Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
Nat Commun ; 15(1): 7722, 2024 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-39242571

RESUMEN

In Candida albicans, Cdr1 pumps azole drugs out of the cells to reduce intracellular accumulation at detrimental concentrations, leading to azole-drug resistance. Milbemycin oxime, a veterinary anti-parasitic drug, strongly and specifically inhibits Cdr1. However, how Cdr1 recognizes and exports azole drugs, and how milbemycin oxime inhibits Cdr1 remain unclear. Here, we report three cryo-EM structures of Cdr1 in distinct states: the apo state (Cdr1Apo), fluconazole-bound state (Cdr1Flu), and milbemycin oxime-inhibited state (Cdr1Mil). Both the fluconazole substrate and the milbemycin oxime inhibitor are primarily recognized within the central cavity of Cdr1 through hydrophobic interactions. The fluconazole is suggested to be exported from the binding site into the environment through a lateral pathway driven by TM2, TM5, TM8 and TM11. Our findings uncover the inhibitory mechanism of milbemycin oxime, which inhibits Cdr1 through competition, hindering export, and obstructing substrate entry. These discoveries advance our understanding of Cdr1-mediated azole resistance in C. albicans and provide the foundation for the development of innovative antifungal drugs targeting Cdr1 to combat azole-drug resistance.


Asunto(s)
Antifúngicos , Azoles , Candida albicans , Microscopía por Crioelectrón , Proteínas Fúngicas , Proteínas de Transporte de Membrana , Candida albicans/efectos de los fármacos , Candida albicans/metabolismo , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/química , Proteínas Fúngicas/antagonistas & inhibidores , Antifúngicos/farmacología , Antifúngicos/química , Azoles/farmacología , Azoles/química , Proteínas de Transporte de Membrana/metabolismo , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/ultraestructura , Farmacorresistencia Fúngica , Fluconazol/farmacología , Sitios de Unión
2.
Nature ; 629(8012): 704-709, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38693257

RESUMEN

Choline is an essential nutrient that the human body needs in vast quantities for cell membrane synthesis, epigenetic modification and neurotransmission. The brain has a particularly high demand for choline, but how it enters the brain remains unknown1-3. The major facilitator superfamily transporter FLVCR1 (also known as MFSD7B or SLC49A1) was recently determined to be a choline transporter but is not highly expressed at the blood-brain barrier, whereas the related protein FLVCR2 (also known as MFSD7C or SLC49A2) is expressed in endothelial cells at the blood-brain barrier4-7. Previous studies have shown that mutations in human Flvcr2 cause cerebral vascular abnormalities, hydrocephalus and embryonic lethality, but the physiological role of FLVCR2 is unknown4,5. Here we demonstrate both in vivo and in vitro that FLVCR2 is a BBB choline transporter and is responsible for the majority of choline uptake into the brain. We also determine the structures of choline-bound FLVCR2 in both inward-facing and outward-facing states using cryo-electron microscopy. These results reveal how the brain obtains choline and provide molecular-level insights into how FLVCR2 binds choline in an aromatic cage and mediates its uptake. Our work could provide a novel framework for the targeted delivery of therapeutic agents into the brain.


Asunto(s)
Encéfalo , Colina , Proteínas de Transporte de Membrana , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Transporte Biológico , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Colina/metabolismo , Microscopía por Crioelectrón , Técnicas In Vitro , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/metabolismo , Proteínas de Transporte de Membrana/ultraestructura , Modelos Moleculares
3.
Nature ; 620(7973): 445-452, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37495693

RESUMEN

To replicate inside macrophages and cause tuberculosis, Mycobacterium tuberculosis must scavenge a variety of nutrients from the host1,2. The mammalian cell entry (MCE) proteins are important virulence factors in M. tuberculosis1,3, where they are encoded by large gene clusters and have been implicated in the transport of fatty acids4-7 and cholesterol1,4,8 across the impermeable mycobacterial cell envelope. Very little is known about how cargos are transported across this barrier, and it remains unclear how the approximately ten proteins encoded by a mycobacterial mce gene cluster assemble to transport cargo across the cell envelope. Here we report the cryo-electron microscopy (cryo-EM) structure of the endogenous Mce1 lipid-import machine of Mycobacterium smegmatis-a non-pathogenic relative of M. tuberculosis. The structure reveals how the proteins of the Mce1 system assemble to form an elongated ABC transporter complex that is long enough to span the cell envelope. The Mce1 complex is dominated by a curved, needle-like domain that appears to be unrelated to previously described protein structures, and creates a protected hydrophobic pathway for lipid transport across the periplasm. Our structural data revealed the presence of a subunit of the Mce1 complex, which we identified using a combination of cryo-EM and AlphaFold2, and name LucB. Our data lead to a structural model for Mce1-mediated lipid import across the mycobacterial cell envelope.


Asunto(s)
Proteínas Bacterianas , Microscopía por Crioelectrón , Lípidos , Proteínas de Transporte de Membrana , Mycobacterium tuberculosis , Internalización del Virus , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/ultraestructura , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/metabolismo , Proteínas de Transporte de Membrana/ultraestructura , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/ultraestructura , Tuberculosis/microbiología , Factores de Virulencia/química , Factores de Virulencia/metabolismo , Transportadoras de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/ultraestructura , Periplasma/metabolismo , Dominios Proteicos , Interacciones Hidrofóbicas e Hidrofílicas , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/ultraestructura
4.
Int J Mol Sci ; 22(22)2021 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-34829983

RESUMEN

The BAM is a macromolecular machine responsible for the folding and the insertion of integral proteins into the outer membrane of diderm Gram-negative bacteria. In Escherichia coli, it consists of a transmembrane ß-barrel subunit, BamA, and four outer membrane lipoproteins (BamB-E). Using BAM-specific antibodies, in E. coli cells, the complex is shown to localize in the lateral wall in foci. The machinery was shown to be enriched at midcell with specific cell cycle timing. The inhibition of septation by aztreonam did not alter the BAM midcell localization substantially. Furthermore, the absence of late cell division proteins at midcell did not impact BAM timing or localization. These results imply that the BAM enrichment at the site of constriction does not require an active cell division machinery. Expression of the Tre1 toxin, which impairs the FtsZ filamentation and therefore midcell localization, resulted in the complete loss of BAM midcell enrichment. A similar effect was observed for YidC, which is involved in the membrane insertion of cell division proteins in the inner membrane. The presence of the Z-ring is needed for preseptal peptidoglycan (PG) synthesis. As BAM was shown to be embedded in the PG layer, it is possible that BAM is inserted preferentially simultaneously with de novo PG synthesis to facilitate the insertion of OMPs in the newly synthesized outer membrane.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/ultraestructura , Proteínas Bacterianas/genética , Proteínas del Citoesqueleto/genética , Proteínas de Escherichia coli/genética , Proteínas de Transporte de Membrana/genética , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas Bacterianas/ultraestructura , División Celular/genética , Proteínas del Citoesqueleto/ultraestructura , Escherichia coli/química , Escherichia coli/genética , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/ultraestructura , Bacterias Gramnegativas/genética , Bacterias Gramnegativas/ultraestructura , Lipoproteínas/genética , Lipoproteínas/ultraestructura , Proteínas de Transporte de Membrana/ultraestructura , Pliegue de Proteína , Multimerización de Proteína/genética
5.
Nucleic Acids Res ; 49(16): 9280-9293, 2021 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-34387667

RESUMEN

Activator proteins 1 (AP-1) comprise one of the largest families of eukaryotic basic leucine zipper transcription factors. Despite advances in the characterization of AP-1 DNA-binding sites, our ability to predict new binding sites and explain how the proteins achieve different gene expression levels remains limited. Here we address the role of sequence-specific DNA flexibility for stability and specific binding of AP-1 factors, using microsecond-long molecular dynamics simulations. As a model system, we employ yeast AP-1 factor Yap1 binding to three different response elements from two genetic environments. Our data show that Yap1 actively exploits the sequence-specific flexibility of DNA within the response element to form stable protein-DNA complexes. The stability also depends on the four to six flanking nucleotides, adjacent to the response elements. The flanking sequences modulate the conformational adaptability of the response element, making it more shape-efficient to form specific contacts with the protein. Bioinformatics analysis of differential expression of the studied genes supports our conclusions: the stability of Yap1-DNA complexes, modulated by the flanking environment, influences the gene expression levels. Our results provide new insights into mechanisms of protein-DNA recognition and the biological regulation of gene expression levels in eukaryotes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , ADN/genética , Proteínas de Saccharomyces cerevisiae/genética , Factor de Transcripción AP-1/genética , Factores de Transcripción/genética , Secuencia de Bases/genética , Sitios de Unión/genética , ADN/ultraestructura , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/ultraestructura , Regulación de la Expresión Génica/genética , Sustancias Macromoleculares/ultraestructura , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/ultraestructura , Simulación de Dinámica Molecular , Elementos de Respuesta/genética , Proteínas de Saccharomyces cerevisiae/ultraestructura , Factor de Transcripción AP-1/ultraestructura , Factores de Transcripción/ultraestructura , Proteínas Señalizadoras YAP
6.
PLoS Biol ; 19(8): e3001370, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34383749

RESUMEN

The mycobacterial membrane protein large 3 (MmpL3) transporter is essential and required for shuttling the lipid trehalose monomycolate (TMM), a precursor of mycolic acid (MA)-containing trehalose dimycolate (TDM) and mycolyl arabinogalactan peptidoglycan (mAGP), in Mycobacterium species, including Mycobacterium tuberculosis and Mycobacterium smegmatis. However, the mechanism that MmpL3 uses to facilitate the transport of fatty acids and lipidic elements to the mycobacterial cell wall remains elusive. Here, we report 7 structures of the M. smegmatis MmpL3 transporter in its unbound state and in complex with trehalose 6-decanoate (T6D) or TMM using single-particle cryo-electron microscopy (cryo-EM) and X-ray crystallography. Combined with calculated results from molecular dynamics (MD) and target MD simulations, we reveal a lipid transport mechanism that involves a coupled movement of the periplasmic domain and transmembrane helices of the MmpL3 transporter that facilitates the shuttling of lipids to the mycobacterial cell wall.


Asunto(s)
Proteínas Bacterianas/metabolismo , Factores Cordón/metabolismo , Metabolismo de los Lípidos , Proteínas de Transporte de Membrana/metabolismo , Mycobacterium smegmatis/metabolismo , Proteínas Bacterianas/ultraestructura , Microscopía por Crioelectrón , Decanoatos/metabolismo , Escherichia coli , Proteínas de Transporte de Membrana/ultraestructura , Simulación de Dinámica Molecular , Mycobacterium smegmatis/ultraestructura , Trehalosa/metabolismo
7.
Nat Commun ; 12(1): 4625, 2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34330923

RESUMEN

Bacteria often secrete diffusible protein toxins (bacteriocins) to kill bystander cells during interbacterial competition. Here, we use biochemical, biophysical and structural analyses to show how a bacteriocin exploits TolC, a major outer-membrane antibiotic efflux channel in Gram-negative bacteria, to transport itself across the outer membrane of target cells. Klebicin C (KlebC), a rRNase toxin produced by Klebsiella pneumoniae, binds TolC of a related species (K. quasipneumoniae) with high affinity through an N-terminal, elongated helical hairpin domain common amongst bacteriocins. The KlebC helical hairpin opens like a switchblade to bind TolC. A cryo-EM structure of this partially translocated state, at 3.1 Å resolution, reveals that KlebC associates along the length of the TolC channel. Thereafter, the unstructured N-terminus of KlebC protrudes beyond the TolC iris, presenting a TonB-box sequence to the periplasm. Association with proton-motive force-linked TonB in the inner membrane drives toxin import through the channel. Finally, we demonstrate that KlebC binding to TolC blocks drug efflux from bacteria. Our results indicate that TolC, in addition to its known role in antibiotic export, can function as a protein import channel for bacteriocins.


Asunto(s)
Antibacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/metabolismo , Bacteriocinas/metabolismo , Canales Iónicos/metabolismo , Klebsiella/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/ultraestructura , Proteínas Bacterianas/química , Proteínas Bacterianas/ultraestructura , Transporte Biológico , Microscopía por Crioelectrón/métodos , Canales Iónicos/química , Canales Iónicos/ultraestructura , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/ultraestructura , Modelos Moleculares , Unión Proteica , Conformación Proteica
8.
Nat Commun ; 12(1): 4236, 2021 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-34244493

RESUMEN

The repertoire of peptides presented by major histocompatibility complex class I (MHC-I) molecules on the cell surface is tailored by the ER-resident peptide loading complex (PLC), which contains the exchange catalyst tapasin. Tapasin stabilizes MHC-I molecules and promotes the formation of stable peptide-MHC-I (pMHC-I) complexes that serve as T cell antigens. Exchange of suboptimal by high-affinity ligands is catalyzed by tapasin, but the underlying mechanism is still elusive. Here we analyze the tapasin-induced changes in MHC-I dynamics, and find the catalyst to exploit two essential features of MHC-I. First, tapasin recognizes a conserved allosteric site underneath the α2-1-helix of MHC-I, 'loosening' the MHC-I F-pocket region that accomodates the C-terminus of the peptide. Second, the scoop loop11-20 of tapasin relies on residue L18 to target the MHC-I F-pocket, enabling peptide exchange. Meanwhile, tapasin residue K16 plays an accessory role in catalysis of MHC-I allotypes bearing an acidic F-pocket. Thus, our results provide an explanation for the observed allele-specificity of catalyzed peptide exchange.


Asunto(s)
Alelos , Presentación de Antígeno/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Regulación Alostérica , Biocatálisis , Cristalografía por Rayos X , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/aislamiento & purificación , Antígenos de Histocompatibilidad Clase I/ultraestructura , Humanos , Inmunoglobulinas/metabolismo , Inmunoglobulinas/ultraestructura , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/ultraestructura , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/aislamiento & purificación , Proteínas de Transporte de Membrana/ultraestructura , Simulación de Dinámica Molecular , Mutación , Resonancia Magnética Nuclear Biomolecular , Conformación Proteica en Hélice alfa , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo
9.
J Biol Chem ; 297(1): 100863, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34118233

RESUMEN

The serotonin transporter (SERT) shapes serotonergic neurotransmission by retrieving its eponymous substrate from the synaptic cleft. Ligands that discriminate between SERT and its close relative, the dopamine transporter DAT, differ in their association rate constant rather than their dissociation rate. The structural basis for this phenomenon is not known. Here we examined the hypothesis that the extracellular loops 2 (EL2) and 4 (EL4) limit access to the ligand-binding site of SERT. We employed an antibody directed against EL4 (residues 388-400) and the antibody fragments 8B6 scFv (directed against EL2 and EL4) and 15B8 Fab (directed against EL2) and analyzed their effects on the transport cycle of and inhibitor binding to SERT. Electrophysiological recordings showed that the EL4 antibody and 8B6 scFv impeded the initial substrate-induced transition from the outward to the inward-facing conformation but not the forward cycling mode of SERT. In contrast, binding of radiolabeled inhibitors to SERT was enhanced by either EL4- or EL2-directed antibodies. We confirmed this observation by determining the association and dissociation rate of the DAT-selective inhibitor methylphenidate via electrophysiological recordings; occupancy of EL2 with 15B8 Fab enhanced the affinity of SERT for methylphenidate by accelerating its binding. Based on these observations, we conclude that (i) EL4 undergoes a major movement during the transition from the outward to the inward-facing state, and (ii) EL2 and EL4 limit access of inhibitors to the binding of SERT, thus acting as a selectivity filter. This insight has repercussions for drug development.


Asunto(s)
Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Membrana/genética , Conformación Proteica/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Secuencia de Aminoácidos/genética , Animales , Sitios de Unión/efectos de los fármacos , Células COS , Chlorocebus aethiops , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/antagonistas & inhibidores , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/ultraestructura , Células HEK293 , Humanos , Ligandos , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/ultraestructura , Técnicas de Placa-Clamp , Dominios Proteicos/genética , Serotonina/química , Serotonina/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/efectos de los fármacos , Proteínas de Transporte de Serotonina en la Membrana Plasmática/ultraestructura , Inhibidores Selectivos de la Recaptación de Serotonina/química
10.
mBio ; 12(3): e0103121, 2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34044590

RESUMEN

Antibiotic-resistant strains of the Gram-negative pathogen Acinetobacter baumannii have emerged as a significant global health threat. One successful therapeutic option to treat bacterial infections has been to target the bacterial ribosome. However, in many cases, multidrug efflux pumps within the bacterium recognize and extrude these clinically important antibiotics designed to inhibit the protein synthesis function of the bacterial ribosome. Thus, multidrug efflux within A. baumannii and other highly drug-resistant strains is a major cause of failure of drug-based treatments of infectious diseases. We here report the first structures of the Acinetobacter drug efflux (Ade)J pump in the presence of the antibiotic eravacycline, using single-particle cryo-electron microscopy (cryo-EM). We also describe cryo-EM structures of the eravacycline-bound forms of the A. baumannii ribosome, including the 70S, 50S, and 30S forms. Our data indicate that the AdeJ pump primarily uses hydrophobic interactions to bind eravacycline, while the 70S ribosome utilizes electrostatic interactions to bind this drug. Our work here highlights how an antibiotic can bind multiple bacterial targets through different mechanisms and potentially enables drug optimization by taking advantage of these different modes of ligand binding. IMPORTANCE Acinetobacter baumannii has developed into a highly antibiotic-resistant Gram-negative pathogen. The prevalent AdeJ multidrug efflux pump mediates resistance to different classes of antibiotics known to inhibit the function of the 70S ribosome. Here, we report the first structures of the A. baumannii AdeJ pump, both in the absence and presence of eravacycline. We also describe structures of the A. baumannii ribosome bound by this antibiotic. Our results indicate that AdeJ and the ribosome use very distinct binding modes for drug recognition. Our work will ultimately enable structure-based drug discovery to combat antibiotic-resistant A. baumannii infection.


Asunto(s)
Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/ultraestructura , Antibacterianos/farmacología , Microscopía por Crioelectrón/métodos , Proteínas de Transporte de Membrana/metabolismo , Ribosomas/metabolismo , Tetraciclinas/metabolismo , Tetraciclinas/farmacología , Acinetobacter baumannii/genética , Acinetobacter baumannii/metabolismo , Farmacorresistencia Bacteriana Múltiple , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/ultraestructura , Ribosomas/química , Ribosomas/ultraestructura
11.
mBio ; 12(2)2021 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-33820823

RESUMEN

Gram-negative bacteria utilize the resistance-nodulation-cell division (RND) superfamily of efflux pumps to expel a variety of toxic compounds from the cell. The Escherichia coli CusA membrane protein, which recognizes and extrudes biocidal Cu(I) and Ag(I) ions, belongs to the heavy-metal efflux (HME) subfamily of RND efflux pumps. We here report four structures of the trimeric CusA heavy-metal efflux pump in the presence of Cu(I) using single-particle cryo-electron microscopy (cryo-EM). We discover that different CusA protomers within the trimer are able to bind Cu(I) ions simultaneously. Our structural data combined with molecular dynamics (MD) simulations allow us to propose a mechanism for ion transport where each CusA protomer functions independently within the trimer.IMPORTANCE The bacterial RND superfamily of efflux pumps mediate resistance to a variety of biocides, including Cu(I) and Ag(I) ions. Here we report four cryo-EM structures of the trimeric CusA pump in the presence of Cu(I). Combined with MD simulations, our data indicate that each CusA protomer within the trimer recognizes and extrudes Cu(I) independently.


Asunto(s)
Microscopía por Crioelectrón , Proteínas de Escherichia coli/química , Escherichia coli/metabolismo , Transporte Iónico , Proteínas de Transporte de Membrana/química , Metales Pesados/metabolismo , Sitios de Unión , Transporte Biológico , Cobre/metabolismo , Escherichia coli/genética , Escherichia coli/ultraestructura , Proteínas de Escherichia coli/ultraestructura , Proteínas de Transporte de Membrana/ultraestructura , Simulación de Dinámica Molecular , Unión Proteica , Plata/metabolismo
12.
mBio ; 12(1)2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33622726

RESUMEN

Acinetobacter baumannii is a Gram-negative pathogen that has emerged as one of the most highly antibiotic-resistant bacteria worldwide. Multidrug efflux within these highly drug-resistant strains and other opportunistic pathogens is a major cause of failure of drug-based treatments of infectious diseases. The best-characterized multidrug efflux system in A. baumannii is the prevalent Acinetobacterdrug efflux B (AdeB) pump, which is a member of the resistance-nodulation-cell division (RND) superfamily. Here, we report six structures of the trimeric AdeB multidrug efflux pump in the presence of ethidium bromide using single-particle cryoelectron microscopy (cryo-EM). These structures allow us to directly observe various novel conformational states of the AdeB trimer, including the transmembrane region of trimeric AdeB can be associated with form a trimer assembly or dissociated into "dimer plus monomer" and "monomer plus monomer plus monomer" configurations. We also discover that a single AdeB protomer can simultaneously anchor a number of ethidium ligands and that different AdeB protomers can bind ethidium molecules simultaneously. Combined with molecular dynamics (MD) simulations, we reveal a drug transport mechanism that involves multiple multidrug-binding sites and various transient states of the AdeB membrane protein. Our data suggest that each AdeB protomer within the trimer binds and exports drugs independently.IMPORTANCEAcinetobacter baumannii has emerged as one of the most highly antibiotic-resistant Gram-negative pathogens. The prevalent AdeB multidrug efflux pump mediates resistance to a broad spectrum of clinically relevant antimicrobial agents. Here, we report six cryo-EM structures of the trimeric AdeB pump in the presence of ethidium bromide. We discover that a single AdeB protomer can simultaneously anchor a number of ligands, and different AdeB protomers can bind ethidium molecules simultaneously. The results indicate that each AdeB protomer within the trimer recognizes and extrudes drugs independently.


Asunto(s)
Acinetobacter baumannii/genética , Proteínas Bacterianas/química , Microscopía por Crioelectrón , Farmacorresistencia Bacteriana Múltiple/genética , Proteínas de Transporte de Membrana/química , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/ultraestructura , Antibacterianos/farmacología , Proteínas Bacterianas/ultraestructura , División Celular/efectos de los fármacos , Etidio/farmacología , Proteínas de Transporte de Membrana/ultraestructura
13.
Nat Struct Mol Biol ; 28(2): 173-180, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33432245

RESUMEN

Nucleic acid-sensing Toll-like receptors (TLRs) play a pivotal role in innate immunity by recognizing foreign DNA and RNA. Compartmentalization of these TLRs in the endosome limits their activation by self-derived nucleic acids and reduces the possibility of autoimmune reactions. Although chaperone Unc-93 homolog B1, TLR signaling regulator (UNC93B1) is indispensable for the trafficking of TLRs from the endoplasmic reticulum to the endosome, mechanisms of UNC93B1-mediated TLR regulation remain largely unknown. Here, we report two cryo-EM structures of human and mouse TLR3-UNC93B1 complexes and a human TLR7-UNC93B1 complex. UNC93B1 exhibits structural similarity to the major facilitator superfamily transporters. Both TLRs interact with the UNC93B1 amino-terminal six-helix bundle through their transmembrane and luminal juxtamembrane regions, but the complexes of TLR3 and TLR7 with UNC93B1 differ in their oligomerization state. The structural information provided here should aid in designing compounds to combat autoimmune diseases.


Asunto(s)
Proteínas de Transporte de Membrana , Receptor Toll-Like 3 , Receptor Toll-Like 7 , Animales , Humanos , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/ultraestructura , Ratones , Unión Proteica , Multimerización de Proteína , Receptor Toll-Like 3/química , Receptor Toll-Like 3/ultraestructura , Receptor Toll-Like 7/química , Receptor Toll-Like 7/ultraestructura
14.
Methods Appl Fluoresc ; 9(1): 015006, 2021 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-33427202

RESUMEN

Synthesis and multiple STED imaging applications of four, red-emitting (610-670 nm), tetrazine-functionalized fluorescent probes (CBRD = Chemical Biology Research group Dye 1-4) with large Stokes-shift is presented. Present studies revealed the super-resolution microscopy applicability of the probes as demonstrated through bioorthogonal labeling scheme of cytoskeletal proteins actin and keratin-19, and mitochondrial protein TOMM20. Furthermore, super-resolved images of insulin receptors in live-cell bioorthogonal labeling schemes through a genetically encoded cyclooctynylated non-canonical amino acid are also presented. The large Stokes-shifts and the wide spectral bands of the probes enabled the use of two common depletion lasers (660 nm and 775 nm). The probes were also found suitable for super-resolution microscopy in combination with two-photon excitation (2P-STED) resulting in improved spatial resolution. One of the dyes was also used together with two commercial dyes in the three-color STED imaging of intracellular structures.


Asunto(s)
Colorantes Fluorescentes , Microscopía Fluorescente/métodos , Actinas/análisis , Actinas/ultraestructura , Línea Celular , Células HEK293 , Células HeLa , Humanos , Queratina-19/análisis , Queratina-19/ultraestructura , Proteínas de Transporte de Membrana/análisis , Proteínas de Transporte de Membrana/ultraestructura , Microscopía Confocal , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Receptor de Insulina/análisis , Receptor de Insulina/ultraestructura , Receptores de Superficie Celular/análisis , Receptores de Superficie Celular/ultraestructura
15.
Proc Natl Acad Sci U S A ; 117(34): 20597-20606, 2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32788370

RESUMEN

The major histocompatibility complex class-I (MHC-I) peptide-loading complex (PLC) is a cornerstone of the human adaptive immune system, being responsible for processing antigens that allow killer T cells to distinguish between healthy and compromised cells. Based on a recent low-resolution cryo-electron microscopy (cryo-EM) structure of this large membrane-bound protein complex, we report an atomistic model of the PLC and study its conformational dynamics on the multimicrosecond time scale using all-atom molecular dynamics (MD) simulations in an explicit lipid bilayer and water environment (1.6 million atoms in total). The PLC has a layered structure, with two editing modules forming a flexible protein belt surrounding a stable, catalytically active core. Tapasin plays a central role in the PLC, stabilizing the MHC-I binding groove in a conformation reminiscent of antigen-loaded MHC-I. The MHC-I-linked glycan steers a tapasin loop involved in peptide editing toward the binding groove. Tapasin conformational dynamics are also affected by calreticulin through a conformational selection mechanism that facilitates MHC-I recruitment into the complex.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Calreticulina/metabolismo , Microscopía por Crioelectrón , Antígenos de Histocompatibilidad Clase I/ultraestructura , Humanos , Proteínas de Transporte de Membrana/metabolismo , Proteínas de Transporte de Membrana/ultraestructura , Simulación de Dinámica Molecular , Polisacáridos/metabolismo , Proteína Disulfuro Isomerasas/metabolismo
16.
Life Sci Alliance ; 3(8)2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32646883

RESUMEN

Hedgehog (HH) signaling is essential for metazoan development. The HH ligand is secreted into the extracellular space by a cell surface protein named Dispatched-1 (DISP1). Here, we report the cryo-EM structure of human DISP1 protein. DISP1 contains 12 transmembrane helices (TMs) and two extracellular domains (ECDs). Its ECDs reveal an open state, in contrast to its structural homologues PTCH1 and NPC1, whose extracellular/luminal domains adopt a closed state. The low-resolution structure of the DISP1 complex with dual lipid-modified HH ligand reveals how the ECDs of DISP1 engage with HH ligand. Moreover, several cholesterol-like molecules are found in the TMs, implying a transport-like function of DISP1.


Asunto(s)
Proteínas de la Membrana/ultraestructura , Proteínas de Transporte de Membrana/ultraestructura , Proteínas Hedgehog/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ligandos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/genética , Proteína Niemann-Pick C1 , Receptor Patched-1/metabolismo , Receptor Patched-1/ultraestructura , Dominios Proteicos/genética , Transducción de Señal , Transactivadores/genética
17.
mBio ; 11(3)2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32457251

RESUMEN

Neisseria gonorrhoeae is an obligate human pathogen and causative agent of the sexually transmitted infection (STI) gonorrhea. The most predominant and clinically important multidrug efflux system in N. gonorrhoeae is the multiple transferrable resistance (Mtr) pump, which mediates resistance to a number of different classes of structurally diverse antimicrobial agents, including clinically used antibiotics (e.g., ß-lactams and macrolides), dyes, detergents and host-derived antimicrobials (e.g., cationic antimicrobial peptides and bile salts). Recently, it has been found that gonococci bearing mosaic-like sequences within the mtrD gene can result in amino acid changes that increase the MtrD multidrug efflux pump activity, probably by influencing antimicrobial recognition and/or extrusion to elevate the level of antibiotic resistance. Here, we report drug-bound solution structures of the MtrD multidrug efflux pump carrying a mosaic-like sequence using single-particle cryo-electron microscopy, with the antibiotics bound deeply inside the periplasmic domain of the pump. Through this structural approach coupled with genetic studies, we identify critical amino acids that are important for drug resistance and propose a mechanism for proton translocation.IMPORTANCENeisseria gonorrhoeae has become a highly antimicrobial-resistant Gram-negative pathogen. Multidrug efflux is a major mechanism that N. gonorrhoeae uses to counteract the action of multiple classes of antibiotics. It appears that gonococci bearing mosaic-like sequences within the gene mtrD, encoding the most predominant and clinically important transporter of any gonococcal multidrug efflux pump, significantly elevate drug resistance and enhance transport function. Here, we report cryo-electron microscopy (EM) structures of N. gonorrhoeae MtrD carrying a mosaic-like sequence that allow us to understand the mechanism of drug recognition. Our work will ultimately inform structure-guided drug design for inhibiting these critical multidrug efflux pumps.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/ultraestructura , Farmacorresistencia Bacteriana Múltiple , Proteínas de Transporte de Membrana/ultraestructura , Neisseria gonorrhoeae/efectos de los fármacos , Proteínas Bacterianas/química , Microscopía por Crioelectrón , Regulación Bacteriana de la Expresión Génica , Proteínas de Transporte de Membrana/química , Neisseria gonorrhoeae/genética
18.
Biomolecules ; 10(4)2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32316569

RESUMEN

Magnesium transporter A (MgtA) is an active transporter responsible for importing magnesium ions into the cytoplasm of prokaryotic cells. This study focuses on the peptide corresponding to the intrinsically disordered N-terminal region of MgtA, referred to as KEIF. Primary-structure and bioinformatic analyses were performed, followed by studies of the undisturbed single chain using a combination of techniques including small-angle X-ray scattering, circular dichroism spectroscopy, and atomistic molecular-dynamics simulations. Moreover, interactions with large unilamellar vesicles were investigated by using dynamic light scattering, laser Doppler velocimetry, cryogenic transmission electron microscopy, and circular dichroism spectroscopy. KEIF was confirmed to be intrinsically disordered in aqueous solution, although extended and containing little ß -structure and possibly PPII structure. An increase of helical content was observed in organic solvent, and a similar effect was also seen in aqueous solution containing anionic vesicles. Interactions of cationic KEIF with anionic vesicles led to the hypothesis that KEIF adsorbs to the vesicle surface through electrostatic and entropic driving forces. Considering this, there is a possibility that the biological role of KEIF is to anchor MgtA in the cell membrane, although further investigation is needed to confirm this hypothesis.


Asunto(s)
Adenosina Trifosfatasas/química , Fenómenos Químicos , Proteínas de Escherichia coli/química , Escherichia coli/metabolismo , Proteínas Intrínsecamente Desordenadas/química , Proteínas de Transporte de Membrana/química , Adenosina Trifosfatasas/ultraestructura , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Dicroismo Circular , Proteínas de Escherichia coli/ultraestructura , Proteínas Intrínsecamente Desordenadas/ultraestructura , Lípidos/química , Proteínas de Transporte de Membrana/ultraestructura , Simulación de Dinámica Molecular , Tamaño de la Partícula , Probabilidad , Estructura Secundaria de Proteína , Dispersión del Ángulo Pequeño , Liposomas Unilamelares/química , Difracción de Rayos X
19.
J Mol Biol ; 432(2): 484-496, 2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31669168

RESUMEN

Proper membrane insertion is crucial for the structure and function of membrane proteins in all cells. The YidC insertase plays an essential role in this process, but the molecular mechanism of YidC-mediated insertion remains unknown. Here we track the stepwise movement of Pf3 coat through YidC by obtaining a series of translational arrested intermediates, and investigate them by thiol cross-linking. We show that Pf3 is inserted as a helical hairpin, i.e., the prospective transmembrane segment moves along the YidC greasy slide comprised of TM3 and TM5, whereas the N-terminal tail transiently folds back into the hydrophilic groove of YidC located in the inner leaflet of the membrane until it is translocated to the periplasm in a subsequent step involving the electrochemical membrane potential. In addition to providing virtual insights about how YidC inserts single-spanning membrane proteins, our study also demonstrates a valuable in vivo tracking method that can be applied to study more complicated substrates or other translocases.


Asunto(s)
Proteínas de Escherichia coli/ultraestructura , Escherichia coli/ultraestructura , Proteínas de la Membrana/ultraestructura , Proteínas de Transporte de Membrana/ultraestructura , Biosíntesis de Proteínas , Membrana Celular/genética , Membrana Celular/ultraestructura , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Interacciones Hidrofóbicas e Hidrofílicas , Proteínas de la Membrana/genética , Proteínas de Transporte de Membrana/genética , Mutación/genética , Periplasma/genética , Transporte de Proteínas/genética , Especificidad por Sustrato
20.
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