Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Biophys J ; 123(14): 2185-2198, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-38678367

RESUMEN

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.


Asunto(s)
Bupropión , Cianobacterias , Canales Iónicos Activados por Ligandos , Bupropión/farmacología , Bupropión/química , Bupropión/metabolismo , Canales Iónicos Activados por Ligandos/metabolismo , Canales Iónicos Activados por Ligandos/química , Sitios de Unión , Cianobacterias/metabolismo , Simulación del Acoplamiento Molecular , Secuencia de Aminoácidos
2.
Biophys J ; 119(12): 2593-2603, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33157122

RESUMEN

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.


Asunto(s)
Receptores de Serotonina 5-HT3 , Serotonina , Canales Iónicos , Iones , Conformación Proteica en Hélice alfa , Receptores de Serotonina 5-HT3/genética , Receptores de Serotonina 5-HT3/metabolismo
3.
Biophys J ; 118(4): 934-943, 2020 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-31870537

RESUMEN

The serotonin type 3A (5-HT3A) receptor is a homopentameric cation-selective member of the pentameric ligand-gated ion channel (pLGIC) superfamily. Members of this superfamily assemble from five subunits, each of which consists of three domains: extracellular (ECD), transmembrane (TMD), and intracellular domain (ICD). Previously, we have demonstrated that the 5-HT3A-ICD is required for the interaction between 5-HT3A and the chaperone protein resistance to inhibitors of choline esterase (RIC-3). Additionally, we have shown that 5-HT3A-ICD fused to maltose-binding protein (MBP) directly interacts with RIC-3, without the involvement of other protein(s). To elucidate the molecular determinants of this interaction, we developed different MBP-fused 5-HT3A-ICD constructs by deleting large segments of its amino acid sequence. We expressed seven engineered ICDs in Escherichia coli and purified them to homogeneity. Using a RIC-3 affinity pull-down assay, the interaction between MBP-5HT3A-ICD constructs and RIC-3 was investigated. In summary, we identify a 24-amino-acid-long segment of the 5-HT3A-ICD as a molecular determinant for the interaction between the 5-HT3A-ICD and RIC-3.


Asunto(s)
Receptores de Serotonina 5-HT3 , Serotonina , Secuencia de Aminoácidos , Escherichia coli/genética , Escherichia coli/metabolismo , Chaperonas Moleculares/genética , Receptores de Serotonina 5-HT3/genética , Receptores de Serotonina 5-HT3/metabolismo
4.
Mol Pharmacol ; 97(3): 171-179, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31871303

RESUMEN

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.


Asunto(s)
Bupropión/metabolismo , Bupropión/farmacología , Receptores de Serotonina 5-HT3/metabolismo , Antagonistas del Receptor de Serotonina 5-HT3/metabolismo , Antagonistas del Receptor de Serotonina 5-HT3/farmacología , Secuencia de Aminoácidos , Animales , Inhibidores de Captación de Dopamina/metabolismo , Inhibidores de Captación de Dopamina/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Ratones , Receptores de Serotonina 5-HT3/genética , Estereoisomerismo , Xenopus laevis
6.
Protein Expr Purif ; 153: 45-52, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30130580

RESUMEN

The main principles of higher-order protein oligomerization are elucidated by many structural and biophysical studies. An astonishing number of proteins self-associate to form dimers or higher-order quaternary structures which further interact with other biomolecules to elicit complex cellular responses. In this study, we describe a simple and convenient approach to determine the oligomeric state of purified protein complexes that combines implementation of a novel form of clear-native gel electrophoresis and size exclusion chromatography in line with multi-angle light scattering. Here, we demonstrate the accuracy of this ensemble approach by characterizing the previously established pentameric state of the intracellular domain of serotonin type 3A (5-HT3A) receptors.


Asunto(s)
Electroforesis en Gel de Poliacrilamida/métodos , Proteínas de Unión a Maltosa/química , Receptores de Serotonina 5-HT3/química , Proteínas Recombinantes de Fusión/química , Clonación Molecular , Densitometría , Electroforesis en Gel de Poliacrilamida/instrumentación , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Luz , Proteínas de Unión a Maltosa/genética , Proteínas de Unión a Maltosa/metabolismo , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Receptores de Serotonina 5-HT3/genética , Receptores de Serotonina 5-HT3/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Dispersión de Radiación
7.
J Neurochem ; 137(4): 528-38, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26875553

RESUMEN

Pentameric ligand-gated ion channels (pLGIC) are expressed in both excitable and non-excitable cells that are targeted by numerous clinically used drugs. Assembly from five identical or homologous subunits yields homo- or heteromeric pentamers, respectively. The protein known as Resistance to Inhibitors of Cholinesterase (RIC-3) was identified to interfere with assembly and functional maturation of pLGICs. We have shown previously for serotonin type 3A homopentamers (5-HT3A ) that the interaction with RIC-3 requires the intracellular domain (ICD) of this pLGIC. After expression in Xenopus laevis oocytes RIC-3 attenuated serotonin-induced currents in 5-HT3A wild-type channels, but not in functional 5-HT3A glvM3M4 channels that have the 115-amino acid ICD replaced by a heptapeptide. In complementary experiments we have shown that engineering the Gloeobacter violaceus ligand-gated ion channel (GLIC) to contain the 5-HT3A -ICD confers sensitivity to RIC-3 in oocytes to otherwise insensitive GLIC. In this study, we identify endogenous RIC-3 protein expression in X. laevis oocytes. We purified RIC-3 to homogeneity after expression in Echericia coli. By using heterologously over-expressed and purified RIC-3 and the chimera consisting of the 5-HT3A -ICD and the extracellular and transmembrane domains of GLIC in pull-down experiments, we demonstrate a direct and specific interaction between the two proteins. This result further underlines that the domain within 5-HT3 A R that mediates the interaction with RIC-3 is the ICD. Importantly, this is the first experimental evidence that the interaction between 5-HT3 A R-ICD and RIC-3 does not require other proteins. In addition, we demonstrate that the pentameric assembly of the GLIC-5-HT3A -ICD chimera interacts with RIC-3. We hypothesized that pentameric ligand-gated ion channels (pLGICs) associate directly with the chaperone protein RIC-3 (resistance to inhibitors of cholinesterase type 3), and that the interaction does not require other protein factors. We found that the two proteins indeed interact directly, that the pLGIC intracellular domain is required for the effect, and that pLGICs in their pentameric form associate with RIC-3. These results provide the basis for future studies aimed at investigating which motifs provide the interaction surfaces, and at delineating the mechanism(s) of RIC-3 modulation of functional pLGIC surface expression.


Asunto(s)
Citoplasma/genética , Citoplasma/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Receptores de Serotonina 5-HT3/genética , Receptores de Serotonina 5-HT3/metabolismo , Animales , Humanos , Oocitos , Unión Proteica/fisiología , Xenopus laevis
8.
Biochemistry ; 54(16): 2670-2682, 2015 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-25861708

RESUMEN

Pentameric ligand-gated ion channels (pLGICs), also called Cys-loop receptors in eukaryotic superfamily members, play diverse roles in neurotransmission and serve as primary targets for many therapeutic drugs. Structural studies of full-length eukaryotic pLGICs have been challenging because of glycosylation, large size, pentameric assembly, and hydrophobicity. X-ray structures of prokaryotic pLGICs, including the Gloeobacter violaceus LGIC (GLIC) and the Erwinia chrysanthemi LGIC (ELIC), and truncated eukaryotic pLGICs have significantly improved and complemented the understanding of structural details previously obtained with acetylcholine-binding protein and Torpedo nicotinic acetylcholine receptors. Prokaryotic pLGICs share their overall structural features with eukaryotic pLGICs for the ligand-binding extracellular and channel-lining transmembrane domains. The large intracellular domain (ICD) is present only in eukaryotic members and is characterized by a low level of sequence conservation and significant variability in length (50-250 amino acids), making the ICD a potential target for the modulation of specific pLGIC subunits. None of the structures includes a complete ICD. Here, we created chimeras by adding the ICD of cation-conducting (nAChR-α7) and anion-conducting (GABAρ1, Glyα1) eukaryotic homopentamer-forming pLGICs to GLIC. GLIC-ICD chimeras assemble into pentamers to form proton-gated channels, as does the parent GLIC. Additionally, the sensitivity of the chimeras toward modulation of functional maturation by chaperone protein RIC-3 is preserved as in those of the parent eukaryotic channels. For a previously described GLIC-5HT3A-ICD chimera, we now provide evidence of its successful large-scale expression and purification to homogeneity. Overall, the chimeras provide valuable tools for functional and structural studies of eukaryotic pLGIC ICDs.


Asunto(s)
Proteínas Bacterianas/química , Dickeya chrysanthemi/química , Proteínas de Peces/química , Proteínas Recombinantes de Fusión/química , Torpedo , Receptor Nicotínico de Acetilcolina alfa 7/química , Animales , Proteínas Bacterianas/genética , Dickeya chrysanthemi/genética , Proteínas de Peces/genética , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Receptor Nicotínico de Acetilcolina alfa 7/genética
9.
J Biol Chem ; 288(16): 11294-303, 2013 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-23463505

RESUMEN

Although the activity of the nicotinic acetylcholine receptor (nAChR) is exquisitely sensitive to its membrane environment, the underlying mechanisms remain poorly defined. The homologous prokaryotic pentameric ligand-gated ion channel, Gloebacter ligand-gated ion channel (GLIC), represents an excellent model for probing the molecular basis of nAChR sensitivity because of its high structural homology, relative ease of expression, and amenability to crystallographic analysis. We show here that membrane-reconstituted GLIC exhibits structural and biophysical properties similar to those of the membrane-reconstituted nAChR, although GLIC is substantially more thermally stable. GLIC, however, does not possess the same exquisite lipid sensitivity. In particular, GLIC does not exhibit the same propensity to adopt an uncoupled conformation where agonist binding is uncoupled from channel gating. Structural comparisons provide insight into the chemical features that may predispose the nAChR to the formation of an uncoupled state.


Asunto(s)
Bacterias , Proteínas Bacterianas , Activación del Canal Iónico/fisiología , Canales Iónicos , Bacterias/química , Bacterias/genética , Bacterias/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Canales Iónicos/química , Canales Iónicos/genética , Canales Iónicos/metabolismo , Estabilidad Proteica , Estructura Cuaternaria de Proteína , Homología Estructural de Proteína
10.
J Neurochem ; 125(6): 843-54, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23565737

RESUMEN

Nicotinic acetylcholine receptors (nAChR) are members of the Cys-loop ligand-gated ion channel superfamily. Muscle nAChR are heteropentamers that assemble from two α, and one each of ß, γ, and δ subunits. Each subunit is composed of three domains, extracellular, transmembrane and intracellular. The transmembrane domain consists of four α-helical segments (M1-M4). Pioneering structural information was obtained using electronmicroscopy of Torpedo nAChR. The recently solved X-ray structure of the first eukaryotic Cys-loop receptor, a truncated (intracellular domain missing) glutamate-gated chloride channel α (GluClα) showed the same overall architecture. However, a significant difference with regard to the vertical alignment between the channel-lining segment M2 and segment M3 was observed. Here, we used functional studies utilizing disulfide trapping experiments in muscle nAChR to determine the spatial orientation between M2 and M3. Our results are in agreement with the vertical alignment as obtained when using the GluClα structure as a template to homology model muscle nAChR, however, they cannot be reconciled with the current Torpedo nAChR model. The vertical M2-M3 alignments as observed in X-ray structures of prokaryotic Gloeobacter violaceus ligand-gated ion channel and GluClα are in agreement. Our results further confirm that this alignment in Cys-loop receptors is conserved between prokaryotes and eukaryotes.


Asunto(s)
Receptores Nicotínicos/química , Animales , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Canales de Cloruro/química , Canales de Cloruro/genética , Disulfuros/química , Femenino , Ratones , Músculos/metabolismo , Mutación , Oocitos/metabolismo , Técnicas de Placa-Clamp , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptores Nicotínicos/genética , Receptores Nicotínicos/fisiología , Especificidad de la Especie , Torpedo , Xenopus laevis
11.
J Gen Physiol ; 155(6)2023 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-37026993

RESUMEN

Serotonin or 5-hydroxytryptamine type 3 (5-HT3) receptors belong to the family of pentameric ligand-gated ion channels (pLGICs) that are therapeutic targets for psychiatric disorders and neurological diseases. Due to structural conservation and significant sequence similarities of pLGICs' extracellular and transmembrane domains, clinical trials for drug candidates targeting these two domains have been hampered by off-subunit modulation. With the present study, we explore the interaction interface of the 5-HT3A subunit intracellular domain (ICD) with the resistance to inhibitors of choline esterase (RIC-3) protein. Previously, we have shown that RIC-3 interacts with the L1-MX segment of the ICD fused to maltose-binding protein. In the present study, synthetic L1-MX-based peptides and Ala-scanning identify positions W347, R349, and L353 as critical for binding to RIC-3. Complementary studies using full-length 5-HT3A subunits confirm that the identified Ala substitutions reduce the RIC-3-mediated modulation of functional surface expression. Additionally, we find and characterize a duplication of the binding motif, DWLR…VLDR, present in both the MX-helix and the transition between the ICD MA-helix and transmembrane segment M4. Analogous Ala substitutions at W447, R449, and L454 disrupt MAM4-peptide RIC-3 interactions and reduce modulation of functional surface expression. In summary, we identify the binding motif for RIC-3 in 5-HT3A subunits at two locations in the ICD, one in the MX-helix and one at the MAM4-helix transition.


Asunto(s)
Receptores de Serotonina 5-HT3 , Serotonina , Humanos , Receptores de Serotonina 5-HT3/genética , Receptores de Serotonina 5-HT3/química , Dominios Proteicos
12.
bioRxiv ; 2023 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-37873398

RESUMEN

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.

13.
Biochemistry ; 51(12): 2425-35, 2012 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-22394379

RESUMEN

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.


Asunto(s)
Azidas/metabolismo , Bupropión/análogos & derivados , Bupropión/metabolismo , Membrana Celular/metabolismo , Receptores Nicotínicos/química , Receptores Nicotínicos/metabolismo , Torpedo , Animales , Antidepresivos/química , Antidepresivos/metabolismo , Antidepresivos/farmacología , Sitios de Unión , Bupropión/farmacología , Antagonistas Nicotínicos/química , Antagonistas Nicotínicos/metabolismo , Antagonistas Nicotínicos/farmacología , Etiquetas de Fotoafinidad/química , Etiquetas de Fotoafinidad/metabolismo , Unión Proteica , Estructura Terciaria de Proteína
14.
J Biol Chem ; 286(40): 34635-42, 2011 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-21844195

RESUMEN

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.


Asunto(s)
Cisteína/química , Aminoácidos/química , Animales , Cianobacterias/metabolismo , Escherichia coli/metabolismo , Caballos , Concentración de Iones de Hidrógeno , Iones , Ligandos , Membrana Dobles de Lípidos/metabolismo , Oocitos/metabolismo , Técnicas de Placa-Clamp , Plásmidos/metabolismo , Conformación Proteica , Ingeniería de Proteínas/métodos , Estructura Terciaria de Proteína , Receptores de Serotonina 5-HT3/química , Xenopus laevis
15.
Methods Mol Biol ; 2507: 425-444, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35773596

RESUMEN

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.


Asunto(s)
Deficiencia de Ácido Fólico , Transportador de Folato Acoplado a Protón , Animales , Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/metabolismo , Lípidos , Mamíferos/metabolismo , Mutación , Transportador de Folato Acoplado a Protón/química , Transportador de Folato Acoplado a Protón/genética , Transportador de Folato Acoplado a Protón/metabolismo
16.
PLoS One ; 16(11): e0253184, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34793461

RESUMEN

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.


Asunto(s)
Microdominios de Membrana/metabolismo , Transportador de Folato Acoplado a Protón/metabolismo , Transporte Biológico , Membrana Celular/metabolismo , Humanos , Lípidos
17.
J Neurosci ; 29(6): 1626-35, 2009 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-19211870

RESUMEN

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.


Asunto(s)
Receptores Nicotínicos/química , Receptores Nicotínicos/genética , Secuencia de Aminoácidos , Animales , Femenino , Ratones , Datos de Secuencia Molecular , Estructura Secundaria de Proteína/genética , Estructura Terciaria de Proteína/genética , Receptores Nicotínicos/metabolismo , Torpedo/genética , Xenopus laevis
18.
J Neurosci ; 29(10): 3083-92, 2009 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-19279245

RESUMEN

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.


Asunto(s)
Anestésicos Intravenosos/metabolismo , Modelos Moleculares , Conformación Proteica , Subunidades de Proteína/metabolismo , Receptores de GABA-A/metabolismo , Homología de Secuencia de Aminoácido , Ácido gamma-Aminobutírico/metabolismo , Animales , Sitios de Unión/fisiología , Femenino , Agonistas de Receptores de GABA-A , Conformación Proteica/efectos de los fármacos , Estructura Terciaria de Proteína/efectos de los fármacos , Estructura Terciaria de Proteína/fisiología , Subunidades de Proteína/agonistas , Subunidades de Proteína/química , Ratas , Receptores de GABA-A/química , Xenopus laevis , Ácido gamma-Aminobutírico/farmacología
19.
J Gen Physiol ; 131(2): 137-46, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18227272

RESUMEN

Cys-loop receptor neurotransmitter-gated ion channels are pentameric assemblies of subunits that contain three domains: extracellular, transmembrane, and intracellular. The extracellular domain forms the agonist binding site. The transmembrane domain forms the ion channel. The cytoplasmic domain is involved in trafficking, localization, and modulation by cytoplasmic second messenger systems but its role in channel assembly and function is poorly understood and little is known about its structure. The intracellular domain is formed by the large (>100 residues) loop between the alpha-helical M3 and M4 transmembrane segments. Putative prokaryotic Cys-loop homologues lack a large M3M4 loop. We replaced the complete M3M4 loop (115 amino acids) in the 5-hydroxytryptamine type 3A (5-HT(3A)) subunit with a heptapeptide from the prokaryotic homologue from Gloeobacter violaceus. The macroscopic electrophysiological and pharmacological characteristics of the homomeric 5-HT(3A)-glvM3M4 receptors were comparable to 5-HT(3A) wild type. The channels remained cation-selective but the 5-HT(3A)-glvM3M4 single channel conductance was 43.5 pS as compared with the subpicosiemens wild-type conductance. Coexpression of hRIC-3, a protein that modulates expression of 5-HT(3) and acetylcholine receptors, significantly attenuated 5-HT-induced currents with wild-type 5-HT(3A) but not 5-HT(3A)-glvM3M4 receptors. A similar deletion of the M3M4 loop in the anion-selective GABA-rho1 receptor yielded functional, GABA-activated, anion-selective channels. These results imply that the M3M4 loop is not essential for receptor assembly and function and suggest that the cytoplasmic domain may fold as an independent module from the transmembrane and extracellular domains.


Asunto(s)
Mutación , Receptores de GABA-B/fisiología , Receptores de Serotonina 5-HT3/fisiología , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Línea Celular , Cianobacterias/genética , Diltiazem/farmacología , Femenino , Antagonistas de Receptores de GABA-B , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Lidocaína/análogos & derivados , Lidocaína/farmacología , Potenciales de la Membrana/efectos de los fármacos , Ratones , Datos de Secuencia Molecular , Ondansetrón/farmacología , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Oocitos/fisiología , Técnicas de Placa-Clamp , Picrotoxina/farmacología , Receptores de GABA-B/genética , Receptores de Serotonina 5-HT3/genética , Homología de Secuencia de Aminoácido , Serotonina/farmacología , Antagonistas del Receptor de Serotonina 5-HT3 , Cloruro de Sodio/farmacología , Xenopus laevis , Ácido gamma-Aminobutírico/farmacología
20.
J Gen Physiol ; 151(9): 1135-1145, 2019 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-31409663

RESUMEN

Serotonin type 3 receptors (5-HT3Rs) are cation-conducting pentameric ligand-gated ion channels and members of the Cys-loop superfamily in eukaryotes. 5-HT3Rs are found in the peripheral and central nervous system, and they are targets for drugs used to treat anxiety, drug dependence, and schizophrenia, as well as chemotherapy-induced and postoperative nausea and emesis. Decades of research of Cys-loop receptors have identified motifs in both the extracellular and transmembrane domains that mediate pentameric assembly. Those efforts have largely ignored the most diverse domain of these channels, the intracellular domain (ICD). Here we identify molecular determinants within the ICD of serotonin type 3A (5-HT3A) subunits for pentameric assembly by first identifying the segments contributing to pentamerization using deletion constructs of, and finally by making defined amino acid substitutions within, an isolated soluble ICD. Our work provides direct experimental evidence for the contribution of three intracellular arginines, previously implicated in governing the low conductance of 5-HT3ARs, in structural features such as pentameric assembly.


Asunto(s)
Arginina/química , Receptores de Serotonina 5-HT3/química , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Escherichia coli , Regulación de la Expresión Génica , Ratones , Conformación Proteica , Dominios Proteicos , Ingeniería de Proteínas , Pliegue de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA