Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; : 107778, 2024 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-39270821

RESUMO

Propofol is a widely used anesthetic and sedative that acts as a positive allosteric modulator (PAM) of gamma-aminobutyric acid type A (GABAA) receptors. Several potential propofol binding sites that may mediate this effect have been identified using propofol-analogue photoaffinity labeling. o-PD labels ß-H267, a pore-lining residue, whereas AziPm labels residues ß-M286, ß-M227 and α-I239 in the two membrane-facing interfaces (ß(+)/α(-) and α(+)/ß(-)) between α and ß subunits. This study used photoaffinity labeling of α1ß3 GABAA receptors to reconcile the apparently conflicting results obtained with AziPm and o-PD labeling, focusing on whether ß3-H267 identifies specific propofol binding site(s). The results show that propofol, but not AziPm protects ß3-H267 from labeling by o-PD, whereas both propofol and o-PD protect against AziPm labeling of ß3-M286, ß3-M227 and α1I239. These data indicate that there are three distinct classes of propofol binding sites, with AziPm binding to two of the classes and o-PD to all three. Analysis of binding stoichiometry using native mass spectrometry in ß3 homomeric receptors, demonstrated a minimum of five AziPm labeled residues and three o-PD labeled residues per pentamer, suggesting that there are two distinct propofol binding sites per ß-subunit. The native MS data, coupled with photolabeling performed in the presence of zinc, indicate that the binding site(s) identified by o-PD are adjacent to, but not within the channel pore, since the pore at the 17' H267 residue can accommodate only one propofol molecule. These data validate the existence of three classes of specific propofol binding sites on α1ß3 GABAA receptors.

2.
Mol Pharmacol ; 2024 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-39214710

RESUMO

The γ-aminobutyric acid type A (GABAA) receptor is modulated by a number of neuroactive steroids. Sulfated steroids and 3ß-hydroxy steroids inhibit while 3α-hydroxy steroids typically potentiate the receptor. Here, we have investigated inhibition of the α1ß3γ2L GABAA receptor by the endogenous neurosteroid 3α-hydroxy-5ß-pregnan-20-one (3α5ßP) and the synthetic neuroactive steroid 3α-hydroxy-5α-androstane-17ß-carbonitrile (ACN). The receptors were expressed in Xenopus oocytes. All experiments were done using two-electrode voltage-clamp electrophysiology. In the presence of low concentrations of GABA, 3α5ßP and ACN potentiate the GABAA receptor. To reveal inhibition, we conducted the experiments on receptors activated by the combination of a saturating concentration of GABA and propofol to fully activate the receptors and mask potentiation, or on mutant receptors in which potentiation is ablated. Under these conditions, both steroids inhibited the receptor with IC50s of ~13 µM and maximal inhibitory effects of 70-90%. Receptor inhibition by 3α5ßP was sensitive to substitution of the α1TM2-2' residue, previously shown to ablate inhibition by pregnenolone sulfate. However, results of coapplication studies and the apparent lack of state dependence suggest that pregnenolone sulfate and 3α5ßP inhibit the GABAA receptor independently and through distinct mechanisms. Mutations to the neurosteroid binding sites in the α1 and ß3 subunits significantly, albeit weakly and incompletely, reduced inhibition by 3α5ßP and ACN. Significance Statement The heteromeric GABAA receptor is inhibited by sulfated steroids and 3ß-hydroxy steroids while 3α-hydroxy steroids are considered to potentiate the receptor. We show here that 3α-hydroxy steroids have inhibitory effects on the α1ß3γ2L receptor, which are observed in specific experimental settings and are expected to manifest under different physiological conditions.

3.
J Anesth ; 38(2): 261-274, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38252143

RESUMO

Neurosteroids (NS) are a class of steroids that are synthesized within the central nervous system (CNS). Various NS can either enhance or inhibit CNS excitability and they play important biological roles in brain development, brain function and as mediators of mood. One class of NS, 3α-hydroxy-pregnane steroids such as allopregnanolone (AlloP) or pregnanolone (Preg), inhibits neuronal excitability; these endogenous NS and their analogues have been therapeutically applied as anti-depressants, anti-epileptics and general anesthetics. While NS have many favorable properties as anesthetics (e.g. rapid onset, rapid recovery, minimal cardiorespiratory depression, neuroprotection), they are not currently in clinical use, largely due to problems with formulation. Recent advances in understanding NS mechanisms of action and improved formulations have rekindled interest in development of NS as sedatives and anesthetics. In this review, the synthesis of NS, and their mechanism of action will be reviewed with specific emphasis on their binding sites and actions on γ-aminobutyric acid type A (GABAA) receptors. The potential advantages of NS analogues as sedative and anesthetic agents will be discussed.


Assuntos
Anestésicos Gerais , Anestésicos , Neuroesteroides , Anestésicos Gerais/efeitos adversos , Anestésicos/efeitos adversos , Pregnanolona/farmacologia , Ácido gama-Aminobutírico , Receptores de GABA-A
4.
PLoS Biol ; 17(3): e3000157, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30845142

RESUMO

Neurosteroids are endogenous modulators of neuronal excitability and nervous system development and are being developed as anesthetic agents and treatments for psychiatric diseases. While gamma amino-butyric acid Type A (GABAA) receptors are the primary molecular targets of neurosteroid action, the structural details of neurosteroid binding to these proteins remain ill defined. We synthesized neurosteroid analogue photolabeling reagents in which the photolabeling groups were placed at three positions around the neurosteroid ring structure, enabling identification of binding sites and mapping of neurosteroid orientation within these sites. Using middle-down mass spectrometry (MS), we identified three clusters of photolabeled residues representing three distinct neurosteroid binding sites in the human α1ß3 GABAA receptor. Novel intrasubunit binding sites were identified within the transmembrane helical bundles of both the α1 (labeled residues α1-N408, Y415) and ß3 (labeled residue ß3-Y442) subunits, adjacent to the extracellular domains (ECDs). An intersubunit site (labeled residues ß3-L294 and G308) in the interface between the ß3(+) and α1(-) subunits of the GABAA receptor pentamer was also identified. Computational docking studies of neurosteroid to the three sites predicted critical residues contributing to neurosteroid interaction with the GABAA receptors. Electrophysiological studies of receptors with mutations based on these predictions (α1-V227W, N408A/Y411F, and Q242L) indicate that both the α1 intrasubunit and ß3-α1 intersubunit sites are critical for neurosteroid action.


Assuntos
Proteínas de Membrana/metabolismo , Receptores de GABA/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Eletrofisiologia , Feminino , Citometria de Fluxo , Humanos , Espectrometria de Massas , Simulação de Acoplamento Molecular , Muscimol/metabolismo , Neurotransmissores/metabolismo , Oócitos/metabolismo , Xenopus laevis
5.
Mol Pharmacol ; 100(1): 19-31, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33958479

RESUMO

Prior work employing functional analysis, photolabeling, and X-ray crystallography have identified three distinct binding sites for potentiating steroids in the heteromeric GABAA receptor. The sites are located in the membrane-spanning domains of the receptor at the ß-α subunit interface (site I) and within the α (site II) and ß subunits (site III). Here, we have investigated the effects of mutations to these sites on potentiation of the rat α1ß2γ2L GABAA receptor by the endogenous neurosteroid allopregnanolone (3α5αP). The mutations were introduced alone or in combination to probe the additivity of effects. We show that the effects of amino acid substitutions in sites I and II are energetically additive, indicating independence of the actions of the two steroid binding sites. In site III, none of the mutations tested reduced potentiation by 3α5αP, nor did a mutation in site III modify the effects of mutations in sites I or II. We infer that the binding sites for 3α5αP act independently. The independence of steroid action at each site is supported by photolabeling data showing that mutations in either site I or site II selectively change steroid orientation in the mutated site without affecting labeling at the unmutated site. The findings are discussed in the context of linking energetic additivity to empirical changes in receptor function and ligand binding. SIGNIFICANCE STATEMENT: Prior work has identified three distinct binding sites for potentiating steroids in the heteromeric γ-aminobutyric acid type A receptor. This study shows that the sites act independently and additively in the presence of the steroid allopregnanolone and provide estimates of energetic contributions made by steroid binding to each site.


Assuntos
Substituição de Aminoácidos , Pregnanolona/farmacologia , Receptores de GABA-A/química , Animais , Sítios de Ligação , Cristalografia por Raios X , Modelos Moleculares , Conformação Molecular , Simulação de Acoplamento Molecular , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Pregnanolona/química , Ratos , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo
6.
Mol Pharmacol ; 98(6): 762-769, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32978327

RESUMO

Synaptic GABAA receptors are alternately exposed to short pulses of a high, millimolar concentration of GABA and prolonged periods of low, micromolar concentration of the transmitter. Prior work has indicated that exposure to micromolar concentrations of GABA can both activate the postsynaptic receptors generating sustained low-amplitude current and desensitize the receptors, thereby reducing the peak amplitude of subsequent synaptic response. However, the precise relationship between tonic activation and reduction of peak response is not known. Here, we have measured the effect of prolonged exposure to GABA or the combination of GABA and the neurosteroid allopregnanolone, which was intended to desensitize a fraction of receptors, on a subsequent response to a high concentration of agonist in human α1ß3γ2L receptors expressed in Xenopus oocytes. We show that the reduction in the peak amplitude of the post-exposure test response correlates with the open probability of the preceding desensitizing response. Curve fitting of the inhibitory relationship yielded an IC50 of 12.5 µM and a Hill coefficient of -1.61. The activation and desensitization data were mechanistically analyzed in the framework of a three-state Resting-Active-Desensitized model. Using the estimated affinity, efficacy, and desensitization parameters, we calculated the amount of desensitization that would accumulate during a long (2-minute) application of GABA or GABA plus allopregnanolone. The results indicate that accumulation of desensitization depends on the level of activity rather than agonist or potentiator concentration per se. We estimate that in the presence of 1 µM GABA, approximately 5% of α1ß3γ2L receptors are functionally eliminated because of desensitization. SIGNIFICANCE STATEMENT: We present an analytical approach to quantify and predict the loss of activatable GABAA receptors due to desensitization in the presence of transmitter and the steroid allopregnanolone. The findings indicate that the peak amplitude of the synaptic response is influenced by ambient GABA and that changes in ambient concentrations of the transmitter and other GABAergic agents can modify tonically and phasically activated synaptic receptors in opposite directions.


Assuntos
Agonistas de Receptores de GABA-A/farmacologia , Receptores de GABA-A/metabolismo , Potenciais Sinápticos/efeitos dos fármacos , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Humanos , Oócitos , Técnicas de Patch-Clamp , Pregnanolona/farmacologia , Proteínas Recombinantes/metabolismo , Fatores de Tempo , Xenopus laevis , Ácido gama-Aminobutírico/farmacologia
7.
Mol Pharmacol ; 98(4): 303-313, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32873746

RESUMO

Muscimol is a psychoactive isoxazole derived from the mushroom Amanita muscaria and a potent orthosteric agonist of the GABAA receptor. The binding of [3H]muscimol has been used to evaluate the distribution of GABAA receptors in the brain, and studies of modulation of [3H]muscimol binding by allosteric GABAergic modulators such as barbiturates and steroid anesthetics have provided insight into the modes of action of these drugs on the GABAA receptor. It has, however, not been feasible to directly apply interaction parameters derived from functional studies to describe the binding of muscimol to the receptor. Here, we employed the Monod-Wyman-Changeux concerted transition model to analyze muscimol binding isotherms. We show that the binding isotherms from recombinant α1ß3 GABAA receptors can be qualitatively predicted using electrophysiological data pertaining to properties of receptor activation and desensitization in the presence of muscimol. The model predicts enhancement of [3H]muscimol binding in the presence of the steroids allopregnanolone and pregnenolone sulfate, although the steroids interact with distinct sites and either enhance (allopregnanolone) or reduce (pregnenolone sulfate) receptor function. We infer that the concerted transition model can be used to link radioligand binding and electrophysiological data. SIGNIFICANCE STATEMENT: The study employs a three-state resting-active-desensitized model to link radioligand binding and electrophysiological data. We show that the binding isotherms can be qualitatively predicted using parameters estimated in electrophysiological experiments and that the model accurately predicts the enhancement of [3H]muscimol binding in the presence of the potentiating steroid allopregnanolone and the inhibitory steroid pregnenolone sulfate.


Assuntos
Agonistas de Receptores de GABA-A/farmacologia , Muscimol/farmacologia , Receptores de GABA-A/metabolismo , Esteroides/farmacologia , Regulação Alostérica/efeitos dos fármacos , Sítios de Ligação , Células HEK293 , Humanos , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Muscimol/química , Pregnanolona/farmacologia , Pregnenolona/farmacologia , Receptores de GABA-A/química , Receptores de GABA-A/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Trítio/química
8.
Mol Pharmacol ; 98(4): 280-291, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32675382

RESUMO

The ρ1 GABAA receptor is prominently expressed in the retina and is present at lower levels in several brain regions and other tissues. Although the ρ1 receptor is insensitive to many anesthetic drugs that modulate the heteromeric GABAA receptor, it maintains a rich and multifaceted steroid pharmacology. The receptor is negatively modulated by 5ß-reduced steroids, sulfated or carboxylated steroids, and ß-estradiol, whereas many 5α-reduced steroids potentiate the receptor. In this study, we analyzed modulation of the human ρ1 GABAA receptor by several neurosteroids, individually and in combination, in the framework of the coagonist concerted transition model. Experiments involving coapplication of two or more steroids revealed that the receptor contains at least three classes of distinct, nonoverlapping sites for steroids, one each for the inhibitory steroids pregnanolone (3α5ßP), 3α5ßP sulfate, and ß-estradiol. The site for 3α5ßP can accommodate the potentiating steroid 5αTHDOC. The findings are discussed with respect to receptor modulation by combinations of endogenous neurosteroids. SIGNIFICANCE STATEMENT: The study describes modulation of the ρ1 GABAA receptor by neurosteroids. The coagonist concerted transition model was used to determine overlap of binding sites for several inhibitory and potentiating steroids.


Assuntos
Desoxicorticosterona/análogos & derivados , Neuroesteroides/farmacologia , Pregnanolona/farmacologia , Receptores de GABA-A/química , Receptores de GABA-A/metabolismo , Xenopus laevis/genética , Animais , Animais Geneticamente Modificados , Sítios de Ligação , Desoxicorticosterona/química , Desoxicorticosterona/farmacologia , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Modelos Moleculares , Estrutura Molecular , Neuroesteroides/química , Pregnanolona/química , Receptores de GABA-A/genética
9.
J Biol Chem ; 293(8): 3013-3027, 2018 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-29301936

RESUMO

Neurosteroids are endogenous sterols that potentiate or inhibit pentameric ligand-gated ion channels (pLGICs) and can be effective anesthetics, analgesics, or anti-epileptic drugs. The complex effects of neurosteroids on pLGICs suggest the presence of multiple binding sites in these receptors. Here, using a series of novel neurosteroid-photolabeling reagents combined with top-down and middle-down mass spectrometry, we have determined the stoichiometry, sites, and orientation of binding for 3α,5α-pregnane neurosteroids in the Gloeobacter ligand-gated ion channel (GLIC), a prototypic pLGIC. The neurosteroid-based reagents photolabeled two sites per GLIC subunit, both within the transmembrane domain; one site was an intrasubunit site, and the other was located in the interface between subunits. By using reagents with photoreactive groups positioned throughout the neurosteroid backbone, we precisely map the orientation of neurosteroid binding within each site. Amino acid substitutions introduced at either site altered neurosteroid modulation of GLIC channel activity, demonstrating the functional role of both sites. These results provide a detailed molecular model of multisite neurosteroid modulation of GLIC, which may be applicable to other mammalian pLGICs.


Assuntos
Proteínas de Bactérias/metabolismo , Desoxicorticosterona/análogos & derivados , Canais Iônicos de Abertura Ativada por Ligante/metabolismo , Modelos Moleculares , Neurotransmissores/metabolismo , Pregnanos/metabolismo , Substituição de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sítios de Ligação , Cianobactérias , Desoxicorticosterona/química , Desoxicorticosterona/metabolismo , Hidroxilação , Cinética , Canais Iônicos de Abertura Ativada por Ligante/química , Canais Iônicos de Abertura Ativada por Ligante/genética , Ligantes , Conformação Molecular , Simulação de Acoplamento Molecular , Mutagênese Sítio-Dirigida , Neurotransmissores/química , Marcadores de Fotoafinidade/química , Mutação Puntual , Pregnanos/química , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Multimerização Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
10.
Mol Pharmacol ; 93(5): 468-476, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29439087

RESUMO

GABAA receptors activated by the transmitter GABA are potentiated by several allosterically acting drugs, including the intravenous anesthetic propofol. Propofol can also directly activate the receptor, albeit at higher concentrations. Previous functional studies have identified amino acid residues whose substitution reduces potentiation of GABA-activated receptors by propofol while enhancing the ability of propofol to directly activate the receptor. One interpretation of such observations is that the mutation has specific effects on the sites or processes involved in potentiation or activation. We show here that divergent effects on potentiation and direct activation can be mediated by increased constitutive open probability in the mutant receptor without any specific effect on the interactions between the allosteric drug and the receptor. By simulating GABAA receptor activity using the concerted transition model, we demonstrate that the predicted degree of potentiation is reduced as the level of constitutive activity increases. The model further predicts that a potentiating effect of an allosteric modulator is a computable value that depends on the level of constitutive activity, the amplitude of the response to the agonist, and the amplitude of the direct activating response to the modulator. Specific predictions were confirmed by electrophysiological data from the binary α1ß3 and concatemeric ternary ß2α1γ2L+ß2α1 GABAA receptors. The corollaries of reduced potentiation due to increased constitutive activity are isobolograms that conform to simple additivity and a loss of separation between the concentration-response relationships for direct activation and potentiation.


Assuntos
Anestésicos Intravenosos/farmacologia , Mutação , Propofol/farmacologia , Receptores de GABA-A/efeitos dos fármacos , Receptores de GABA-A/genética , Regulação Alostérica , Anestésicos Intravenosos/metabolismo , Animais , Células Cultivadas , Sinergismo Farmacológico , Agonistas GABAérgicos/farmacologia , Humanos , Propofol/metabolismo , Receptores de GABA-A/metabolismo , Xenopus , Ácido gama-Aminobutírico/metabolismo
11.
J Biol Chem ; 292(22): 9294-9304, 2017 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-28396346

RESUMO

Voltage-dependent anion channel-1 (VDAC1) is a highly regulated ß-barrel membrane protein that mediates transport of ions and metabolites between the mitochondria and cytosol of the cell. VDAC1 co-purifies with cholesterol and is functionally regulated by cholesterol, among other endogenous lipids. Molecular modeling studies based on NMR observations have suggested five cholesterol-binding sites in VDAC1, but direct experimental evidence for these sites is lacking. Here, to determine the sites of cholesterol binding, we photolabeled purified mouse VDAC1 (mVDAC1) with photoactivatable cholesterol analogues and analyzed the photolabeled sites with both top-down mass spectrometry (MS), and bottom-up MS paired with a clickable, stable isotope-labeled tag, FLI-tag. Using cholesterol analogues with a diazirine in either the 7 position of the steroid ring (LKM38) or the aliphatic tail (KK174), we mapped a binding pocket in mVDAC1 localized to Thr83 and Glu73, respectively. When Glu73 was mutated to a glutamine, KK174 no longer photolabeled this residue, but instead labeled the nearby Tyr62 within this same binding pocket. The combination of analytical strategies employed in this work permits detailed molecular mapping of a cholesterol-binding site in a protein, including an orientation of the sterol within the site. Our work raises the interesting possibility that cholesterol-mediated regulation of VDAC1 may be facilitated through a specific binding site at the functionally important Glu73 residue.


Assuntos
Colesterol/química , Canal de Ânion 1 Dependente de Voltagem/química , Marcadores de Afinidade , Animais , Sítios de Ligação , Camundongos , Ressonância Magnética Nuclear Biomolecular , Canal de Ânion 1 Dependente de Voltagem/genética
12.
N Engl J Med ; 372(9): 796-7, 2015 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-25714157

RESUMO

Some anesthetics and sedatives have been shown to cause neurotoxic effects in laboratory animals. The FDA collaboration SmartTots recommends undertaking large-scale clinical studies and avoiding nonurgent surgical procedures requiring anesthesia in children younger than 3 years of age.


Assuntos
Anestésicos/efeitos adversos , Encéfalo/efeitos dos fármacos , Hipnóticos e Sedativos/efeitos adversos , Deficiências da Aprendizagem/induzido quimicamente , Modelos Animais , Guias de Prática Clínica como Assunto , Procedimentos Cirúrgicos Operatórios , Animais , Pré-Escolar , Humanos , Complicações Pós-Operatórias , Receptores de GABA/efeitos dos fármacos , Receptores de Glutamato/efeitos dos fármacos
13.
Anesth Analg ; 126(4): 1393-1396, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29401076

RESUMO

SmartTots (http://smarttots.org/) represents a public-private partnership between the International Anesthesia Research Society and the US Food and Drug Administration. Over the past 7 years, SmartTots has worked in collaboration with various stakeholders to determine whether anesthetic drugs have detrimental effects on the developing brain. SmartTots has funded clinical and preclinical studies, organized meetings, served as a repository of peer-reviewed information, and facilitated the development of consensus-based statements. Here, we report advances in the field of anesthetic neurotoxicity and provide an update on SmartTots' activities. Clinical studies have provided some reassurance that a brief exposure to anesthetic drugs does not cause overt, persistent cognitive deficits. New recommendations aim to increase the reproducibility and "clinical relevance" of data from studies of laboratory animals. Overall, the field has advanced substantially; however, it remains paramount to definitively resolve whether anesthetic drugs are neurotoxic to the immature brain. The results of SmartTots efforts will either ally unwarranted fears or substantially change pediatric anesthetic practice and prompt studies to identify neuroprotective strategies.


Assuntos
Desenvolvimento do Adolescente/efeitos dos fármacos , Anestésicos Gerais/efeitos adversos , Pesquisa Biomédica/métodos , Encéfalo/efeitos dos fármacos , Desenvolvimento Infantil/efeitos dos fármacos , Síndromes Neurotóxicas/etiologia , Adolescente , Comportamento do Adolescente/efeitos dos fármacos , Fatores Etários , Animais , Encéfalo/crescimento & desenvolvimento , Criança , Comportamento Infantil/efeitos dos fármacos , Pré-Escolar , Humanos , Lactente , Recém-Nascido , Síndromes Neurotóxicas/fisiopatologia , Síndromes Neurotóxicas/psicologia , Parcerias Público-Privadas , Medição de Risco , Fatores de Risco , Sociedades Científicas , Estados Unidos , United States Food and Drug Administration
14.
Anal Chem ; 89(4): 2636-2644, 2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-28194953

RESUMO

Identifying sites of protein-ligand interaction is important for structure-based drug discovery and understanding protein structure-function relationships. Mass spectrometry (MS) has emerged as a useful tool for identifying residues covalently modified by ligands. Current methods use database searches that are dependent on acquiring interpretable fragmentation spectra (MS2) of peptide-ligand adducts. This is problematic for identifying sites of hydrophobic ligand incorporation in integral membrane proteins (IMPs), where poor aqueous solubility and ionization of peptide-ligand adducts and collision-induced adduct loss hinder the acquisition of quality MS2 spectra. To address these issues, we developed a fast ligand identification (FLI) tag that can be attached to any alkyne-containing ligand via Cu(I)-catalyzed cycloaddition. The FLI tag adds charge to increase solubility and ionization, and utilizes stable isotope labeling for MS1 level identification of hydrophobic peptide-ligand adducts. The FLI tag was coupled to an alkyne-containing neurosteroid photolabeling reagent and used to identify peptide-steroid adducts in MS1 spectra via the stable heavy isotope pair. Peptide-steroid adducts were not identified in MS2-based database searches because collision-induced adduct loss was the dominant feature of collision-induced dissociation (CID) fragmentation, but targeted analysis of MS1 pairs using electron transfer dissociation (ETD) markedly reduced adduct loss. Using the FLI tag and ETD, we identified Glu73 as the site of photoincorporation of our neurosteroid ligand in the IMP, mouse voltage-dependent anion channel-1 (mVDAC1), and top-down MS confirmed a single site of photolabeling.


Assuntos
Ligantes , Peptídeos/química , Espectrometria de Massas em Tandem , Canal de Ânion 1 Dependente de Voltagem/metabolismo , Alcinos/química , Sequência de Aminoácidos , Animais , Cromatografia Líquida de Alta Pressão , Química Click , Interações Hidrofóbicas e Hidrofílicas , Marcação por Isótopo , Camundongos , Peptídeos/metabolismo , Solubilidade , Raios Ultravioleta , Canal de Ânion 1 Dependente de Voltagem/química
17.
Mol Pharmacol ; 88(4): 736-45, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26206487

RESUMO

Propofol is a sedative and anesthetic agent that can both activate GABA(A) receptors and potentiate receptor activation elicited by submaximal concentrations of the transmitter. A recent modeling study of the ß3 homomeric GABA(A) receptor postulated a high-affinity propofol binding site in a hydrophobic pocket in the middle of a triangular cleft lined by the M1 and M2 membrane-spanning domains of one subunit and the M2 domain of the neighboring subunit. The goal of the present study was to gain functional evidence for the involvement of this pocket in the actions of propofol. Human ß3 and α1ß3 receptors were expressed in Xenopus oocytes, and the effects of substitutions of selected residues were probed on channel activation by propofol and pentobarbital. The data demonstrate the vital role of the ß3(Y143), ß3(F221), ß3(Q224), and ß3(T266) residues in the actions of propofol but not pentobarbital in ß3 receptors. The effects of ß3(Y143W) and ß3(Q224W) on activation by propofol are likely steric because propofol analogs with less bulky ortho substituents activated both wild-type and mutant receptors. The T266W mutation removed activation by propofol in ß3 homomeric receptors; however, this mutation alone or in combination with a homologous mutation (I271W) in the α1 subunit had almost no effect on activation properties in α1ß3 heteromeric receptors. We hypothesize that heteromeric α1ß3 receptors can be activated by propofol interactions with ß3-ß3, α1-ß3, and ß3-α1 interfaces, but the exact locations of the binding site and/or nature of interactions vary in different classes of interfaces.


Assuntos
Análise Mutacional de DNA/métodos , Mutação/genética , Propofol/metabolismo , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Animais , Sítios de Ligação/genética , Relação Dose-Resposta a Droga , Feminino , Humanos , Estrutura Secundária de Proteína , Receptores de GABA-A/química , Xenopus laevis
18.
Nat Chem Biol ; 9(11): 715-20, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24056400

RESUMO

Propofol is the most important intravenous general anesthetic in current clinical use. It acts by potentiating GABAA (γ-aminobutyric acid type A) receptors, but where it binds to this receptor is not known and has been a matter of some debate. We synthesized a new propofol analog photolabeling reagent whose biological activity is very similar to that of propofol. We confirmed that this reagent labeled known propofol binding sites in human serum albumin that have been identified using X-ray crystallography. Using a combination of protiated and deuterated versions of the reagent to label mammalian receptors in intact membranes, we identified a new binding site for propofol in GABAA receptors consisting of both ß3 homopentamers and α1ß3 heteropentamers. The binding site is located within the ß subunit at the interface between the transmembrane domains and the extracellular domain and lies close to known determinants of anesthetic sensitivity in the transmembrane segments TM1 and TM2.


Assuntos
Marcadores de Fotoafinidade/análise , Propofol/metabolismo , Receptores de GABA-A/química , Receptores de GABA-A/metabolismo , Sítios de Ligação , Humanos , Modelos Moleculares , Estrutura Molecular , Marcadores de Fotoafinidade/química , Propofol/química , Albumina Sérica/química , Relação Estrutura-Atividade
19.
N Engl J Med ; 365(7): 591-600, 2011 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-21848460

RESUMO

BACKGROUND: Unintended intraoperative awareness, which occurs when general anesthesia is not achieved or maintained, affects up to 1% of patients at high risk for this complication. We tested the hypothesis that a protocol incorporating the electroencephalogram-derived bispectral index (BIS) is superior to a protocol incorporating standard monitoring of end-tidal anesthetic-agent concentration (ETAC) for the prevention of awareness. METHODS: We conducted a prospective, randomized, evaluator-blinded trial at three medical centers. We randomly assigned 6041 patients at high risk for awareness to BIS-guided anesthesia (with an audible alert if the BIS value was <40 or >60, on a scale of 0 to 100, with 0 indicating the suppression of detectable brain electrical activity and 100 indicating the awake state) or ETAC-guided anesthesia (with an audible alert if the ETAC was <0.7 or >1.3 minimum alveolar concentration). In addition to audible alerts, the protocols included structured education and checklists. Superiority of the BIS protocol was assessed with the use of a one-sided Fisher's exact test. RESULTS: A total of 7 of 2861 patients (0.24%) in the BIS group, as compared with 2 of 2852 (0.07%) in the ETAC group, who were interviewed postoperatively had definite intraoperative awareness (a difference of 0.17 percentage points; 95% confidence interval [CI], -0.03 to 0.38; P=0.98). Thus, the superiority of the BIS protocol was not demonstrated. A total of 19 cases of definite or possible intraoperative awareness (0.66%) occurred in the BIS group, as compared with 8 (0.28%) in the ETAC group (a difference of 0.38 percentage points; 95% CI, 0.03 to 0.74; P=0.99), with the superiority of the BIS protocol again not demonstrated. There was no difference between the groups with respect to the amount of anesthesia administered or the rate of major postoperative adverse outcomes. CONCLUSIONS: The superiority of the BIS protocol was not established; contrary to expectations, fewer patients in the ETAC group than in the BIS group experienced awareness. (Funded by the Foundation for Anesthesia Education and Research and others; BAG-RECALL ClinicalTrials.gov number, NCT00682825.).


Assuntos
Anestesia Geral , Anestésicos Inalatórios/análise , Monitores de Consciência , Consciência no Peroperatório/prevenção & controle , Monitorização Intraoperatória/métodos , Alvéolos Pulmonares/química , Adulto , Idoso , Eletroencefalografia , Feminino , Humanos , Consciência no Peroperatório/epidemiologia , Masculino , Pessoa de Meia-Idade , Complicações Pós-Operatórias , Estudos Prospectivos , Fatores de Risco , Método Simples-Cego
20.
Mol Cell Proteomics ; 11(1): M111.011445, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22338125

RESUMO

Mass spectrometric sequencing of low abundance, integral membrane proteins, particularly the transmembrane domains, presents challenges that span the multiple phases of sample preparation including solubilization, purification, enzymatic digestion, peptide extraction, and chromatographic separation. We describe a method through which we have obtained high peptide coverage for 12 γ-aminobutyric acid type A receptor (GABAA receptor) subunits from 2 picomoles of affinity-purified GABAA receptors from rat brain neocortex. Focusing on the α1 subunit, we identified peptides covering 96% of the protein sequence from fragmentation spectra (MS2) using a database searching algorithm and deduced 80% of the amino acid residues in the protein from de novo sequencing of Orbitrap spectra. The workflow combined microscale membrane protein solubilization, protein delipidation, in-solution multi-enzyme digestion, multiple stationary phases for peptide extraction, and acquisition of high-resolution full scan and fragmentation spectra. For de novo sequencing of peptides containing the transmembrane domains, timed digestions with chymotrypsin were utilized to generate peptides with overlapping sequences that were then recovered by sequential solid phase extraction using a C4 followed by a porous graphitic carbon stationary phase. The specificity of peptide identifications and amino acid residue sequences was increased by high mass accuracy and charge state assignment to parent and fragment ions. Analysis of three separate brain samples demonstrated that 78% of the sequence of the α1 subunit was observed in all three replicates with an additional 13% covered in two of the three replicates, indicating a high degree of sequence coverage reproducibility. Label-free quantitative analysis was applied to the three replicates to determine the relative abundances of 11 γ-aminobutyric acid type A receptor subunits. The deep sequence MS data also revealed two N-glycosylation sites on the α1 subunit, confirmed two splice variants of the γ2 subunit (γ2L and γ2S) and resolved a database discrepancy in the sequence of the α5 subunit.


Assuntos
Espectrometria de Massas/métodos , Receptores de GABA-A/química , Análise de Sequência de Proteína/métodos , Sequência de Aminoácidos , Animais , Cromatografia Líquida , Eletroforese em Gel de Poliacrilamida , Dados de Sequência Molecular , Neocórtex , Peptídeos/química , Isoformas de Proteínas , Subunidades Proteicas/química , Ratos , Extração em Fase Sólida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA