Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 2476, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38509088

RESUMEN

Cellular entry of the hepatitis B and D viruses (HBV/HDV) requires binding of the viral surface polypeptide preS1 to the hepatobiliary transporter Na+-taurocholate co-transporting polypeptide (NTCP). This interaction can be blocked by bulevirtide (BLV, formerly Myrcludex B), a preS1 derivative and approved drug for treating HDV infection. Here, to elucidate the basis of this inhibitory function, we determined a cryo-EM structure of BLV-bound human NTCP. BLV forms two domains, a plug lodged in the bile salt transport tunnel of NTCP and a string that covers the receptor's extracellular surface. The N-terminally attached myristoyl group of BLV interacts with the lipid-exposed surface of NTCP. Our structure reveals how BLV inhibits bile salt transport, rationalizes NTCP mutations that decrease the risk of HBV/HDV infection, and provides a basis for understanding the host specificity of HBV/HDV. Our results provide opportunities for structure-guided development of inhibitors that target HBV/HDV docking to NTCP.


Asunto(s)
Hepatitis B , Lipopéptidos , Simportadores , Humanos , Virus de la Hepatitis B/fisiología , Antivirales/uso terapéutico , Receptores Virales/metabolismo , Ácidos y Sales Biliares/metabolismo , Virus de la Hepatitis Delta/fisiología , Simportadores/metabolismo , Internalización del Virus , Hepatocitos/metabolismo
2.
Nat Commun ; 14(1): 7296, 2023 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-37949847

RESUMEN

BSEP (ABCB11) is an ATP-binding cassette transporter that is expressed in hepatocytes and extrudes bile salts into the canaliculi of the liver. BSEP dysfunction, caused by mutations or induced by drugs, is frequently associated with severe cholestatic liver disease. We report the cryo-EM structure of glibenclamide-bound human BSEP in nanodiscs, revealing the basis of small-molecule inhibition. Glibenclamide binds the apex of a central binding pocket between the transmembrane domains, preventing BSEP from undergoing conformational changes, and thus rationalizing the reduced uptake of bile salts. We further report two high-resolution structures of BSEP trapped in distinct nucleotide-bound states by using a catalytically inactivated BSEP variant (BSEPE1244Q) to visualize a pre-hydrolysis state, and wild-type BSEP trapped by vanadate to visualize a post-hydrolysis state. Our studies provide structural and functional insight into the mechanism of bile salt extrusion and into small-molecule inhibition of BSEP, which may rationalize drug-induced liver toxicity.


Asunto(s)
Colestasis , Gliburida , Humanos , Transportadoras de Casetes de Unión a ATP/metabolismo , Ácidos y Sales Biliares/metabolismo , Colestasis/metabolismo , Gliburida/metabolismo , Gliburida/farmacología , Hígado/metabolismo
3.
Nat Commun ; 14(1): 5774, 2023 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-37723174

RESUMEN

The organic anion transporting polypeptides OATP1B1 and OATP1B3 are membrane proteins that mediate uptake of drugs into the liver for subsequent conjugation and biliary excretion, a key step in drug elimination from the human body. Polymorphic variants of these transporters can cause reduced drug clearance and adverse drug effects such as statin-induced rhabdomyolysis, and co-administration of OATP substrates can lead to damaging drug-drug interaction. Despite their clinical relevance in drug disposition and pharmacokinetics, the structure and mechanism of OATPs are unknown. Here we present cryo-EM structures of human OATP1B1 and OATP1B3 bound to synthetic Fab fragments and in functionally distinct states. A single estrone-3-sulfate molecule is bound in a pocket located in the C-terminal half of OATP1B1. The shape and chemical nature of the pocket rationalize the preference for diverse organic anions and allow in silico docking of statins. The structure of OATP1B3 is determined in a drug-free state but reveals a bicarbonate molecule bound to the conserved signature motif and a histidine residue that is prevalent in OATPs exhibiting pH-dependent activity.


Asunto(s)
Transportador 1 de Anión Orgánico Específico del Hígado , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos , Humanos , Transporte Biológico , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Proteínas de Transporte de Membrana , Transportador 1 de Anión Orgánico Específico del Hígado/química , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos/química
4.
Nat Commun ; 14(1): 4368, 2023 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-37474505

RESUMEN

The host proteins SERINC3 and SERINC5 are HIV-1 restriction factors that reduce infectivity when incorporated into the viral envelope. The HIV-1 accessory protein Nef abrogates incorporation of SERINCs via binding to intracellular loop 4 (ICL4). Here, we determine cryoEM maps of full-length human SERINC3 and an ICL4 deletion construct, which reveal that hSERINC3 is comprised of two α-helical bundles connected by a ~ 40-residue, highly tilted, "crossmember" helix. The design resembles non-ATP-dependent lipid transporters. Consistently, purified hSERINCs reconstituted into proteoliposomes induce flipping of phosphatidylserine (PS), phosphatidylethanolamine and phosphatidylcholine. Furthermore, SERINC3, SERINC5 and the scramblase TMEM16F expose PS on the surface of HIV-1 and reduce infectivity, with similar results in MLV. SERINC effects in HIV-1 and MLV are counteracted by Nef and GlycoGag, respectively. Our results demonstrate that SERINCs are membrane transporters that flip lipids, resulting in a loss of membrane asymmetry that is strongly correlated with changes in Env conformation and loss of infectivity.


Asunto(s)
Infecciones por VIH , VIH-1 , Humanos , Proteínas de la Membrana/metabolismo , VIH-1/metabolismo , Factores de Restricción Antivirales , Glicoproteínas de Membrana , Antivirales
6.
Methods Mol Biol ; 2507: 273-294, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35773587

RESUMEN

Structural studies of membrane proteins require high-quality samples. The target proteins should not only be pure and homogeneous but should also be active and allow the capture of a functionally relevant state. Here we present optimized methods for the expression and purification of human ABC transporters and oligosaccharyltransferase (OST) complexes that can be used for high-resolution structure determination using single-particle cryo-electron microscopy (cryo-EM). The protocols are based on the generation of stable cell lines that enable tetracycline-inducible expression of the target proteins. For the multidrug exporter ABCB1, we describe a protocol for reconstitution into nanodiscs and evaluation of the ATPase activity in the presence of drugs. For human OST, we describe a strategy for the purification of OST-A and OST-B complexes, including techniques to evaluate their integrity and activity using in vitro glycosylation assays. These protocols can be adapted for the production of other human ABC transporters and multimeric membrane protein complexes.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Hexosiltransferasas , Microscopía por Crioelectrón , Hexosiltransferasas/química , Hexosiltransferasas/genética , Hexosiltransferasas/metabolismo , Humanos , Proteínas de la Membrana/metabolismo
7.
Nature ; 604(7905): 371-376, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35388216

RESUMEN

The outer membrane of Gram-negative bacteria has an external leaflet that is largely composed of lipopolysaccharide, which provides a selective permeation barrier, particularly against antimicrobials1. The final and crucial step in the biosynthesis of lipopolysaccharide is the addition of a species-dependent O-antigen to the lipid A core oligosaccharide, which is catalysed by the O-antigen ligase WaaL2. Here we present structures of WaaL from Cupriavidus metallidurans, both in the apo state and in complex with its lipid carrier undecaprenyl pyrophosphate, determined by single-particle cryo-electron microscopy. The structures reveal that WaaL comprises 12 transmembrane helices and a predominantly α-helical periplasmic region, which we show contains many of the conserved residues that are required for catalysis. We observe a conserved fold within the GT-C family of glycosyltransferases and hypothesize that they have a common mechanism for shuttling the undecaprenyl-based carrier to and from the active site. The structures, combined with genetic, biochemical, bioinformatics and molecular dynamics simulation experiments, offer molecular details on how the ligands come in apposition, and allows us to propose a mechanistic model for catalysis. Together, our work provides a structural basis for lipopolysaccharide maturation in a member of the GT-C superfamily of glycosyltransferases.


Asunto(s)
Ligasas , Lipopolisacáridos , Antígenos O , Proteínas Bacterianas/química , Ligasas de Carbono-Oxígeno/química , Ligasas de Carbono-Oxígeno/genética , Microscopía por Crioelectrón , Glicosiltransferasas , Bacterias Gramnegativas , Lipopolisacáridos/química , Lipopolisacáridos/metabolismo
8.
J Med Chem ; 65(1): 191-216, 2022 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-34928144

RESUMEN

Targeted concurrent inhibition of intestinal drug efflux transporter P-glycoprotein (P-gp) and drug metabolizing enzyme cytochrome P450 3A4 (CYP3A4) is a promising approach to improve oral bioavailability of their common substrates such as docetaxel, while avoiding side effects arising from their pan inhibitions. Herein, we report the discovery and characterization of potent small molecule inhibitors of P-gp and CYP3A4 with encequidar (minimally absorbed P-gp inhibitor) as a starting point for optimization. To aid in the design of these dual inhibitors, we solved the high-resolution cryo-EM structure of encequidar bound to human P-gp. The structure guided us to prudently decorate the encequidar scaffold with CYP3A4 pharmacophores, leading to the identification of several analogues with dual potency against P-gp and CYP3A4. In vivo, dual P-gp and CYP3A4 inhibitor 3a improved the oral absorption of docetaxel by 3-fold as compared to vehicle, while 3a itself remained poorly absorbed.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Microscopía por Crioelectrón/métodos , Inhibidores del Citocromo P-450 CYP3A/farmacología , Citocromo P-450 CYP3A/química , Diseño de Fármacos , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Inhibidores del Citocromo P-450 CYP3A/química , Docetaxel/administración & dosificación , Inhibidores Enzimáticos/química , Humanos , Ratones
9.
Proc Natl Acad Sci U S A ; 118(33)2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34385322

RESUMEN

ABCB4 is expressed in hepatocytes and translocates phosphatidylcholine into bile canaliculi. The mechanism of specific lipid recruitment from the canalicular membrane, which is essential to mitigate the cytotoxicity of bile salts, is poorly understood. We present cryogenic electron microscopy structures of human ABCB4 in three distinct functional conformations. An apo-inward structure reveals how phospholipid can be recruited from the inner leaflet of the membrane without flipping its orientation. An occluded structure reveals a single phospholipid molecule in a central cavity. Its choline moiety is stabilized by cation-π interactions with an essential tryptophan residue, rationalizing the specificity of ABCB4 for phosphatidylcholine. In an inhibitor-bound structure, a posaconazole molecule blocks phospholipids from reaching the central cavity. Using a proteoliposome-based translocation assay with fluorescently labeled phosphatidylcholine analogs, we recapitulated the substrate specificity of ABCB4 in vitro and confirmed the role of the key tryptophan residue. Our results provide a structural basis for understanding an essential translocation step in the generation of bile and its sensitivity to azole drugs.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Fosfatidilcolinas/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/química , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Transporte Biológico , Técnicas de Visualización de Superficie Celular , Microscopía por Crioelectrón , Epítopos , Regulación de la Expresión Génica , Variación Genética , Células HEK293 , Humanos , Fragmentos Fab de Inmunoglobulinas , Modelos Moleculares , Conformación Proteica
10.
Proc Natl Acad Sci U S A ; 117(42): 26245-26253, 2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-33020312

RESUMEN

ABCB1 detoxifies cells by exporting diverse xenobiotic compounds, thereby limiting drug disposition and contributing to multidrug resistance in cancer cells. Multiple small-molecule inhibitors and inhibitory antibodies have been developed for therapeutic applications, but the structural basis of their activity is insufficiently understood. We determined cryo-EM structures of nanodisc-reconstituted, human ABCB1 in complex with the Fab fragment of the inhibitory, monoclonal antibody MRK16 and bound to a substrate (the antitumor drug vincristine) or to the potent inhibitors elacridar, tariquidar, or zosuquidar. We found that inhibitors bound in pairs, with one molecule lodged in the central drug-binding pocket and a second extending into a phenylalanine-rich cavity that we termed the "access tunnel." This finding explains how inhibitors can act as substrates at low concentration, but interfere with the early steps of the peristaltic extrusion mechanism at higher concentration. Our structural data will also help the development of more potent and selective ABCB1 inhibitors.


Asunto(s)
Acridinas/metabolismo , Anticuerpos Monoclonales/metabolismo , Microscopía por Crioelectrón/métodos , Tetrahidroisoquinolinas/metabolismo , Vincristina/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/antagonistas & inhibidores , Subfamilia B de Transportador de Casetes de Unión a ATP/química , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Antineoplásicos Fitogénicos/metabolismo , Proliferación Celular , Resistencia a Múltiples Medicamentos , Células HEK293 , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica
11.
Nature ; 579(7799): 443-447, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32103179

RESUMEN

In eukaryotic protein N-glycosylation, a series of glycosyltransferases catalyse the biosynthesis of a dolichylpyrophosphate-linked oligosaccharide before its transfer onto acceptor proteins1. The final seven steps occur in the lumen of the endoplasmic reticulum (ER) and require dolichylphosphate-activated mannose and glucose as donor substrates2. The responsible enzymes-ALG3, ALG9, ALG12, ALG6, ALG8 and ALG10-are glycosyltransferases of the C-superfamily (GT-Cs), which are loosely defined as containing membrane-spanning helices and processing an isoprenoid-linked carbohydrate donor substrate3,4. Here we present the cryo-electron microscopy structure of yeast ALG6 at 3.0 Å resolution, which reveals a previously undescribed transmembrane protein fold. Comparison with reported GT-C structures suggests that GT-C enzymes contain a modular architecture with a conserved module and a variable module, each with distinct functional roles. We used synthetic analogues of dolichylphosphate-linked and dolichylpyrophosphate-linked sugars and enzymatic glycan extension to generate donor and acceptor substrates using purified enzymes of the ALG pathway to recapitulate the activity of ALG6 in vitro. A second cryo-electron microscopy structure of ALG6 bound to an analogue of dolichylphosphate-glucose at 3.9 Å resolution revealed the active site of the enzyme. Functional analysis of ALG6 variants identified a catalytic aspartate residue that probably acts as a general base. This residue is conserved in the GT-C superfamily. Our results define the architecture of ER-luminal GT-C enzymes and provide a structural basis for understanding their catalytic mechanisms.


Asunto(s)
Microscopía por Crioelectrón , Retículo Endoplásmico/enzimología , Glicosiltransferasas/genética , Glicosiltransferasas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Biocatálisis , Dominio Catalítico , Secuencia Conservada , Monofosfato de Dolicol Manosa/metabolismo , Fosfatos de Dolicol/metabolismo , Glucosa/análogos & derivados , Glucosa/metabolismo , Glicosiltransferasas/deficiencia , Técnicas In Vitro , Lípidos , Proteínas de la Membrana/deficiencia , Modelos Moleculares , Mutación , Monosacáridos de Poliisoprenil Fosfato/química , Monosacáridos de Poliisoprenil Fosfato/metabolismo , Unión Proteica , Saccharomyces cerevisiae/genética , Especificidad por Sustrato
12.
Nature ; 576(7786): 315-320, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31776516

RESUMEN

The emergence and spread of drug-resistant Plasmodium falciparum impedes global efforts to control and eliminate malaria. For decades, treatment of malaria has relied on chloroquine (CQ), a safe and affordable 4-aminoquinoline that was highly effective against intra-erythrocytic asexual blood-stage parasites, until resistance arose in Southeast Asia and South America and spread worldwide1. Clinical resistance to the chemically related current first-line combination drug piperaquine (PPQ) has now emerged regionally, reducing its efficacy2. Resistance to CQ and PPQ has been associated with distinct sets of point mutations in the P. falciparum CQ-resistance transporter PfCRT, a 49-kDa member of the drug/metabolite transporter superfamily that traverses the membrane of the acidic digestive vacuole of the parasite3-9. Here we present the structure, at 3.2 Å resolution, of the PfCRT isoform of CQ-resistant, PPQ-sensitive South American 7G8 parasites, using single-particle cryo-electron microscopy and antigen-binding fragment technology. Mutations that contribute to CQ and PPQ resistance localize primarily to moderately conserved sites on distinct helices that line a central negatively charged cavity, indicating that this cavity is the principal site of interaction with the positively charged CQ and PPQ. Binding and transport studies reveal that the 7G8 isoform binds both drugs with comparable affinities, and that these drugs are mutually competitive. The 7G8 isoform transports CQ in a membrane potential- and pH-dependent manner, consistent with an active efflux mechanism that drives CQ resistance5, but does not transport PPQ. Functional studies on the newly emerging PfCRT F145I and C350R mutations, associated with decreased PPQ susceptibility in Asia and South America, respectively6,9, reveal their ability to mediate PPQ transport in 7G8 variant proteins and to confer resistance in gene-edited parasites. Structural, functional and in silico analyses suggest that distinct mechanistic features mediate the resistance to CQ and PPQ in PfCRT variants. These data provide atomic-level insights into the molecular mechanism of this key mediator of antimalarial treatment failures.


Asunto(s)
Microscopía por Crioelectrón , Resistencia a Medicamentos/efectos de los fármacos , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/ultraestructura , Plasmodium falciparum/química , Proteínas Protozoarias/química , Proteínas Protozoarias/ultraestructura , Cloroquina/metabolismo , Cloroquina/farmacología , Resistencia a Medicamentos/genética , Concentración de Iones de Hidrógeno , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/parasitología , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Modelos Moleculares , Mutación , Plasmodium falciparum/genética , Plasmodium falciparum/ultraestructura , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Quinolinas/metabolismo , Quinolinas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA