Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biophys J ; 2024 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-38678367

RESUMO

Bupropion is an atypical antidepressant and smoking cessation drug that causes adverse effects such as insomnia, irritability, and anxiety. Bupropion inhibits dopamine and norepinephrine reuptake transporters and eukaryotic cation-conducting pentameric ligand-gated ion channels, such as nicotinic acetylcholine and serotonin type 3A receptors, at clinically relevant concentrations. Here, we demonstrate that bupropion also inhibits a prokaryotic homolog of pentameric ligand-gated ion channels, the Gloeobacter violaceus ligand-gated ion channel (GLIC). Using the GLIC as a model, we used molecular docking to predict binding sites for bupropion. Bupropion was found to bind to several sites within the transmembrane domain, with the predominant site being localized to the interface between transmembrane segments M1 and M3 of two adjacent subunits. Residues W213, T214, and W217 in the first transmembrane segment, M1, and F267 and I271 in the third transmembrane segment, M3, most frequently reside within a 4 Å distance from bupropion. We then used single amino acid substitutions at these positions and two-electrode voltage-clamp recordings to determine their impact on bupropion inhibitory effects. The substitution T214F alters bupropion potency by shifting the half-maximal inhibitory concentration to a 13-fold higher value compared to wild-type GLIC. Residue T214 is found within a previously identified binding pocket for neurosteroids and lipids in the GLIC. This intersubunit binding pocket is structurally conserved and almost identical to a binding pocket described for neurosteroids in γ-aminobutyric acid type A receptors. Our data thus suggest that the T214 that lines a previously identified lipophilic binding pocket in GLIC and γ-aminobutyric acid type A receptors is also a modulatory site for bupropion interaction with the GLIC.

2.
bioRxiv ; 2023 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-37873398

RESUMO

Bupropion is an atypical antidepressant and smoking cessation drug which causes adverse effects such as insomnia, irritability, and anxiety. Bupropion inhibits dopamine and norepinephrine reuptake transporters and eukaryotic cation-conducting pentameric ligand-gated ion channels (pLGICs), such as nicotinic acetylcholine (nACh) and serotonin type 3A (5-HT3A) receptors, at clinically relevant concentrations. However, the binding sites and binding mechanisms of bupropion are still elusive. To further understand the inhibition of pLGICs by bupropion, in this work, using a prokaryotic homologue of pLGICs as a model, we examined the inhibitory potency of bupropion in Gloeobacter violaceus ligand-gated ion channel (GLIC), a proton-gated ion channel. Bupropion inhibited proton-induced currents in GLIC with an inhibitory potency of 14.9 ± 2.0 µM, comparable to clinically attainable concentrations previously shown to also modulate eukaryotic pLGICs. Using single amino acid substitutions in GLIC and two-electrode voltage-clamp recordings, we further determined a binding site for bupropion in the lower third of the first transmembrane segment M1 at residue T214. The sidechain of M1 T214 together with additional residues of M1 and also of M3 of the adjacent subunit have previously been shown to contribute to binding of other lipophilic molecules like allopregnanolone and pregnanolone.

3.
Methods Mol Biol ; 2507: 425-444, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35773596

RESUMO

Proton coupled folate transporter (PCFT) is an integral membrane protein with 12 transmembrane segments localized to the plasma membrane. PCFT is the main route by which folate, vitamin B9, from dietary sources enters mammalian cells in the small intestine. Loss-of-function mutations in this membrane transport protein cause hereditary folate malabsorption, and upregulation of PCFT has been reported in cancer cells. Currently, a complete translocation mechanism of folate via PCFT is still missing. To reveal this mechanism via studies of structural architecture and structure-function relationships, soluble and stable PCFT in a phospholipid bilayer environment is needed. We therefore develop an approach to screen lipid environments in which PCFT is most soluble. Traditional in vitro expression and reconstitution into lipid bilayers of integral membrane proteins requires separate steps, which are costly and time-consuming. In this chapter, we describe a protocol for in vitro translation of PCFT into preformed lipid nanodiscs using a cell-free expression system, which helps to accelerate and reduce the cost of the sample preparation.


Assuntos
Deficiência de Ácido Fólico , Transportador de Folato Acoplado a Próton , Animais , Ácido Fólico/metabolismo , Deficiência de Ácido Fólico/metabolismo , Lipídeos , Mamíferos/metabolismo , Mutação , Transportador de Folato Acoplado a Próton/química , Transportador de Folato Acoplado a Próton/genética , Transportador de Folato Acoplado a Próton/metabolismo
4.
PLoS One ; 16(11): e0253184, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34793461

RESUMO

The Proton-Coupled Folate Transporter (PCFT) is a transmembrane transport protein that controls the absorption of dietary folates in the small intestine. PCFT also mediates uptake of chemotherapeutically used antifolates into tumor cells. PCFT has been identified within lipid rafts observed in phospholipid bilayers of plasma membranes, a micro environment that is altered in tumor cells. The present study aimed at investigating the impact of different lipids within Lipid-protein nanodiscs (LPNs), discoidal lipid structures stabilized by membrane scaffold proteins, to yield soluble PCFT expression in an E. coli lysate-based cell-free transcription/translation system. In the absence of detergents or lipids, we observed PCFT quantitatively as precipitate in this system. We then explored the ability of LPNs to support solubilized PCFT expression when present during in-vitro translation. LPNs consisted of either dimyristoyl phosphatidylcholine (DMPC), palmitoyl-oleoyl phosphatidylcholine (POPC), or dimyristoyl phosphatidylglycerol (DMPG). While POPC did not lead to soluble PCFT expression, both DMPG and DMPC supported PCFT translation directly into LPNs, the latter in a concentration dependent manner. The results obtained through this study provide insights into the lipid preferences of PCFT. Membrane-embedded or solubilized PCFT will enable further studies with diverse biophysical approaches to enhance the understanding of the structure and molecular mechanism of folate transport through PCFT.


Assuntos
Microdomínios da Membrana/metabolismo , Transportador de Folato Acoplado a Próton/metabolismo , Transporte Biológico , Membrana Celular/metabolismo , Humanos , Lipídeos
5.
Biophys J ; 119(12): 2593-2603, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33157122

RESUMO

The intracellular domain of the serotonin type 3A receptor, a pentameric ligand-gated ion channel, is crucial for regulating conductance. Ion permeation through the extracellular vestibule and the transmembrane channel is well understood, whereas the specific ion conduction pathway through the intracellular domain is less clear. The intracellular domain starts with a short loop after the third transmembrane segment, followed by a short α-helical segment, a large unstructured loop, and finally, the membrane-associated MA-helix that continues into the last transmembrane segment. The MA-helices from all five subunits form the extension of the transmembrane ion channel and shape what has been described as a "closed vestibule," with their lateral portals obstructed by loops and their cytosolic ends forming a tight hydrophobic constriction. The question remains whether the lateral portals or cytosolic constriction conduct ions upon channel opening. In our study, we used disulfide bond formation between pairs of engineered cysteines to probe the proximity and mobility of segments of the MA-helices most distal to the membrane bilayer. Our results indicate that the proximity and orientation for cysteine pairs at I409C/R410C, in close proximity to the lateral windows, and L402C/L403C, at the cytosolic ends of the MA-helices, are conducive for disulfide bond formation. Although conformational changes associated with gating promote cross-linking for I409C/R410C, which in turn decreases channel currents, cross-linking of L402C/L403C is functionally silent in macroscopic currents. These results support the hypothesis that concerted conformational changes open the lateral portals for ion conduction, rendering ion conduction through the vertical portal unlikely.


Assuntos
Receptores 5-HT3 de Serotonina , Serotonina , Canais Iônicos , Íons , Conformação Proteica em alfa-Hélice , Receptores 5-HT3 de Serotonina/genética , Receptores 5-HT3 de Serotonina/metabolismo
6.
Mol Pharmacol ; 97(3): 171-179, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31871303

RESUMO

Bupropion, a Food and Drug Administration-approved antidepressant and smoking cessation aid, blocks dopamine and norepinephrine reuptake transporters and noncompetitively inhibits nicotinic acetylcholine and serotonin (5-HT) type 3A receptors (5-HT3ARs). 5-HT3 receptors are pentameric ligand-gated ion channels that regulate synaptic activity in the central and peripheral nervous system, presynaptically and postsynaptically. In the present study, we examined and compared the effect of bupropion and its active metabolite hydroxybupropion on mouse homomeric 5-HT3A and heteromeric 5-HT3AB receptors expressed in Xenopus laevis oocytes using two-electrode voltage clamp experiments. Coapplication of bupropion or hydroxybupropion with 5-HT dose dependently inhibited 5-HT-induced currents in heteromeric 5-HT type 3AB receptors (5-HT3ABRs) (IC50 = 840 and 526 µM, respectively). The corresponding IC50s for bupropion and hydroxybupropion for homomeric 5-HT3ARs were 10- and 5-fold lower, respectively (87 and 113 µM). The inhibition of 5-HT3ARs and 5-HT3ABRs was non-use dependent and voltage independent, suggesting bupropion is not an open channel blocker. The inhibition by bupropion was reversible and time-dependent. Of note, preincubation with a low concentration of bupropion that mimics therapeutic drug conditions inhibits 5-HT-induced currents in 5-HT3A and 5-HT3AB receptors considerably. In summary, we demonstrate that bupropion inhibits heteromeric 5-HT3ABRs as well as homomeric 5-HT3ARs. This inhibition occurs at clinically relevant concentrations and may contribute to bupropion's clinical effects. SIGNIFICANCE STATEMENT: Clinical studies indicate that antagonizing serotonin (5-HT) type 3AB (5-HT3AB) receptors in brain areas involved in mood regulation is successful in treating mood and anxiety disorders. Previously, bupropion was shown to be an antagonist at homopentameric 5-HT type 3A receptors. The present work provides novel insights into the pharmacological effects that bupropion exerts on heteromeric 5-HT3AB receptors, in particular when constantly present at low, clinically attainable concentrations. The results advance the knowledge on the clinical effects of bupropion as an antidepressant.


Assuntos
Bupropiona/metabolismo , Bupropiona/farmacologia , Receptores 5-HT3 de Serotonina/metabolismo , Antagonistas do Receptor 5-HT3 de Serotonina/metabolismo , Antagonistas do Receptor 5-HT3 de Serotonina/farmacologia , Sequência de Aminoácidos , Animais , Inibidores da Captação de Dopamina/metabolismo , Inibidores da Captação de Dopamina/farmacologia , Relação Dose-Resposta a Droga , Feminino , Camundongos , Receptores 5-HT3 de Serotonina/genética , Estereoisomerismo , Xenopus laevis
7.
PLoS One ; 12(5): e0177572, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28493963

RESUMO

The proton-coupled folate transporter (PCFT) provides an essential uptake route for the vitamin folic acid (B9) in mammals. In addition, it is currently of high interest for targeting chemotherapeutic agents to tumors due to the increased folic acid requirement of rapidly dividing tumor cells as well as the upregulated PCFT expression in several tumors. To understand its function, determination of its atomic structure and molecular mechanism of transport are essential goals that require large amounts of functional PCFT. Here, we present a high-level heterologous expression system for human PCFT using a recombinant baculovirus and Spodoptera frugiperda (Sf9) insect cells. We demonstrate folate transport functionality along the PCFT expression, isolation, and purification process. Importantly, purified PCFT transports folic acid after reconstitution. We thus succeeded in overcoming heterologous expression as a major bottleneck of PCFT research. The availability of an overexpression system for human PCFT provides the basis for future biochemical, biophysical and structural studies.


Assuntos
Expressão Gênica , Transportador de Folato Acoplado a Próton/isolamento & purificação , Transportador de Folato Acoplado a Próton/metabolismo , Células Sf9/metabolismo , Animais , Cromatografia de Afinidade , Cromatografia em Gel , Detergentes/farmacologia , Ácido Fólico/metabolismo , Humanos , Lipossomos/metabolismo , Solubilidade , Trítio/metabolismo
8.
Neuropharmacology ; 113(Pt A): 89-99, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27671323

RESUMO

The FDA-approved antidepressant and smoking cessation drug bupropion is known to inhibit dopamine and norepinephrine reuptake transporters, as well as nicotinic acetylcholine receptors (nAChRs) which are cation-conducting members of the Cys-loop superfamily of ion channels, and more broadly pentameric ligand-gated ion channels (pLGICs). In the present study, we examined the ability of bupropion and its primary metabolite hydroxybupropion to block the function of cation-selective serotonin type 3A receptors (5-HT3ARs), and further characterized bupropion's pharmacological effects at these receptors. Mouse 5-HT3ARs were heterologously expressed in HEK-293 cells or Xenopus laevis oocytes for equilibrium binding studies. In addition, the latter expression system was utilized for functional studies by employing two-electrode voltage-clamp recordings. Both bupropion and hydroxybupropion inhibited serotonin-gated currents from 5-HT3ARs reversibly and dose-dependently with inhibitory potencies of 87 µM and 112 µM, respectively. Notably, the measured IC50 value for hydroxybupropion is within its therapeutically-relevant concentrations. The blockade by bupropion was largely non-competitive and non-use-dependent. Unlike its modulation at cation-selective pLGICs, bupropion displayed no significant inhibition of the function of anion-selective pLGICs. In summary, our results demonstrate allosteric blockade by bupropion of the 5-HT3AR. Importantly, given the possibility that bupropion's major active metabolite may achieve clinically relevant concentrations in the brain, our novel findings delineate a not yet identified pharmacological principle underlying its antidepressant effect.


Assuntos
Antidepressivos de Segunda Geração/farmacocinética , Bupropiona/análogos & derivados , Bupropiona/farmacocinética , Receptores 5-HT3 de Serotonina/metabolismo , Antagonistas do Receptor 5-HT3 de Serotonina/farmacocinética , Serotonina/metabolismo , Regulação Alostérica , Animais , Relação Dose-Resposta a Droga , Granisetron/farmacocinética , Células HEK293 , Humanos , Camundongos , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Serotonina/análise , Serotonina/farmacocinética , Xenopus laevis
9.
FEBS Open Bio ; 6(3): 216-30, 2016 03.
Artigo em Inglês | MEDLINE | ID: mdl-27047750

RESUMO

The proton-coupled folate transporter (PCFT, SLC46A1) transports folic acid across the plasma membrane, together with an excess of protons such that the net charge translocation is positive. We developed 3D structural models of PCFT threaded onto the X-ray structures of major facilitator superfamily (MFS) members that were identified as close structural homologues. The model of PCFT threaded onto the glycerol-3-phosphate transporter (GlpT) structure is consistent with detailed accessibility studies in the absence of extracellular substrate and at pH 7.4 presented here, and additionally with a multitude of other mutagenesis and functional studies. Characteristic MFS structural features are preserved in this PCFT model, such as 12 transmembrane helices divided into two pseudosymmetric bundles, and a high density of positive charges on the periphery of the cytoplasmic site that allow interactions with negatively charged lipid head-groups. Under the experimental conditions, PCFT predominantly samples the resting state, which in this case is inward-open. Several positions lining the substrate cavity have been identified. Motif A, a helix-turn-helix motif that is a hallmark of MFS transporters between transmembrane segments II and III is oriented appropriately to interact with residues from transmembrane segments IV as well as XI upon conformational transition to the outward-open state. A charge-relay system between three charged residues as well as apposing glycines in two α-helices, both contributed to by motif A, become engaged when PCFT is modeled on the outward-open state of a putative proton-driven transporter (YajR).

10.
PLoS One ; 8(10): e78301, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24205192

RESUMO

The proton-coupled folate transporter (PCFT) was recently identified as the major uptake route for dietary folates in humans. The three-dimensional structure of PCFT and its detailed interplay with function remain to be determined. We screened the water-accessible extracellular surface of HsPCFT using the substituted-cysteine accessibility method, to investigate the boundaries between the water-accessible surface and inaccessible buried protein segments. Single-cysteines, engineered individually at 40 positions in a functional cysteine-less HsPCFT background construct, were probed for plasma-membrane expression in Xenopus oocytes with a bilayer-impermeant primary-amine-reactive biotinylating agent (sulfosuccinimidyl 6-(biotinamido) hexanoate), and additionally for water-accessibility of the respective engineered cysteine with the sulfhydryl-selective biotinylating agent 2-((biotinoyl)amino)ethyl methanethiosulfonate. The ratio between Cys-selective over amine-selective labeling was further used to evaluate three-dimensional models of HsPCFT generated by homology / threading modeling. The closest homologues of HsPCFT with a known experimentally-determined three-dimensional structure are all members of one of the largest membrane protein super-families, the major facilitator superfamily (MFS). The low sequence identity--14% or less--between HsPCFT and these templates necessitates experiment-based evaluation and model refinement of homology/threading models. With the present set of single-cysteine accessibilities, the models based on GlpT and PepTSt are most promising for further refinement.


Assuntos
Transportador de Folato Acoplado a Próton/metabolismo , Água/metabolismo , Sequência de Aminoácidos , Animais , Biotinilação/métodos , Membrana Celular/metabolismo , Cisteína/metabolismo , Feminino , Proteínas de Membrana/metabolismo , Oócitos/metabolismo , Xenopus laevis/metabolismo
11.
FEBS J ; 280(12): 2900-15, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23601781

RESUMO

Folic acid is an essential vitamin required for de novo biosynthesis of nucleotides and amino acids. The proton-coupled folate transporter (PCFT; SLC46A1) has been identified as the major contributor for intestinal folate uptake. It is also involved in folate transport across the blood-brain barrier and into solid tumors. PCFT belongs to the major facilitator superfamily. Major facilitator superfamily members can exist in either monomeric or homo-oligomeric form. Here, we utilized blue native polyacrylamide gel electrophoresis (BN/PAGE) and crosslinking with bi-functional chemicals to investigate the quaternary structure of human PCFT after heterologous expression in Xenopus laevis oocytes and CHO cells. PCFT was expressed in the plasma membrane in both expression systems. The functionality of the utilized PCFT construct was confirmed in oocytes by folic acid induced currents at acidic pH. For both the oocyte and CHO expression system [(3)H]folic acid uptake studies indicated that PCFT was functional. To analyze the oligomeric state of PCFT in the plasma membrane, plasma membranes were isolated by polymerization with colloidal silica and polyacrylic acid and subsequent centrifugation. The digitonin-solubilized non-denatured PCFT migrated during BN/PAGE as a monomer, as judged by comparison with a membrane protein (5-HT(3A) receptor) of known pentameric assembly that was used to create a molecular sizing ladder. The chemical crosslinkers glutaraldehyde and dimethyl adipimidate were not able to covalently link potential higher order PCFT structures to form oligomers that were stable following SDS treatment. Together, our results demonstrate that plasma-membrane PCFT functions as a monomeric protein.


Assuntos
Membrana Celular/metabolismo , Transportador de Folato Acoplado a Próton/metabolismo , Animais , Transporte Biológico , Células CHO , Calibragem , Cricetinae , Eletroforese em Gel de Poliacrilamida/normas , Feminino , Ácido Fólico/metabolismo , Glicosilação , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Potenciais da Membrana , Peso Molecular , Oócitos/metabolismo , Processamento de Proteína Pós-Traducional , Estrutura Quaternária de Proteína , Transportador de Folato Acoplado a Próton/química , Padrões de Referência , Xenopus laevis
12.
Biochemistry ; 51(12): 2425-35, 2012 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-22394379

RESUMO

Bupropion, a clinically used antidepressant and smoking-cessation drug, acts as a noncompetitive antagonist of nicotinic acetylcholine receptors (nAChRs). To identify its binding site(s) in nAChRs, we developed a photoreactive bupropion analogue, (±)-2-(N-tert-butylamino)-3'-[(125)I]-iodo-4'-azidopropiophenone (SADU-3-72). Based on inhibition of [(125)I]SADU-3-72 binding, SADU-3-72 binds with high affinity (IC(50) = 0.8 µM) to the Torpedo nAChR in the resting (closed channel) state and in the agonist-induced desensitized state, and bupropion binds to that site with 3-fold higher affinity in the desensitized (IC(50) = 1.2 µM) than in the resting state. Photolabeling of Torpedo nAChRs with [(125)I]SADU-3-72 followed by limited in-gel digestion of nAChR subunits with endoproteinase Glu-C established the presence of [(125)I]SADU-3-72 photoincorporation within nAChR subunit fragments containing M1-M2-M3 helices (αV8-20K, ßV8-22/23K, and γV8-24K) or M1-M2 helices (δV8-14). Photolabeling within ßV8-22/23K, γV8-24K, and δV8-14 was reduced in the desensitized state and inhibited by ion channel blockers selective for the resting (tetracaine) or desensitized (thienycyclohexylpiperidine (TCP)) state, and this pharmacologically specific photolabeling was localized to the M2-9 leucine ring (δLeu(265), ßLeu(257)) within the ion channel. In contrast, photolabeling within the αV8-20K was enhanced in the desensitized state and not inhibited by TCP but was inhibited by bupropion. This agonist-enhanced photolabeling was localized to αTyr(213) in αM1. These results establish the presence of two distinct bupropion binding sites within the Torpedo nAChR transmembrane domain: a high affinity site at the middle (M2-9) of the ion channel and a second site near the extracellular end of αM1 within a previously described halothane (general anesthetic) binding pocket.


Assuntos
Azidas/metabolismo , Bupropiona/análogos & derivados , Bupropiona/metabolismo , Membrana Celular/metabolismo , Receptores Nicotínicos/química , Receptores Nicotínicos/metabolismo , Torpedo , Animais , Antidepressivos/química , Antidepressivos/metabolismo , Antidepressivos/farmacologia , Sítios de Ligação , Bupropiona/farmacologia , Antagonistas Nicotínicos/química , Antagonistas Nicotínicos/metabolismo , Antagonistas Nicotínicos/farmacologia , Marcadores de Fotoafinidade/química , Marcadores de Fotoafinidade/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína
13.
J Biol Chem ; 286(40): 34635-42, 2011 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-21844195

RESUMO

Prokaryotic members of the Cys-loop receptor ligand-gated ion channel superfamily were recently identified. Previously, Cys-loop receptors were only known from multicellular organisms (metazoans). Contrary to the metazoan Cys-loop receptors, the prokaryotic ones consist of an extracellular (ECD) and a transmembrane domain (TMD), lacking the large intracellular domain (ICD) present in metazoa (between transmembrane segments M3 and M4). Using a chimera approach, we added the 115-amino acid ICD from mammalian serotonin type 3A receptors (5-HT(3A)) to the prokaryotic proton-activated Gloeobacter violaceus ligand-gated ion channel (GLIC). We created 12 GLIC-5-HT(3A)-ICD chimeras by replacing a variable number of amino acids in the short GLIC M3M4 linker with the entire 5-HT(3A)-ICD. Two-electrode voltage clamp recordings after expression in Xenopus laevis oocytes showed that only two chimeras were functional and produced currents upon acidification. The pH(50) was comparable with wild-type GLIC. 5-HT(3A) receptor expression can be inhibited by the chaperone protein RIC-3. We have shown previously that the 5-HT(3A)-ICD is required for the attenuation of 5-HT-induced currents when RIC-3 is co-expressed with 5-HT(3A) receptors in X. laevis oocytes. Expression of both functional 5-HT(3A) chimeras was inhibited by RIC-3 co-expression, indicating appropriate folding of the 5-HT(3A)-ICD in the chimeras. Our results indicate that the ICD can be considered a separate domain that can be removed from or added to the ECD and TMD while maintaining the overall structure and function of the ECD and TMD.


Assuntos
Cisteína/química , Aminoácidos/química , Animais , Cianobactérias/metabolismo , Escherichia coli/metabolismo , Cavalos , Concentração de Íons de Hidrogênio , Íons , Ligantes , Bicamadas Lipídicas/metabolismo , Oócitos/metabolismo , Técnicas de Patch-Clamp , Plasmídeos/metabolismo , Conformação Proteica , Engenharia de Proteínas/métodos , Estrutura Terciária de Proteína , Receptores 5-HT3 de Serotonina/química , Xenopus laevis
14.
J Neurosci ; 29(10): 3083-92, 2009 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-19279245

RESUMO

The molecular basis of general anesthetic interactions with GABA(A) receptors is uncertain. An accurate homology model would facilitate studies of anesthetic action. Construction of a GABA(A) model based on the 4 A resolution acetylcholine receptor structure is complicated by alignment uncertainty between the acetylcholine and GABA(A) receptor M3 and M4 transmembrane segments. Using disulfide crosslinking we previously established the orientation of M2 and M3 within a single GABA(A) subunit. The resultant model predicts that the betaM3 residue beta2M286, implicated in anesthetic binding, faces the adjacent alpha1-M1 segment and not into the beta2 subunit interior as some models have suggested. To assess the proximity of beta2M286 to the alpha1-M1 segment we expressed beta2M286C and gamma2 with 10 consecutive alpha1-M1 cysteine (Cys) mutants, alpha1I223C to alpha1L232C, in and flanking the extracellular end of alpha1-M1. In activated states, beta2M286C formed disulfide bonds with alpha1Y225C and alpha1Q229C based on electrophysiological assays and dimers on Western blots, but not with other alpha1-M1 mutants. beta2F289, one helical turn below beta2M286, formed disulfide bonds with alpha1I228C, alpha1Q229C and alpha1L232C in activated states. The intervening residues, beta2G287C and beta2C288, did not form disulfide bonds with alpha1-M1 Cys mutants. We conclude that the beta2-M3 residues beta2M286 and beta2F289 face the intersubunit interface in close proximity to alpha1-M1 and that channel gating induces a structural rearrangement in the transmembrane subunit interface that reduces the betaM3 to alphaM1 separation by approximately 7 A. This supports the hypothesis that some intravenous anesthetics bind in the betaM3-alphaM1 subunit interface consistent with azi-etomidate photoaffinity labeling.


Assuntos
Anestésicos Intravenosos/metabolismo , Modelos Moleculares , Conformação Proteica , Subunidades Proteicas/metabolismo , Receptores de GABA-A/metabolismo , Homologia de Sequência de Aminoácidos , Ácido gama-Aminobutírico/metabolismo , Animais , Sítios de Ligação/fisiologia , Feminino , Agonistas de Receptores de GABA-A , Conformação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína/efeitos dos fármacos , Estrutura Terciária de Proteína/fisiologia , Subunidades Proteicas/agonistas , Subunidades Proteicas/química , Ratos , Receptores de GABA-A/química , Xenopus laevis , Ácido gama-Aminobutírico/farmacologia
15.
J Neurosci ; 29(6): 1626-35, 2009 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-19211870

RESUMO

Nicotinic acetylcholine receptors (nAChR) are cation-selective, ligand-gated ion channels of the cysteine (Cys)-loop gene superfamily. The recent crystal structure of a bacterial homolog from Erwinia chrysanthemi (ELIC) agrees with previous structures of the N-terminal domain of AChBP (acetylcholine-binding protein) and of the electron-microscopy-derived Torpedo nAChR structure. However, the ELIC transmembrane domain is significantly more tightly packed than the corresponding region of the Torpedo nAChR. We investigated the tightness of protein packing surrounding the extracellular end of the M2 transmembrane segment and around the loop connecting the M2 and M3 segments using the substituted cysteine accessibility method. The M2 20' to 27' residues were highly water accessible and the variation in reaction rates were consistent with this region being alpha-helical. At all positions tested, the presence of ACh changed methanethiosulfonate ethylammonium (MTSEA) modification rates by <10-fold. In the presence of ACh, reaction rates for residues in the last extracellular alpha-helical turn of M2 and in the M2M3 loop increased, whereas rates in the penultimate alpha-helical turn of M2 decreased. Only three of eight M2M3 loop residues were accessible to MTSEA in both the presence and absence of ACh. We infer that the protein packing around the M2M3 loop is tight, consistent with its location at the interdomain interface where it is involved in the transduction of ligand binding in the extracellular domain to gating in the transmembrane domain. Our data indicate that the Torpedo nAChR transmembrane domain structure is a better model than the ELIC structure for eukaryotic Cys-loop receptors.


Assuntos
Receptores Nicotínicos/química , Receptores Nicotínicos/genética , Sequência de Aminoácidos , Animais , Feminino , Camundongos , Dados de Sequência Molecular , Estrutura Secundária de Proteína/genética , Estrutura Terciária de Proteína/genética , Receptores Nicotínicos/metabolismo , Torpedo/genética , Xenopus laevis
16.
J Med Chem ; 51(15): 4430-48, 2008 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-18651727

RESUMO

Drugs used to treat various disorders target GABA A receptors. To develop alpha subunit selective compounds, we synthesized 5-(4-piperidyl)-3-isoxazolol (4-PIOL) derivatives. The 3-isoxazolol moiety was substituted by 1,3,5-oxadiazol-2-one, 1,3,5-oxadiazol-2-thione, and substituted 1,2,4-triazol-3-ol heterocycles with modifications to the basic piperidine substituent as well as substituents without basic nitrogen. Compounds were screened by [(3)H]muscimol binding and in patch-clamp experiments with heterologously expressed GABA A alpha ibeta 3gamma 2 receptors (i = 1-6). The effects of 5-aminomethyl-3 H-[1,3,4]oxadiazol-2-one 5d were comparable to GABA for all alpha subunit isoforms. 5-piperidin-4-yl-3 H-[1,3,4]oxadiazol-2-one 5a and 5-piperidin-4-yl-3 H-[1,3,4]oxadiazol-2-thione 6a were weak agonists at alpha 2-, alpha 3-, and alpha 5-containing receptors. When coapplied with GABA, they were antagonistic in alpha 2-, alpha 4-, and alpha 6-containing receptors and potentiated alpha 3-containing receptors. 6a protected GABA binding site cysteine-substitution mutants alpha 1F64C and alpha 1S68C from reacting with methanethiosulfonate-ethylsulfonate. 6a specifically covalently modified the alpha 1R66C thiol, in the GABA binding site, through its oxadiazolethione sulfur. These results demonstrate the feasibility of synthesizing alpha subtype selective GABA mimetic drugs.


Assuntos
Agonistas de Receptores de GABA-A , Receptores de GABA-A/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Sítios de Ligação , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Linhagem Celular , Eletrofisiologia , Feminino , Humanos , Isoxazóis/síntese química , Isoxazóis/química , Isoxazóis/farmacologia , Modelos Moleculares , Estrutura Molecular , Muscimol/síntese química , Muscimol/química , Muscimol/farmacologia , Mutação/genética , Oócitos , Técnicas de Patch-Clamp , Piperidinas/síntese química , Piperidinas/química , Piperidinas/farmacologia , Subunidades Proteicas/agonistas , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Ratos , Receptores de GABA-A/química , Receptores de GABA-A/genética , Relação Estrutura-Atividade , Xenopus laevis
17.
Mol Pharmacol ; 74(3): 854-62, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18524888

RESUMO

The reduced folate carrier (RFC) and the proton-coupled folate transporter (PCFT) are ubiquitously expressed in normal and malignant mammalian tissues and in human solid tumor cell lines. This article addresses the extent to which PCFT contributes to transport of pemetrexed and to the activities of this and other antifolates relative to RFC at physiological pH. Either RFC or PCFT cDNA was stably transfected into a transporter-null HeLa cell variant to achieve activities similar to their endogenous function in wild-type HeLa cells. PCFT and RFC produced comparable increases in pemetrexed activity in growth medium with 5-formyltetrahydrofolate. However, PCFT had little or no effect on the activities of methotrexate, N-(5-[N-(3,4-dihydro-2-methyl-4-oxyquinazolin-6-ylmethyl)-N-methyl-amino]-2-thenoyl)-l-glutamic acid (raltitrexed, Tomudex; ZD1694), or N(alpha)-(4-amino-4-deoxypteroyl)-N(delta)-hemiphthaloyl-l-ornithine (PT523) in comparison with RFC irrespective of the folate growth source. PCFT, expressed at high levels in Xenopus laevis oocytes and in transporter-competent HepG2 cells, exhibited a high affinity for pemetrexed, with an influx K(m) value of 0.2 to 0.8 muM at pH 5.5. PCFT increased the growth inhibitory activity of pemetrexed, but not that of the other antifolates in HepG2 cells grown with 5-formyltetrahydrofolate at physiological pH. These findings illustrate the unique role that PCFT plays in the transport and pharmacological activity of pemetrexed. Because of the ubiquitous expression of PCFT in human tumors, and the ability of PCFT to sustain pemetrexed activity even in the absence of RFC, tumor cells are unlikely to become resistant to pemetrexed as a result of impaired transport because of the redundancy of these genetically distinct routes.


Assuntos
Antagonistas do Ácido Fólico/metabolismo , Glutamatos/metabolismo , Guanina/análogos & derivados , Proteínas de Membrana Transportadoras/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Clonais , Antagonistas do Ácido Fólico/farmacologia , Guanina/metabolismo , Células HeLa , Humanos , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Espaço Intracelular/metabolismo , Cinética , Pemetrexede , Prótons , Proteína Carregadora de Folato Reduzido , Transfecção , Trítio , Xenopus
18.
Cell ; 127(5): 917-28, 2006 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-17129779

RESUMO

Folates are essential nutrients that are required for one-carbon biosynthetic and epigenetic processes. While folates are absorbed in the acidic milieu of the upper small intestine, the underlying absorption mechanism has not been defined. We now report the identification of a human proton-coupled, high-affinity folate transporter that recapitulates properties of folate transport and absorption in intestine and in various cell types at low pH. We demonstrate that a loss-of-function mutation in this gene is the molecular basis for hereditary folate malabsorption in a family with this disease. This transporter was previously reported to be a lower-affinity, pH-independent heme carrier protein, HCP1. However, the current study establishes that a major function of this gene product is proton-coupled folate transport required for folate homeostasis in man, and we have thus amended the name to PCFT/HCP1.


Assuntos
Ácido Fólico/metabolismo , Mucosa Intestinal/metabolismo , Síndromes de Malabsorção/genética , Proteínas de Membrana Transportadoras/metabolismo , Animais , Transporte Biológico , Células CACO-2 , Linhagem Celular Tumoral , Eletrofisiologia , Ácido Fólico/química , Regulação da Expressão Gênica , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Cinética , Proteínas de Membrana Transportadoras/genética , Oócitos , Linhagem , Transportador de Folato Acoplado a Próton , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Especificidade por Substrato , Xenopus
19.
J Neurosci ; 26(17): 4492-9, 2006 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-16641228

RESUMO

Construction of a GABAA receptor homology model based on the acetylcholine (ACh) receptor structure is complicated by the low sequence similarity between GABAA and ACh M3 transmembrane segments that creates significant uncertainty in their alignment. We determined the orientation of the GABAA M2 and M3 transmembrane segments using disulfide cross-linking. The M2 residues alpha1M266 (11') and alpha1T267 (12') were mutated to cysteine in either wild type or single M3 cysteine mutant (alpha1V297C, alpha1A300C to alpha1A305C) backgrounds. We assayed spontaneous and induced disulfide bond formation. Reduction with DTT significantly potentiated GABA-induced currents in alpha1T267C-L301C and alpha1T267C-F304C. Copper phenanthroline-induced oxidation inhibited GABA-induced currents in these mutants and in alpha1T267C-A305C. Intrasubunit disulfide bonds formed between these Cys pairs, implying that the alpha-carbon separation was at most 5.6 A. The reactive alpha1M3 residues (L301, F304, A305) lie on the same face of an alpha-helix. The unresponsive ones (A300, I302, E303) lie on the opposite face. In the resting state, the reactive side of alpha1M3 faces M2-alpha1T267. In conjunction with the ACh structure, our data indicate that alignment of GABAA and ACh M3 requires a single gap in the GABAA M2-M3 loop. In the presence of GABA, oxidation of alpha1T267C-L301C and alpha1T267C-F304C had no effect, but oxidation of alpha1T267C-A305C caused a significant increase in spontaneous channel opening. We infer that, as the channel opens, the distance and/or orientation between M2-alpha1T267 and M3-alpha1A305 changes such that the disulfide bond stabilizes the open state. This begins to define the conformational motion that M2 undergoes during channel opening.


Assuntos
Oócitos/metabolismo , Receptores Colinérgicos/química , Receptores Colinérgicos/metabolismo , Receptores de GABA-A/química , Receptores de GABA-A/metabolismo , Alinhamento de Sequência , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células Cultivadas , Reagentes de Ligações Cruzadas/química , Reagentes de Ligações Cruzadas/metabolismo , Dissulfetos/química , Dissulfetos/metabolismo , Dados de Sequência Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Xenopus laevis
20.
Curr Med Chem ; 12(24): 2805-47, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16305474

RESUMO

It is known that DNA is a well-characterized intracellular target but its size and sequential characteristics make it an elusive target for selective drug action. Binding of low molecular weight ligands to DNA causes a variety of significant biological responses. In this context the main consideration is given to recent developments in DNA sequence selective binding agents bearing conjugated effectors because of their potential application in treatment of cancers, in diagnosis as well as in molecular biology. In the present review recent results about analogues of netropsins, distamycin A and of some lexitropsins and combilexins or related hybrid molecules with sequence reading, intercalating or alkylating activity are described and evaluated for prospective applications. Furthermore there exists DNA minor groove binder with different basic structures which does not possess the typical polyamide chain, including dimeric intercalating chromophores. Finally new results about peptide nucleic acids and related nucleic acid bases linked with polyamides are reported. In pronounced examples the structural chemistry, synthesis, DNA binding with several biophysical methods, molecular aspects, structure activity relationship, topoisomerase inhibition, antitumour and antibacterial effects are discussed in detail.


Assuntos
Alquilantes/química , DNA/química , Substâncias Intercalantes/química , Ligantes , Alquilantes/metabolismo , Sítios de Ligação , DNA/metabolismo , Desenho de Fármacos , Substâncias Intercalantes/metabolismo , Modelos Moleculares , Estrutura Molecular , Conformação de Ácido Nucleico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA