Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Intervalo de año de publicación
1.
PLoS Pathog ; 17(9): e1009488, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34492091

RESUMEN

Arenavirus entry into host cells occurs through a low pH-dependent fusion with late endosomes that is mediated by the viral glycoprotein complex (GPC). The mechanisms of GPC-mediated membrane fusion and of virus targeting to late endosomes are not well understood. To gain insights into arenavirus fusion, we examined cell-cell fusion induced by the Old World Lassa virus (LASV) GPC complex. LASV GPC-mediated cell fusion is more efficient and occurs at higher pH with target cells expressing human LAMP1 compared to cells lacking this cognate receptor. However, human LAMP1 is not absolutely required for cell-cell fusion or LASV entry. We found that GPC-induced fusion progresses through the same lipid intermediates as fusion mediated by other viral glycoproteins-a lipid curvature-sensitive intermediate upstream of hemifusion and a hemifusion intermediate downstream of acid-dependent steps that can be arrested in the cold. Importantly, GPC-mediated fusion and LASV pseudovirus entry are specifically augmented by an anionic lipid, bis(monoacylglycero)phosphate (BMP), which is highly enriched in late endosomes. This lipid also specifically promotes cell fusion mediated by Junin virus GPC, an unrelated New World arenavirus. We show that BMP promotes late steps of LASV fusion downstream of hemifusion-the formation and enlargement of fusion pores. The BMP-dependence of post-hemifusion stages of arenavirus fusion suggests that these viruses evolved to use this lipid as a cofactor to selectively fuse with late endosomes.


Asunto(s)
Endosomas/metabolismo , Fiebre de Lassa/metabolismo , Virus Lassa/fisiología , Lisofosfolípidos/metabolismo , Monoglicéridos/metabolismo , Internalización del Virus , Animales , Células COS , Chlorocebus aethiops , Células HEK293 , Humanos , Proteínas del Envoltorio Viral/metabolismo
2.
J Virol ; 93(23)2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31511384

RESUMEN

Lassa virus (LASV) is the causative agent of a fatal hemorrhagic fever in humans. The glycoprotein (GP) of LASV mediates viral entry into host cells, and correct processing and modification of GP by host factors is a prerequisite for virus replication. Here, using an affinity purification-coupled mass spectrometry (AP-MS) strategy, 591 host proteins were identified as interactors of LASV GP. Gene ontology analysis was performed to functionally annotate these proteins, and the oligosaccharyltransferase (OST) complex was highly enriched. Functional studies conducted by using CRISPR-Cas9-mediated knockouts showed that STT3A and STT3B, the two catalytically active isoforms of the OST complex, are essential for the propagation of the recombinant arenavirus rLCMV/LASV glycoprotein precursor, mainly via affecting virus infectivity. Knockout of STT3B, but not STT3A, caused hypoglycosylation of LASV GP, indicating a preferential requirement of LASV for the STT3B-OST isoform. Furthermore, double knockout of magnesium transporter 1 (MAGT1) and tumor suppressor candidate 3 (TUSC3), two specific subunits of STT3B-OST, also caused hypoglycosylation of LASV GP and affected virus propagation. Site-directed mutagenesis analysis revealed that the oxidoreductase CXXC active-site motif of MAGT1 or TUSC3 is essential for the glycosylation of LASV GP. NGI-1, a small-molecule OST inhibitor, can effectively reduce virus infectivity without affecting cell viability. The STT3B-dependent N-glycosylation of GP is conserved among other arenaviruses, including both the Old World and New World groups. Our study provided a systematic view of LASV GP-host interactions and revealed the preferential requirement of STT3B for LASV GP N-glycosylation.IMPORTANCE Glycoproteins play vital roles in the arenavirus life cycle by facilitating virus entry and participating in the virus budding process. N-glycosylation of GPs is responsible for their proper functioning; however, little is known about the host factors on which the virus depends for this process. In this study, a comprehensive LASV GP interactome was characterized, and further study revealed that STT3B-dependent N-glycosylation was preferentially required by arenavirus GPs and critical for virus infectivity. The two specific thioredoxin subunits of STT3B-OST MAGT1 and TUSC3 were found to be essential for the N-glycosylation of viral GP. NGI-1, a small-molecule inhibitor of OST, also showed a robust inhibitory effect on arenavirus. Our study provides new insights into LASV GP-host interactions and extends the potential targets for the development of novel therapeutics against Lassa fever in the future.


Asunto(s)
Glicoproteínas/metabolismo , Hexosiltransferasas/metabolismo , Fiebre de Lassa/metabolismo , Virus Lassa/metabolismo , Proteínas de la Membrana/metabolismo , Sistemas CRISPR-Cas , Proteínas de Transporte de Catión , Línea Celular , Técnicas de Inactivación de Genes , Glicosilación , Células HEK293 , Células HeLa , Hexosiltransferasas/genética , Humanos , Virus Lassa/genética , Virus Lassa/patogenicidad , Proteínas de la Membrana/genética , Mutagénesis Sitio-Dirigida , Proteínas del Tejido Nervioso , Oxidorreductasas/metabolismo , Isoformas de Proteínas , Receptores de Superficie Celular , Proteínas Supresoras de Tumor/genética , Internalización del Virus
3.
EBioMedicine ; 40: 605-613, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30711514

RESUMEN

BACKGROUND: Lassa virus (LASV) is the etiologic agent of an acute hemorrhagic fever endemic in West Africa. Natural killer (NK) cells control viral infections in part through the interaction between killer cell immunoglobulin-like receptors (KIRs) and their ligands. LASV infection is associated with defective immune responses, including inhibition of NK cell activity in the presence of MHC-class 1+-infected target cells. METHODS: We compared individual KIR and HLA-class 1 genotypes of 68 healthy volunteers to 51 patients infected with LASV in Sierra Leone, including 37 survivors and 14 fatalities. Next, potential HLA-C1, HLA-C2, and HLA-Bw4 binding epitopes were in silico screened among LASV nucleoprotein (NP) and envelope glycoprotein (GP). Selected 10-mer peptides were then tested in peptide-HLA stabilization, KIR binding and polyfunction assays. FINDINGS: LASV-infected patients were similar to healthy controls, except for the inhibitory KIR2DL2 gene. We found a specific increase in the HLA-C1:KIR2DL2 interaction in fatalities (10/11) as compared to survivors (12/19) and controls (19/29). We also identified that strong of NP and GP viral epitopes was only observed with HLA-C molecules, and associated with strong inhibition of degranulation in the presence of KIR2DL+ NK cells. This inhibitory effect significantly increased in the presence of the vGP420 variant, detected in 28.1% of LASV sequences. INTERPRETATION: Our finding suggests that presentation of specific LASV epitopes by HLA-C alleles to the inhibitory KIR2DL2 receptor on NK cells could potentially prevent the killing of infected cells and provides insights into the mechanisms by which LASV can escape NK-cell-mediated immune pressure.


Asunto(s)
Epítopos/inmunología , Antígenos HLA-C/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Fiebre de Lassa/inmunología , Fiebre de Lassa/metabolismo , Virus Lassa/inmunología , Receptores KIR2DL2/metabolismo , Antígenos Virales/inmunología , Línea Celular , Citotoxicidad Inmunológica , Mapeo Epitopo/métodos , Genotipo , Antígenos HLA-C/genética , Humanos , Tolerancia Inmunológica , Inmunomodulación , Inmunofenotipificación , Fiebre de Lassa/genética , Fiebre de Lassa/virología , Unión Proteica , Receptores KIR2DL2/genética
4.
J Virol ; 90(22): 10329-10338, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27605678

RESUMEN

To effectively infect cells, Lassa virus needs to switch in an endosomal compartment from its primary receptor, α-dystroglycan, to a protein termed LAMP1. A unique histidine triad on the surface of the receptor-binding domain from the glycoprotein spike complex of Lassa virus is important for LAMP1 binding. Here we investigate mutated spikes that have an impaired ability to interact with LAMP1 and show that although LAMP1 is important for efficient infectivity, it is not required for spike-mediated membrane fusion per se Our studies reveal important regulatory roles for histidines from the triad in sensing acidic pH and preventing premature spike triggering. We further show that LAMP1 requires a positively charged His230 residue to engage with the spike complex and that LAMP1 binding promotes membrane fusion. These results elucidate the molecular role of LAMP1 binding during Lassa virus cell entry and provide new insights into how pH is sensed by the spike. IMPORTANCE: Lassa virus is a devastating disease-causing agent in West Africa, with a significant yearly death toll and severe long-term complications associated with its infection in survivors. In recent years, we learned that Lassa virus needs to switch receptors in a pH-dependent manner to efficiently infect cells, but neither the molecular mechanisms that allow switching nor the actual effects of switching were known. Here we investigate the activity of the viral spike complex after abrogation of its ability to switch receptors. These studies inform us about the role of switching receptors and provide new insights into how the spike senses acidic pH.


Asunto(s)
Fiebre de Lassa/metabolismo , Fiebre de Lassa/virología , Virus Lassa/metabolismo , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Unión Proteica/fisiología , África Occidental , Animales , Línea Celular , Chlorocebus aethiops , Distroglicanos/metabolismo , Endosomas/metabolismo , Endosomas/virología , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Fusión de Membrana/fisiología , Receptores Virales/metabolismo , Células Vero , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus
5.
J Virol ; 90(14): 6412-6429, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27147735

RESUMEN

UNLABELLED: The pathogenic Old World arenavirus Lassa virus (LASV) causes a severe hemorrhagic fever with a high rate of mortality in humans. Several LASV receptors, including dystroglycan (DG), TAM receptor tyrosine kinases, and C-type lectins, have been identified, suggesting complex receptor use. Upon receptor binding, LASV enters the host cell via an unknown clathrin- and dynamin-independent pathway that delivers the virus to late endosomes, where fusion occurs. Here we investigated the mechanisms underlying LASV endocytosis in human cells in the context of productive arenavirus infection, using recombinant lymphocytic choriomeningitis virus (rLCMV) expressing the LASV glycoprotein (rLCMV-LASVGP). We found that rLCMV-LASVGP entered human epithelial cells via DG using a macropinocytosis-related pathway independently of alternative receptors. Dystroglycan-mediated entry of rLCMV-LASVGP required sodium hydrogen exchangers, actin, and the GTPase Cdc42 and its downstream targets, p21-activating kinase-1 (PAK1) and Wiskott-Aldrich syndrome protein (N-Wasp). Unlike other viruses that enter cells via macropinocytosis, rLCMV-LASVGP entry did not induce overt changes in cellular morphology and hardly affected actin dynamics or fluid uptake. Screening of kinase inhibitors identified protein kinase C, phosphoinositide 3-kinase, and the receptor tyrosine kinase human hepatocyte growth factor receptor (HGFR) to be regulators of rLCMV-LASVGP entry. The HGFR inhibitor EMD 1214063, a candidate anticancer drug, showed antiviral activity against rLCMV-LASVGP at the level of entry. When combined with ribavirin, which is currently used to treat human arenavirus infection, EMD 1214063 showed additive antiviral effects. In sum, our study reveals that DG can link LASV to an unusual pathway of macropinocytosis that causes only minimal perturbation of the host cell and identifies cellular kinases to be possible novel targets for therapeutic intervention. IMPORTANCE: Lassa virus (LASV) causes several hundred thousand infections per year in Western Africa, with the mortality rate among hospitalized patients being high. The current lack of a vaccine and the limited therapeutic options at hand make the development of new drugs against LASV a high priority. In the present study, we uncover that LASV entry into human cells via its major receptor, dystroglycan, involves an unusual pathway of macropinocytosis and define a set of cellular factors implicated in the regulation of LASV entry. A screen of kinase inhibitors revealed HGFR to be a possible candidate target for antiviral drugs against LASV. An HGFR candidate inhibitor currently being evaluated for cancer treatment showed potent antiviral activity and additive drug effects with ribavirin, which is used in the clinic to treat human LASV infection. In sum, our study reveals novel fundamental aspects of the LASV-host cell interaction and highlights a possible candidate drug target for therapeutic intervention.


Asunto(s)
Distroglicanos/metabolismo , Fiebre de Lassa/virología , Virus Lassa/patogenicidad , Pinocitosis/fisiología , Internalización del Virus , Células A549 , Endosomas/metabolismo , Endosomas/virología , Células Epiteliales/metabolismo , Células Epiteliales/virología , Humanos , Fiebre de Lassa/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores Virales/metabolismo , Transducción de Señal , Proteína del Síndrome de Wiskott-Aldrich/metabolismo
6.
J Virol ; 89(15): 7584-92, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25972533

RESUMEN

UNLABELLED: Lassa virus is a notorious human pathogen that infects many thousands of people each year in West Africa, causing severe viral hemorrhagic fevers and significant mortality. The surface glycoprotein of Lassa virus mediates receptor recognition through its GP1 subunit. Here we report the crystal structure of GP1 from Lassa virus, which is the first representative GP1 structure for Old World arenaviruses. We identify a unique triad of histidines that forms a binding site for LAMP1, a known lysosomal protein recently discovered to be a critical receptor for internalized Lassa virus at acidic pH. We demonstrate that mutation of this histidine triad, which is highly conserved among Old World arenaviruses, impairs LAMP1 recognition. Our biochemical and structural data further suggest that GP1 from Lassa virus may undergo irreversible conformational changes that could serve as an immunological decoy mechanism. Together with a variable region that we identify on the surface of GP1, those could be two distinct mechanisms that Lassa virus utilizes to avoid antibody-based immune response. IMPORTANCE: Structural data at atomic resolution for viral proteins is key for understanding their function at the molecular level and can facilitate novel avenues for combating viral infections. Here we used X-ray protein crystallography to decipher the crystal structure of the receptor-binding domain (GP1) from Lassa virus. This is a pathogenic virus that causes significant illness and mortality in West Africa. This structure reveals the overall architecture of GP1 domains from the group of viruses known as the Old World arenaviruses. Using this structural information, we elucidated the mechanisms for pH switch and binding of Lassa virus to LAMP1, a recently identified host receptor that is critical for successful infection. Lastly, our structural analysis suggests two novel immune evasion mechanisms that Lassa virus may utilize to escape antibody-based immune response.


Asunto(s)
Fiebre de Lassa/metabolismo , Virus Lassa/metabolismo , Proteínas de Membrana de los Lisosomas/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Secuencia de Aminoácidos , Línea Celular , Humanos , Fiebre de Lassa/genética , Fiebre de Lassa/virología , Virus Lassa/química , Virus Lassa/genética , Proteínas de Membrana de los Lisosomas/química , Proteínas de Membrana de los Lisosomas/genética , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Receptores Virales/química , Receptores Virales/genética , Receptores Virales/metabolismo , Alineación de Secuencia , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética
7.
J Virol ; 86(4): 2067-78, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22156524

RESUMEN

Although O-mannosylated dystroglycan is a receptor for Lassa virus, a causative agent of Lassa fever, recent findings suggest the existence of an alternative receptor(s). Here we identified four molecules as receptors for Lassa virus: Axl and Tyro3, from the TAM family, and dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin (DC-SIGN) and liver and lymph node sinusoidal endothelial calcium-dependent lectin (LSECtin), from the C-type lectin family. These molecules enhanced the binding of Lassa virus to cells and mediated infection independently of dystroglycan. Axl- or Tyro3-mediated infection required intracellular signaling via the tyrosine kinase activity of Axl or Tyro3, whereas DC-SIGN- or LSECtin-mediated infection and binding were dependent on a specific carbohydrate and on ions. The identification of these four molecules as Lassa virus receptors advances our understanding of Lassa virus cell entry.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Fiebre de Lassa/metabolismo , Virus Lassa/fisiología , Lectinas Tipo C/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Virales/metabolismo , Internalización del Virus , Moléculas de Adhesión Celular/genética , Línea Celular , Células Dendríticas/metabolismo , Células Dendríticas/virología , Distroglicanos/genética , Distroglicanos/metabolismo , Humanos , Fiebre de Lassa/genética , Fiebre de Lassa/virología , Virus Lassa/genética , Lectinas Tipo C/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Superficie Celular/genética , Receptores Virales/genética , Tirosina Quinasa del Receptor Axl
8.
Curr Med Chem ; 18(18): 2770-82, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21568902

RESUMEN

BACKGROUND: Subtilisin Kexin Isozyme-1 (SKI-1)/Site1Protease (S1P) is a Ca(+2)-dependent membrane bound pyrolysin-type serine protease of mammalian subtilase super family Proprotein Convertases (PCs)/Proprotein Convertase Subtilisin Kexins (PCSKs). It cleaves precursor proteins at the carboxy terminus of a non basic amino acid characterized by the sequence Arg/Lys-θ-φ-Leu/Ser/Thr↓, where θ = any amino acid except Cys, φ = the alkyl side chain containing hydrophobic amino acid. SKI-1 cleaves pro-BDNF, pro-SREBP2, pro-ATF6, pro-somatostatin and viral glycoproteins to generate their active forms. As a result SKI-1 plays important roles in cartilage development, bone mineralization, cholesterol metabolism, fatty acid synthesis and infections caused by Arina viruses of hemorrhagic type. Interest has grown to develop inhibitors of SKI-1 that may find useful therapeutic and biochemical applications. OBJECTIVE: Our objective is to develop small molecule inhibitors of SKI-1/S1P and study their kinetic and biochemical properties. RESULTS: Peptide analogs were designed by inserting a protease resistant methylene-oxy (-CH(2)-O-) pseudoamide function at the cleavage site of (251)Asp-Ile-Tyr-Ile-Ser-Arg-Arg-Leu-Leu↓Gly-Thr-Phe-Thr(263), derived from SKI-1 processing site of Lassa virus glycoprotein. The synthesis was conducted by substituting Leu-Gly with previously made Leu-CH(2)-O-Gly. Flexible linear and conformationally constrained circular and disulphide bridged cyclic peptides were prepared by solid phase method. Circular and cyclic peptides inhibited SKI-1 more potently (K(i)~14-20 µM) than the corresponding acyclic peptide (K(i)~51 µM). They also blocked SKI-1-mediated processing of pro-h(human)SREBP2 into its mature form in HepG2 cells. Circular pseudopeptides designed from hATF6 and hSREBP2 also inhibited SKI-1. This is the first report of circular and cyclic Ψ(CH(2)-O) containing peptides as SKI-1 inhibitors with potential therapeutic applications in cholesterol synthesis.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Péptidos Cíclicos/antagonistas & inhibidores , Proproteína Convertasas/antagonistas & inhibidores , Secuencia de Aminoácidos , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/patología , Evaluación de Medicamentos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Células Hep G2 , Humanos , Cinética , Fiebre de Lassa/metabolismo , Fiebre de Lassa/patología , Virus Lassa/metabolismo , Datos de Secuencia Molecular , Péptidos Cíclicos/química , Péptidos Cíclicos/metabolismo , Proproteína Convertasas/química , Proproteína Convertasas/metabolismo , Serina Endopeptidasas/química , Serina Endopeptidasas/metabolismo , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/química , Proteínas Virales/metabolismo
9.
Vopr Virusol ; 39(6): 257-60, 1994.
Artículo en Ruso | MEDLINE | ID: mdl-7536373

RESUMEN

Some immunity parameters (interferon, tumor necrosis factor, interleukin 1, etc.) were studied in CBA/Calac mice infected with Lassa virus. The results permit a hypothesis that a pathologic inflammatory reaction is responsible for the death of animals in experimental Lassa fever. One of the components of this reaction is endogenous shock involving a manifest production of immune response mediators, such as interferon, interleukin 1. and tumor necrosis factor.


Asunto(s)
Fiebre de Lassa/inmunología , Animales , Línea Celular , Chlorocebus aethiops , Citotoxicidad Inmunológica , Humanos , Interferones/biosíntesis , Interleucina-1/biosíntesis , Células Asesinas Naturales/inmunología , Fiebre de Lassa/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos CBA , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/biosíntesis , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA