expressed in Xenopus laevis oocytes. We found that a triad of residues aligning to Thr-152, Glu-209, and Lys-211 in the 5-HT3AR can be exchanged between the homomeric 5-HT3AR and the muscle-type nAChR α-subunit with small functional consequences. Via triple mutant cycle analysis, we demonstrated that this triad forms an interdependent network in the muscle-type nAChR. Furthermore, nAChR-type mutations of the 5-HT3AR affect the affinity of nicotine, a competitive antagonist of 5-HT3ARs, in a cooperative manner. Using mutant cycle analyses between the 5-HT3A triad, loop A residues Asn-101 and Glu-102, ß9 residue Lys-197, and the channel gate at Thr-257, we observed that residues in this region are energetically linked to the channel gate and are particularly sensitive to mutations that introduce a net positive charge. This study expands our understanding of the differences and similarities in the activation mechanisms of Cys-loop receptors.


Assuntos
Modelos Moleculares , Receptores Nicotínicos/metabolismo , Receptores 5-HT3 de Serotonina/metabolismo , Acetilcolina/química , Acetilcolina/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sítios de Ligação , Ligação Competitiva , Agonistas Colinérgicos/química , Agonistas Colinérgicos/metabolismo , Humanos , Cinética , Ligantes , Camundongos , Mutagênese Sítio-Dirigida , Mutação , Nicotina/química , Nicotina/metabolismo , Antagonistas Nicotínicos/química , Antagonistas Nicotínicos/metabolismo , Conformação Proteica , Receptores Nicotínicos/química , Receptores Nicotínicos/genética , Receptores 5-HT3 de Serotonina/química , Receptores 5-HT3 de Serotonina/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência
7.
J Am Chem Soc ; 141(40): 15840-15849, 2019 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-31518499

RESUMO

Nicotinic acetylcholine receptors (nAChRs) are crucial for communication between synapses in the central nervous system. As such, they are also implicated in several neuropsychiatric and addictive diseases. Cytisine is a partial agonist of some nAChRs and has been used for smoking cessation. Previous studies have established a binding model for several agonists to several nAChR subtypes. Here, we evaluate the extent to which this model applies to cytisine at the α4ß2 nAChR, which is a subtype that is known to play a prominent role in nicotine addiction. Along with the commonly seen cation-π interaction and two hydrogen bonds, we find that cytisine makes a second cation-π interaction at the agonist binding site. We also evaluated a series of C(10)-substituted cytisine derivatives, using two-electrode voltage-clamp electrophysiology and noncanonical amino acid mutagenesis. Double-mutant cycle analyses revealed that C(10) substitution generally strengthens the newly established second cation-π interaction, while it weakens the hydrogen bond typically seen to LeuE in the complementary subunit. The results suggest a model for how cytisine derivatives substituted at C(10) (as well as C(9)/C(10)) adjust their binding orientation, in response to pyridone ring substitution.


Assuntos
Alcaloides/química , Agonistas Nicotínicos/química , Receptores Nicotínicos/química , Alcaloides/genética , Animais , Azocinas/química , Sítios de Ligação , Relação Dose-Resposta a Droga , Eletrofisiologia , Ligação de Hidrogênio , Estrutura Molecular , Mutagênese Sítio-Dirigida , Mutação , Oócitos/metabolismo , Técnicas de Patch-Clamp , Ligação Proteica , Quinolizinas/química , Ratos , Receptores Nicotínicos/genética , Xenopus laevis
8.
Biochemistry ; 57(27): 4036-4043, 2018 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-29927250

RESUMO

Erwinia ligand-gated ion channel (ELIC) is a bacterial homologue of vertebrate pentameric ligand-gated ion channels (pLGICs) and has proven to be a valuable model for understanding the structure and function of this important protein family. There is nevertheless still a question about whether molecular details can be accurately extrapolated from this protein to those found in eukaryotes. Here we explore the role of proline residues (Pros) in ELIC by creating mutant receptors, expressing them in Xenopus laevis oocytes, and using whole-cell voltage-clamp electrophysiology to monitor channel activity. In contrast to eukaryotic pLGICs, proline-to-alanine (Pro-to-Ala) substitution in ELIC mostly resulted in gain of function, and even altering highly conserved Pro residues in M1 and the M2-M3 loop did not ablate function. These substitutions also mostly resulted in ablation of the modulation by Ca2+ observed in wild-type receptors. Substitution of the Pro in the "Cys loop", however, did result in nonfunctional receptors. Probing this residue with noncanonical amino acids revealed a requirement for a substituted amine at this position, as well as a general preference for Pro analogues with greater intrinsic cis biases. We propose there is likely a cis bond at the apex of the Cys loop in this protein, which is consistent with some, but not all, findings from other pLGICs. Overall, the data show that the roles of proline residues are less critical in ELIC than in other pLGICs, supporting other studies that suggest caution must be applied in using data from this prokaryotic receptor to understand molecular details of eukaryotic pLGIC receptor function.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Erwinia/química , Erwinia/metabolismo , Canais Iônicos de Abertura Ativada por Ligante/química , Canais Iônicos de Abertura Ativada por Ligante/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Proteínas de Bactérias/genética , Erwinia/genética , Canais Iônicos de Abertura Ativada por Ligante/genética , Modelos Moleculares , Prolina/química , Prolina/genética , Prolina/metabolismo , Conformação Proteica , Alinhamento de Sequência , Xenopus
9.
Biochemistry ; 56(13): 1836-1840, 2017 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-28287260

RESUMO

Designing subtype-selective agonists for neuronal nicotinic acetylcholine receptors is a challenging and significant goal aided by intricate knowledge of each subtype's binding patterns. We previously reported that in α6ß2 receptors, acetylcholine makes a functional cation-π interaction with Trp149, but nicotine and TC299423 do not, suggesting a distinctive binding site. This work explores hydrogen binding at the backbone carbonyl associated with α6ß2 Trp149. Substituting residue i + 1, Thr150, with its α-hydroxy analogue (Tah) attenuates the carbonyl's hydrogen bond accepting ability. At α6(T150Tah)ß2, nicotine shows a 24-fold loss of function, TC299423 shows a modest loss, and acetylcholine shows no effect. Nicotine was further analyzed via a double-mutant cycle analysis utilizing N'-methylnicotinium, which indicated a hydrogen bond in α6ß2 with a ΔΔG of 2.6 kcal/mol. Thus, even though nicotine does not make the conserved cation-π interaction with Trp149, it still makes a functional hydrogen bond to its associated backbone carbonyl.


Assuntos
Acetilcolina/química , Nicotina/análogos & derivados , Agonistas Nicotínicos/química , Receptores Nicotínicos/química , Acetilcolina/farmacologia , Animais , Expressão Gênica , Ligação de Hidrogênio , Mutagênese Sítio-Dirigida , Mutação , Nicotina/química , Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Técnicas de Patch-Clamp , Ligação Proteica , Ratos , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Termodinâmica , Triptofano/química , Triptofano/metabolismo , Xenopus laevis
10.
J Biol Chem ; 291(12): 6272-80, 2016 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-26668320

RESUMO

Gloeobacter violaceus ligand-gated ion channel (GLIC) has served as a valuable structural and functional model for the eukaryotic Cys-loop receptor superfamily. In Cys-loop and other receptors, we have previously demonstrated the crucial roles played by several conserved prolines. Here we explore the role of prolines in the gating transitions of GLIC. As conventional substitutions at some positions resulted in nonfunctional proteins, we used in vivo non-canonical amino acid mutagenesis to determine the specific structural requirements at these sites. Receptors were expressed heterologously in Xenopus laevis oocytes, and whole-cell electrophysiology was used to monitor channel activity. Pro-119 in the Cys-loop, Pro-198 and Pro-203 in the M1 helix, and Pro-299 in the M4 helix were sensitive to substitution, and distinct roles in receptor activity were revealed for each. In the context of the available structural data for GLIC, the behaviors of Pro-119, Pro-203, and Pro-299 mutants are consistent with earlier proline mutagenesis work. However, the Pro-198 site displays a unique phenotype that gives evidence of the importance of the region surrounding this residue for the correct functioning of GLIC.


Assuntos
Proteínas de Bactérias/química , Cianobactérias , Canais Iônicos de Abertura Ativada por Ligante/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Substituição de Aminoácidos , Ligação de Hidrogênio , Ativação do Canal Iônico , Modelos Moleculares , Dados de Sequência Molecular , Prolina , Estrutura Secundária de Proteína
11.
J Am Chem Soc ; 139(13): 4655-4658, 2017 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-28324654

RESUMO

Visible-light triggered quinone trimethyl locks are reported as a general design for long-wavelength photoremovable protecting groups for alcohols and amines. Intramolecular photoreduction unmasks a reactive phenol that undergoes fast lactonization to release alcohols and amines. Model substrates are released in quantitative yield along with well-defined, colorless hydroquinone byproducts. Substituent modifications of the quinone core allow absorption from 400 to 600 nm.

12.
J Am Chem Soc ; 139(13): 4729-4736, 2017 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-28199106

RESUMO

Mechanistic studies of a general reaction that decages a wide range of substrates on exposure to visible light are described. The reaction involves a photochemically initiated reduction of a quinone mediated by an appended thioether. After reduction, a trimethyl lock system incorporated into the quinone leads to thermal decaging. The reaction could be viewed as an electron-transfer initiated reduction of the quinone or as a hydrogen abstraction-Norrish Type II-reaction. Product analysis, kinetic isotope effects, stereochemical labeling, radical clock, and transient absorption studies support the electron transfer mechanism. The differing reactivities of the singlet and triplet states are determined, and the ways in which this process deviates from typical quinone photochemistry are discussed. The mechanism suggests strategies for extending the reaction to longer wavelengths that would be of interest for applications in chemical biology and in a therapeutic setting.

13.
Chembiochem ; 17(14): 1323-7, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27123791

RESUMO

We describe the reversible photoactivation of the acid sensitive ligand-gated ion channel ASIC2a, a mammalian channel found throughout the central and peripheral nervous systems that is associated with vision and pain. We also show the activation of GLIC, an acid-sensitive prokaryotic homologue of the nicotinic acetylcholine receptor. Photoactivation was achieved by using visible light irradiation of a newly synthesized water-soluble merocyanine photoacid, 1, which was designed to remove adverse channel blocking effects of a related system. Activation of ASIC2a and GLIC occurs reversibly, in a benign manner, and only upon irradiation. Further studies using transient absorption spectroscopy showed that protonation of a colorimetric base occurred rapidly (ca. 100 µs) after excitation of 1. These results demonstrate that irradiation of 1 can induce rapid, local pH changes that can be used to investigate both biological and chemical proton transfer reactions.


Assuntos
Canais Iônicos Sensíveis a Ácido/efeitos da radiação , Luz , Canais Iônicos Sensíveis a Ácido/fisiologia , Animais , Humanos , Concentração de Íons de Hidrogênio , Ativação do Canal Iônico , Ligantes , Dor/fisiopatologia , Receptores Nicotínicos/efeitos da radiação , Visão Ocular/fisiologia
14.
Phys Chem Chem Phys ; 17(43): 29262-70, 2015 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-26467787

RESUMO

Cation-π interactions are common in biological systems, and many structural studies have revealed the aromatic box as a common motif. With the aim of understanding the nature of the aromatic box, several computational methods were evaluated for their ability to reproduce experimental cation-π binding energies. We find the DFT method M06 with the 6-31G(d,p) basis set performs best of several methods tested. The binding of benzene to a number of different cations (sodium, potassium, ammonium, tetramethylammonium, and guanidinium) was studied. In addition, the binding of the organic cations NH4(+) and NMe4(+) to ab initio generated aromatic boxes as well as examples of aromatic boxes from protein crystal structures were investigated. These data, along with a study of the distance dependence of the cation-π interaction, indicate that multiple aromatic residues can meaningfully contribute to cation binding, even with displacements of more than an angstrom from the optimal cation-π interaction. Progressive fluorination of benzene and indole was studied as well, and binding energies obtained were used to reaffirm the validity of the "fluorination strategy" to study cation-π interactions in vivo.


Assuntos
Benzeno/química , Modelos Moleculares , Compostos de Amônio/química , Cátions/química , Guanidina/química , Halogenação , Potássio/química , Sódio/química , Termodinâmica
15.
Nature ; 458(7237): 534-7, 2009 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-19252481

RESUMO

Nicotine addiction begins with high-affinity binding of nicotine to acetylcholine (ACh) receptors in the brain. The end result is over 4,000,000 smoking-related deaths annually worldwide and the largest source of preventable mortality in developed countries. Stress reduction, pleasure, improved cognition and other central nervous system effects are strongly associated with smoking. However, if nicotine activated ACh receptors found in muscle as potently as it does brain ACh receptors, smoking would cause intolerable and perhaps fatal muscle contractions. Despite extensive pharmacological, functional and structural studies of ACh receptors, the basis for the differential action of nicotine on brain compared with muscle ACh receptors has not been determined. Here we show that at the alpha4beta2 brain receptors thought to underlie nicotine addiction, the high affinity for nicotine is the result of a strong cation-pi interaction to a specific aromatic amino acid of the receptor, TrpB. In contrast, the low affinity for nicotine at the muscle-type ACh receptor is largely due to the fact that this key interaction is absent, even though the immediate binding site residues, including the key amino acid TrpB, are identical in the brain and muscle receptors. At the same time a hydrogen bond from nicotine to the backbone carbonyl of TrpB is enhanced in the neuronal receptor relative to the muscle type. A point mutation near TrpB that differentiates alpha4beta2 and muscle-type receptors seems to influence the shape of the binding site, allowing nicotine to interact more strongly with TrpB in the neuronal receptor. ACh receptors are established therapeutic targets for Alzheimer's disease, schizophrenia, Parkinson's disease, smoking cessation, pain, attention-deficit hyperactivity disorder, epilepsy, autism and depression. Along with solving a chemical mystery in nicotine addiction, our results provide guidance for efforts to develop drugs that target specific types of nicotinic receptors.


Assuntos
Encéfalo/metabolismo , Nicotina/metabolismo , Receptores Nicotínicos/metabolismo , Acetilcolina/química , Acetilcolina/metabolismo , Animais , Sítios de Ligação , Cátions/metabolismo , Halogenação , Camundongos , Modelos Moleculares , Nicotina/química , Agonistas Nicotínicos/metabolismo , Oócitos/metabolismo , Especificidade de Órgãos , Ligação Proteica , Conformação Proteica , Ratos , Receptores Nicotínicos/química , Receptores Nicotínicos/genética , Fumar/efeitos adversos , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Triptofano/química , Triptofano/metabolismo , Xenopus laevis
16.
Proc Natl Acad Sci U S A ; 109(44): E3028-34, 2012 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-23035248

RESUMO

GABA(A) receptors are pentameric ligand-gated ion channels involved in fast inhibitory neurotransmission and are allosterically modulated by the anxiolytic, anticonvulsant, and sedative-hypnotic benzodiazepines. Here we show that the prokaryotic homolog ELIC also is activated by GABA and is modulated by benzodiazepines with effects comparable to those at GABA(A) receptors. Crystal structures reveal important features of GABA recognition and indicate that benzodiazepines, depending on their concentration, occupy two possible sites in ELIC. An intrasubunit site is adjacent to the GABA-recognition site but faces the channel vestibule. A second intersubunit site partially overlaps with the GABA site and likely corresponds to a low-affinity benzodiazepine-binding site in GABA(A) receptors that mediates inhibitory effects of the benzodiazepine flurazepam. Our study offers a structural view how GABA and benzodiazepines are recognized at a GABA-activated ion channel.


Assuntos
Benzodiazepinas/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Canais Iônicos/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Benzodiazepinas/metabolismo , Sítios de Ligação , Biopolímeros , Cristalografia por Raios X , Canais Iônicos/química , Ligantes , Modelos Moleculares , Receptores de GABA-A/metabolismo , Xenopus
17.
Mol Pharmacol ; 86(3): 263-74, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24966348

RESUMO

P2X receptors and nicotinic acetylcholine receptors (nAChRs) display functional and physical interactions in many cell types and heterologous expression systems, but interactions between α6ß4-containing (α6ß4*) nAChRs and P2X2 receptors and/or P2X3 receptors have not been fully characterized. We measured several types of crosstalk in oocytes coexpressing α6ß4 nAChRs and P2X2, P2X3, or P2X2/3 receptors. A novel form of crosstalk occurs between α6ß4 nAChRs and P2X2 receptors. P2X2 receptors were forced into a prolonged desensitized state upon activation by ATP through a mechanism that does not depend on the intracellular C terminus of the P2X2 receptors. Coexpression of α6ß4 nAChRs with P2X3 receptors shifts the ATP dose-response relation to the right, even in the absence of acetylcholine (ACh). Moreover, currents become nonadditive when ACh and ATP are coapplied, as previously reported for other Cys-loop receptors interacting with P2X receptors, and this crosstalk is dependent on the presence of the P2X3 C-terminal domain. P2X2 receptors also functionally interact with α6ß4ß3 but through a different mechanism from α6ß4. The interaction with P2X3 receptors is less pronounced for the α6ß4ß3 nAChR than the α6ß4 nAChR. We also measured a functional interaction between the α6ß4 nAChRs and the heteromeric P2X2/3 receptor. Experiments with the nAChR channel blocker mecamylamine on P2X2-α6ß4 oocytes point to the loss of P2X2 channel activity during the crosstalk, whereas the ion channel pores of the P2X receptors were fully functional and unaltered by the receptor interaction for P2X2-α6ß4ß3, P2X2/3-α6ß4, and P2X2/3-α6ß4ß3. These results may be relevant to dorsal root ganglion cells and to other neurons that coexpress these receptor subunits.


Assuntos
Subunidades Proteicas/metabolismo , Receptores Nicotínicos/metabolismo , Receptores Purinérgicos P2X/metabolismo , Acetilcolina/farmacologia , Trifosfato de Adenosina/farmacologia , Animais , Feminino , Mecamilamina/farmacologia , Camundongos , Agonistas Nicotínicos/farmacologia , Antagonistas Nicotínicos/farmacologia , Oócitos/metabolismo , Multimerização Proteica , Agonistas do Receptor Purinérgico P2X/farmacologia , Ratos , Receptor Cross-Talk , Receptores Purinérgicos P2X2/metabolismo , Receptores Purinérgicos P2X3/metabolismo , Xenopus laevis
18.
J Biol Chem ; 288(10): 6991-7, 2013 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-23349463

RESUMO

The agonist-binding site of nicotinic acetylcholine receptors (nAChRs) spans an interface between two subunits of the pentameric receptor. The principal component of this binding site is contributed by an α subunit, and it binds the cationic moiety of the nicotinic pharmacophore. The other part of the pharmacophore, a hydrogen bond acceptor, has recently been shown to bind to the complementary non-α subunit via the backbone NH of a conserved Leu. This interaction was predicted by studies of ACh-binding proteins and confirmed by functional studies of the neuronal (CNS) nAChR, α4ß2. The ACh-binding protein structures further suggested that the hydrogen bond to the backbone NH is mediated by a water molecule and that a second hydrogen bonding interaction occurs between the water molecule and the backbone CO of a conserved Asn, also on the non-α subunit. Here, we provide new insights into the nature of the interactions between the hydrogen bond acceptor of nicotinic agonists and the complementary subunit backbone. We studied both the nAChR of the neuromuscular junction (muscle-type) and a neuronal subtype, (α4)2(ß4)3. In the muscle-type receptor, both ACh and nicotine showed a strong interaction with the Leu NH, but the potent nicotine analog epibatidine did not. This interaction was much attenuated in the α4ß4 receptor. Surprisingly, we found no evidence for a functionally significant interaction with the backbone carbonyl of the relevant Asn in either receptor with an array of agonists.


Assuntos
Acetilcolina/metabolismo , Nicotina/metabolismo , Receptores Nicotínicos/metabolismo , Acetilcolina/química , Acetilcolina/farmacologia , Sequência de Aminoácidos , Animais , Asparagina/química , Asparagina/genética , Asparagina/metabolismo , Sítios de Ligação/genética , Compostos Bicíclicos Heterocíclicos com Pontes/química , Compostos Bicíclicos Heterocíclicos com Pontes/metabolismo , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Feminino , Humanos , Ligação de Hidrogênio , Leucina/química , Leucina/genética , Leucina/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Mutação , Nicotina/química , Nicotina/farmacologia , Oócitos/metabolismo , Oócitos/fisiologia , Técnicas de Patch-Clamp , Ligação Proteica , Piridinas/química , Piridinas/metabolismo , Piridinas/farmacologia , Receptores Nicotínicos/química , Receptores Nicotínicos/genética , Homologia de Sequência de Aminoácidos , Xenopus laevis
19.
Chembiochem ; 15(12): 1710-20, 2014 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-24990307

RESUMO

We describe a strategy for incorporating non-canonical amino acids site-specifically into proteins expressed in living cells, involving organic synthesis to chemically aminoacylate a suppressor tRNA, protein expression in Xenopus oocytes, and monitoring protein function, primarily by electrophysiology. With this protocol, a very wide range of non-canonical amino acids can be employed, allowing both systematic structure-function studies and the incorporation of reactive functionalities. Here, we present an overview of the methodology and examples meant to illustrate the versatility and power of the method as a tool for investigating protein structure and function.


Assuntos
Aminoácidos/química , Aminoácidos/metabolismo , Proteínas/química , Proteínas/metabolismo , Aminoacil-RNA de Transferência/química , Aminoacilação de RNA de Transferência , Animais , Humanos , Aminoacil-RNA de Transferência/metabolismo
20.
Acc Chem Res ; 46(4): 885-93, 2013 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-23214924

RESUMO

The chemistry community now recognizes the cation-π interaction as a major force for molecular recognition, joining the hydrophobic effect, the hydrogen bond, and the ion pair in determining macromolecular structure and drug-receptor interactions. This Account provides the author's perspective on the intellectual origins and fundamental nature of the cation-π interaction. Early studies on cyclophanes established that water-soluble, cationic molecules would forego aqueous solvation to enter a hydrophobic cavity if that cavity was lined with π systems. Important gas phase studies established the fundamental nature of the cation-π interaction. The strength of the cation-π interaction (Li(+) binds to benzene with 38 kcal/mol of binding energy; NH4(+) with 19 kcal/mol) distinguishes it from the weaker polar-π interactions observed in the benzene dimer or water-benzene complexes. In addition to the substantial intrinsic strength of the cation-π interaction in gas phase studies, the cation-π interaction remains energetically significant in aqueous media and under biological conditions. Many studies have shown that cation-π interactions can enhance binding energies by 2-5 kcal/mol, making them competitive with hydrogen bonds and ion pairs in drug-receptor and protein-protein interactions. As with other noncovalent interactions involving aromatic systems, the cation-π interaction includes a substantial electrostatic component. The six (four) C(δ-)-H(δ+) bond dipoles of a molecule like benzene (ethylene) combine to produce a region of negative electrostatic potential on the face of the π system. Simple electrostatics facilitate a natural attraction of cations to the surface. The trend for (gas phase) binding energies is Li(+) > Na(+) > K(+) > Rb(+): as the ion gets larger the charge is dispersed over a larger sphere and binding interactions weaken, a classical electrostatic effect. On other hand, polarizability does not define these interactions. Cyclohexane is more polarizable than benzene but a decidedly poorer cation binder. Many studies have documented cation-π interactions in protein structures, where lysine or arginine side chains interact with phenylalanine, tyrosine, or tryptophan. In addition, countless studies have established the importance of the cation-π interaction in a range of biological processes. Our work has focused on molecular neurobiology, and we have shown that neurotransmitters generally use a cation-π interaction to bind to their receptors. We have also shown that many drug-receptor interactions involve cation-π interactions. A cation-π interaction plays a critical role in the binding of nicotine to ACh receptors in the brain, an especially significant case. Other researchers have established important cation-π interactions in the recognition of the "histone code," in terpene biosynthesis, in chemical catalysis, and in many other systems.


Assuntos
Cátions/química , Ânions , Benzeno/química , Carbono/química , Hidrogênio/química , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Fenilalanina/química , Proteínas/química , Eletricidade Estática , Triptofano/química , Água/química
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa