Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 574(7777): 259-263, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31554973

RESUMEN

Chikungunya virus (CHIKV) is a re-emerging alphavirus that is transmitted to humans by mosquito bites and causes musculoskeletal and joint pain1,2. Despite intensive investigations, the human cellular factors that are critical for CHIKV infection remain unknown, hampering the understanding of viral pathogenesis and the development of anti-CHIKV therapies. Here we identified the four-and-a-half LIM domain protein 1 (FHL1)3 as a host factor that is required for CHIKV permissiveness and pathogenesis in humans and mice. Ablation of FHL1 expression results in the inhibition of infection by several CHIKV strains and o'nyong-nyong virus, but not by other alphaviruses and flaviviruses. Conversely, expression of FHL1 promotes CHIKV infection in cells that do not normally express it. FHL1 interacts directly with the hypervariable domain of the nsP3 protein of CHIKV and is essential for the replication of viral RNA. FHL1 is highly expressed in CHIKV-target cells and is particularly abundant in muscles3,4. Dermal fibroblasts and muscle cells derived from patients with Emery-Dreifuss muscular dystrophy that lack functional FHL15 are resistant to CHIKV infection. Furthermore,  CHIKV infection  is undetectable in Fhl1-knockout mice. Overall, this study shows that FHL1 is a key factor expressed by the host that enables CHIKV infection and identifies the interaction between nsP3 and FHL1 as a promising target for the development of anti-CHIKV therapies.


Asunto(s)
Fiebre Chikungunya/virología , Virus Chikungunya/patogenicidad , Factores Celulares Derivados del Huésped/metabolismo , Interacciones Huésped-Patógeno , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas con Dominio LIM/metabolismo , Proteínas Musculares/metabolismo , Animales , Células Cultivadas , Fiebre Chikungunya/tratamiento farmacológico , Virus Chikungunya/efectos de los fármacos , Virus Chikungunya/genética , Virus Chikungunya/crecimiento & desarrollo , Femenino , Fibroblastos/virología , Células HEK293 , Factores Celulares Derivados del Huésped/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas con Dominio LIM/deficiencia , Proteínas con Dominio LIM/genética , Masculino , Ratones , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Mioblastos/virología , Virus O'nyong-nyong/crecimiento & desarrollo , Virus O'nyong-nyong/patogenicidad , Unión Proteica , ARN Viral/biosíntesis , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Replicación Viral
2.
J Virol ; 96(7): e0196221, 2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35266803

RESUMEN

Dengue virus (DENV) is a mosquito-borne flavivirus responsible for dengue disease, a major human health concern for which no effective treatment is available. DENV relies heavily on the host cellular machinery for productive infection. Here, we show that the scaffold protein RACK1, which is part of the DENV replication complex, mediates infection by binding to the 40S ribosomal subunit. Mass spectrometry analysis of RACK1 partners coupled to an RNA interference screen-identified Vigilin and SERBP1 as DENV host-dependency factors. Both are RNA-binding proteins that interact with the DENV genome. Genetic ablation of Vigilin or SERBP1 rendered cells poorly susceptible to DENV, as well as related flaviviruses, by hampering the translation and replication steps. Finally, we established that a Vigilin or SERBP1 mutant lacking RACK1 binding but still interacting with the viral RNA is unable to mediate DENV infection. We propose that RACK1 recruits Vigilin and SERBP1, linking the DENV genome to the translation machinery for efficient infection. IMPORTANCE We recently identified the scaffolding RACK1 protein as an important host-dependency factor for dengue virus (DENV), a positive-stranded RNA virus responsible for the most prevalent mosquito-borne viral disease worldwide. Here, we have performed the first RACK1 interactome in human cells and identified Vigilin and SERBP1 as DENV host-dependency factors. Both are RNA-binding proteins that interact with the DENV RNA to regulate viral replication. Importantly, Vigilin and SERBP1 interact with RACK1 and the DENV viral RNA (vRNA) to mediate viral replication. Overall, our results suggest that RACK1 acts as a binding platform at the surface of the 40S ribosomal subunit to recruit Vigilin and SERBP1, which may therefore function as linkers between the viral RNA and the translation machinery to facilitate infection.


Asunto(s)
Virus del Dengue , Dengue , Proteínas de Unión al ARN , Animales , Dengue/fisiopatología , Virus del Dengue/fisiología , Interacciones Microbiota-Huesped/fisiología , Humanos , Proteínas de Neoplasias/metabolismo , ARN Viral/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Receptores de Cinasa C Activada/metabolismo , Replicación Viral
3.
Antimicrob Agents Chemother ; 66(8): e0008322, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35861550

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the infectious agent that has caused the current coronavirus disease (COVID) pandemic. Viral infection relies on the viral S (spike) protein/cellular receptor ACE2 interaction. Disrupting this interaction would lead to early blockage of viral replication. To identify chemical tools to further study these functional interfaces, 139,146 compounds from different chemical libraries were screened through an S/ACE2 in silico virtual molecular model. The best compounds were selected for further characterization using both cellular and biochemical approaches, reiterating SARS-CoV-2 entry and the S/ACE2 interaction. We report here two selected hits, bis-indolyl pyridine AB-00011778 and triphenylamine AB-00047476. Both of these compounds can block the infectivity of lentiviral vectors pseudotyped with the SARS-CoV-2 S protein as well as wild-type and circulating variant SARS-CoV-2 strains in various human cell lines, including pulmonary cells naturally susceptible to infection. AlphaLISA and biolayer interferometry confirmed a direct inhibitory effect of these drugs on the S/ACE2 association. A specific study of the AB-00011778 inhibitory properties showed that this drug inhibits viral replication with a 50% effective concentration (EC50) between 0.1 and 0.5 µM depending on the cell lines. Molecular docking calculations of the interaction parameters of the molecules within the S/ACE2 complex from both wild-type and circulating variants of the virus showed that the molecules may target multiple sites within the S/ACE2 interface. Our work indicates that AB-00011778 constitutes a good tool for modulating this interface and a strong lead compound for further therapeutic purposes.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , SARS-CoV-2 , Enzima Convertidora de Angiotensina 2 , Humanos , Simulación del Acoplamiento Molecular , Peptidil-Dipeptidasa A/química , Peptidil-Dipeptidasa A/metabolismo , Peptidil-Dipeptidasa A/farmacología , Unión Proteica , Piridinas/farmacología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus
4.
EMBO J ; 36(12): 1653-1668, 2017 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-28473450

RESUMEN

The cytopathic effects of Zika virus (ZIKV) are poorly characterized. Innate immunity controls ZIKV infection and disease in most infected patients through mechanisms that remain to be understood. Here, we studied the morphological cellular changes induced by ZIKV and addressed the role of interferon-induced transmembrane proteins (IFITM), a family of broad-spectrum antiviral factors, during viral replication. We report that ZIKV induces massive vacuolization followed by "implosive" cell death in human epithelial cells, primary skin fibroblasts and astrocytes, a phenomenon which is exacerbated when IFITM3 levels are low. It is reminiscent of paraptosis, a caspase-independent, non-apoptotic form of cell death associated with the formation of large cytoplasmic vacuoles. We further show that ZIKV-induced vacuoles are derived from the endoplasmic reticulum (ER) and dependent on the PI3K/Akt signaling axis. Inhibiting the Sec61 ER translocon in ZIKV-infected cells blocked vacuole formation and viral production. Our results provide mechanistic insight behind the ZIKV-induced cytopathic effect and indicate that IFITM3, by acting as a gatekeeper for incoming virus, restricts virus takeover of the ER and subsequent cell death.


Asunto(s)
Astrocitos/virología , Muerte Celular , Efecto Citopatogénico Viral , Células Epiteliales/virología , Fibroblastos/virología , Vacuolas/metabolismo , Virus Zika/patogenicidad , Astrocitos/citología , Astrocitos/fisiología , Células Cultivadas , Retículo Endoplásmico/metabolismo , Células Epiteliales/citología , Células Epiteliales/fisiología , Fibroblastos/citología , Fibroblastos/fisiología , Humanos , Proteínas de la Membrana/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas de Unión al ARN/metabolismo , Canales de Translocación SEC/metabolismo , Transducción de Señal
5.
J Virol ; 94(7)2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-31915280

RESUMEN

Dengue virus (DENV) is a mosquito-borne flavivirus responsible for dengue disease, a major human health concern for which no specific therapies are available. Like other viruses, DENV relies heavily on the host cellular machinery for productive infection. In this study, we performed a genome-wide CRISPR-Cas9 screen using haploid HAP1 cells to identify host genes important for DENV infection. We identified DPM1 and -3, two subunits of the endoplasmic reticulum (ER) resident dolichol-phosphate mannose synthase (DPMS) complex, as host dependency factors for DENV and other related flaviviruses, such as Zika virus (ZIKV). The DPMS complex catalyzes the synthesis of dolichol-phosphate mannose (DPM), which serves as mannosyl donor in pathways leading to N-glycosylation, glycosylphosphatidylinositol (GPI) anchor biosynthesis, and C- or O-mannosylation of proteins in the ER lumen. Mutation in the DXD motif of DPM1, which is essential for its catalytic activity, abolished DPMS-mediated DENV infection. Similarly, genetic ablation of ALG3, a mannosyltransferase that transfers mannose to lipid-linked oligosaccharide (LLO), rendered cells poorly susceptible to DENV. We also established that in cells deficient for DPMS activity, viral RNA amplification is hampered and truncated oligosaccharides are transferred to the viral prM and E glycoproteins, affecting their proper folding. Overall, our study provides new insights into the host-dependent mechanisms of DENV infection and supports current therapeutic approaches using glycosylation inhibitors to treat DENV infection.IMPORTANCE Dengue disease, which is caused by dengue virus (DENV), has emerged as the most important mosquito-borne viral disease in humans and is a major global health concern. DENV encodes only few proteins and relies on the host cell machinery to accomplish its life cycle. The identification of the host factors important for DENV infection is needed to propose new targets for antiviral intervention. Using a genome-wide CRISPR-Cas9 screen, we identified DPM1 and -3, two subunits of the DPMS complex, as important host factors for the replication of DENV as well as other related viruses such as Zika virus. We established that DPMS complex plays dual roles during viral infection, both regulating viral RNA replication and promoting viral structural glycoprotein folding/stability. These results provide insights into the host molecules exploited by DENV and other flaviviruses to facilitate their life cycle.


Asunto(s)
Sistemas CRISPR-Cas , Virus del Dengue/fisiología , Dengue/virología , Manosiltransferasas/metabolismo , Animales , Chlorocebus aethiops , Retículo Endoplásmico/metabolismo , Fibroblastos/metabolismo , Glicoproteínas/metabolismo , Glicosilación , Glicosilfosfatidilinositoles/metabolismo , Células HEK293 , Humanos , Manosa/química , Oligosacáridos/química , ARN Guía de Kinetoplastida/metabolismo , ARN Viral/química , Células Vero , Replicación Viral
6.
J Virol ; 90(1): 92-102, 2016 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-26468529

RESUMEN

UNLABELLED: Dengue virus (DENV) is the etiological agent of the major human arboviral disease. We previously demonstrated that the TIM and TAM families of phosphatidylserine (PtdSer) receptors involved in the phagocytosis of apoptotic cells mediate DENV entry into target cells. We show here that human CD300a, a recently identified phospholipid receptor, also binds directly DENV particles and enhances viral entry. CD300a facilitates infection of the four DENV serotypes, as well as of other mosquito-borne viruses such as West Nile virus and Chikungunya virus. CD300a acts as an attachment factor that enhances DENV internalization through clathrin-mediated endocytosis. CD300a recognizes predominantly phosphatidylethanolamine (PtdEth) and to a lesser extent PtdSer associated with viral particles. Mutation of residues in the IgV domain critical for phospholipid binding abrogate CD300a-mediated enhancement of DENV infection. Finally, we show that CD300a is expressed at the surface of primary macrophages and anti-CD300a polyclonal antibodies partially inhibited DENV infection of these cells. Overall, these data indicate that CD300a is a novel DENV binding receptor that recognizes PtdEth and PtdSer present on virions and enhance infection. IMPORTANCE: Dengue disease, caused by dengue virus (DENV), has emerged as the most important mosquito-borne viral disease of humans and is a major global health concern. The molecular bases of DENV-host cell interactions during virus entry are poorly understood, hampering the discovery of new targets for antiviral intervention. We recently discovered that the TIM and TAM proteins, two receptor families involved in the phosphatidylserine (PtdSer)-dependent phagocytic removal of apoptotic cells, interact with DENV particles-associated PtdSer through a mechanism that mimics the recognition of apoptotic cells and mediate DENV infection. In this study, we show that CD300a, a novel identified phospholipid receptor, mediates DENV infection. CD300a-dependent DENV infection relies on the direct recognition of phosphatidylethanolamine and to a lesser extent PtdSer associated with viral particles. This study provides novel insights into the mechanisms that mediate DENV entry and reinforce the concept that DENV uses an apoptotic mimicry strategy for viral entry.


Asunto(s)
Antígenos CD/metabolismo , Virus del Dengue/fisiología , Interacciones Huésped-Patógeno , Receptores Inmunológicos/metabolismo , Receptores Virales/metabolismo , Internalización del Virus , Línea Celular , Virus Chikungunya/fisiología , Endocitosis , Humanos , Macrófagos/química , Proteínas de la Membrana/análisis , Fosfatidiletanolaminas/metabolismo , Fosfatidilserinas/metabolismo , Unión Proteica , Virus del Nilo Occidental/fisiología
7.
Virologie (Montrouge) ; 18(6): 325-336, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-33065925

RESUMEN

Apoptotic cells clearance, or efferocytosis, is an essential and highly conserved cellular process mainly based on the recognition of the phosphatidylserine (PtdSer) exposed on the surface of apoptotic bodies by the phagocyte. Since a decade, several studies have shown that many viruses can hijack this biological process by exposing PtdSer on their viral envelope to facilitate infection. This apoptotic mimicry concept has been recently strengthened by recent discoveries showing that multiple enveloped virus families bind directly or indirectly to PtdSer receptors in order to initiate their infectious cycle. This review focus on recent advances in this topic and discuss about PtdSer receptors function, especially TIM (T-Cell Immunoglobulin and Mucin domain) and TAM (Tyro3, Axl, Mer) families, during infection and viral entry.

8.
Methods Mol Biol ; 2824: 203-219, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39039415

RESUMEN

Like all the RNA viruses, Rift Valley fever virus (RVFV) encodes only few viral proteins and relies heavily on the host cellular machinery for productive infection. This dependence creates a potential "Achille's heel" that may be exploited to develop new approaches to treat RVFV infection. The recent development of lentiviral sgRNAs pool has enabled the creation of genome-scale CRISPR-Cas9 knockout libraries that has been used to identify host factors required for virus replication. In this chapter, we describe the preparation and execution of a pooled CRISPR-Cas9 loss-of-function screen using virus-induced cell death phenotypic readout. Using this technique, we outline a strategy for the identification of host factors essential for important human emerging viruses such as RVFV.


Asunto(s)
Sistemas CRISPR-Cas , Virus de la Fiebre del Valle del Rift , Humanos , Virus de la Fiebre del Valle del Rift/genética , Replicación Viral/genética , Interacciones Huésped-Patógeno/genética , Técnicas de Inactivación de Genes , ARN Guía de Sistemas CRISPR-Cas/genética
9.
Nat Commun ; 15(1): 640, 2024 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-38245532

RESUMEN

Considerable progress has been made in understanding the molecular host-virus battlefield during SARS-CoV-2 infection. Nevertheless, the assembly and egress of newly formed virions are less understood. To identify host proteins involved in viral morphogenesis, we characterize the proteome of SARS-CoV-2 virions produced from A549-ACE2 and Calu-3 cells, isolated via ultracentrifugation on sucrose cushion or by ACE-2 affinity capture. Bioinformatic analysis unveils 92 SARS-CoV-2 virion-associated host factors, providing a valuable resource to better understand the molecular environment of virion production. We reveal that G3BP1 and G3BP2 (G3BP1/2), two major stress granule nucleators, are embedded within virions and unexpectedly favor virion production. Furthermore, we show that G3BP1/2 participate in the formation of cytoplasmic membrane vesicles, that are likely virion assembly sites, consistent with a proviral role of G3BP1/2 in SARS-CoV-2 dissemination. Altogether, these findings provide new insights into host factors required for SARS-CoV-2 assembly with potential implications for future therapeutic targeting.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/metabolismo , Replicación Viral , ADN Helicasas/metabolismo , Proteómica , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , COVID-19/metabolismo , ARN Helicasas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Ensamble de Virus , Virión/metabolismo
10.
J Virol ; 85(6): 2980-9, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21191016

RESUMEN

The ubiquitin ligase CBLL1 (also known as HAKAI) has been proposed to be a critical cellular factor exploited by West Nile virus (WNV) for productive infection. CBLL1 has emerged as a major hit in a recent RNA interference screen designed to identify cellular factors required for the early stages of the WNV life cycle. Follow-up experiments showed that HeLa cells knocked down for CBLL1 by a small interfering RNA (siRNA) failed to internalize WNV particles and resisted infection. Furthermore, depletion of a free-ubiquitin pool by the proteasome inhibitor MG132 abolished WNV endocytosis, suggesting that CBLL1 acts in concert with the ubiquitin proteasome system to mediate virus internalization. Here, we examined the effect of CBLL1 knockdown and proteasome inhibitors on infection by WNV and other flaviviruses. We identified new siRNAs that repress the CBLL1 protein and strongly inhibit the endocytosis of Listeria monocytogenes, a bacterial pathogen known to require CBLL1 to invade host cells. Strikingly, however, we detected efficient WNV, dengue virus, and yellow fever virus infection of human cells, despite potent downregulation of CBLL1 by RNA interference. In addition, we found that the proteasome inhibitors MG132 and lactacystin did not affect WNV internalization but strongly repressed flavivirus RNA translation and replication. Together, these data do not support a requirement for CBLL1 during flavivirus entry and rather suggest an essential role of the ubiquitin/proteasome pathway for flavivirus genome amplification.


Asunto(s)
Virus del Dengue/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Internalización del Virus , Replicación Viral , Virus del Nilo Occidental/fisiología , Virus de la Fiebre Amarilla/fisiología , Acetilcisteína/análogos & derivados , Acetilcisteína/metabolismo , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Leupeptinas/metabolismo , Listeria monocytogenes/patogenicidad , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , ARN Interferente Pequeño/metabolismo , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores
11.
J Infect Dis ; 203(9): 1316-23, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21459818

RESUMEN

BACKGROUND: The presence and origin of endemic foci of human T-lymphotropic virus type 2 (HTLV2) infection in Africa remain a matter of debate. METHODS: To better appreciate such determinants, we performed a survey of 1918 inhabitants from Cameroon forest areas, including 1051 Bakola Pygmies and 867 Bantus. RESULTS: The overall HTLV-1/2 seroprevalence was 4% (49 cases of HTLV-1 and 27 cases of HTLV-2 infection). Both infections were mainly restricted to the Bakola Pygmies, with surprisingly no HTLV-2 infections in the Bantu population. Both HTLV-1 and HTLV-2 seroprevalences increased with age. There was evidence of ongoing HTLV-2 transmission in this population. Lymphoid T cell lines producing HTLV-2 were established. HTLV-2 long terminal repeat sequences (672 base pairs) obtained from 7 infected Bakola were highly similar to each other (<1% nucleotide divergence), as well as to Amerindian HTLV-2B strains. Analyses on a complete sequence (8954 base pairs) confirmed that it was a typical HTLV-2 subtype B strain. Along with molecular clock analysis, these data strongly suggest that HTLV-2 has been endemic in the Bakola Pygmy population for a long time. CONCLUSIONS: This study demonstrates clearly an HTLV-2 endemicity with ongoing transmission in an African population. Furthermore, it give insights into central questions regarding the origins and evolution rate of HTLV-2 and the migrations of infected populations.


Asunto(s)
Enfermedades Endémicas , Infecciones por HTLV-II/epidemiología , Virus Linfotrópico T Tipo 2 Humano/aislamiento & purificación , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Camerún/epidemiología , Niño , Preescolar , Análisis por Conglomerados , Femenino , Virus Linfotrópico T Tipo 1 Humano/aislamiento & purificación , Virus Linfotrópico T Tipo 2 Humano/genética , Humanos , Masculino , Persona de Mediana Edad , Epidemiología Molecular , Datos de Secuencia Molecular , Grupos de Población , ARN Viral/genética , Análisis de Secuencia de ADN , Homología de Secuencia , Estudios Seroepidemiológicos , Secuencias Repetidas Terminales , Adulto Joven
12.
Cell Rep ; 39(4): 110744, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35477000

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the COVID-19 pandemic, which has led to a devastating global health crisis. The emergence of variants that escape neutralizing responses emphasizes the urgent need to deepen our understanding of SARS-CoV-2 biology. Using a comprehensive identification of RNA-binding proteins (RBPs) by mass spectrometry (ChIRP-MS) approach, we identify 107 high-confidence cellular factors that interact with the SARS-CoV-2 genome during infection. By systematically knocking down their expression in human lung epithelial cells, we find that the majority of the identified RBPs are SARS-CoV-2 proviral factors. In particular, we show that HNRNPA2B1, ILF3, QKI, and SFPQ interact with the SARS-CoV-2 genome and promote viral RNA amplification. Our study provides valuable resources for future investigations into the mechanisms of SARS-CoV-2 replication and the identification of host-centered antiviral therapies.


Asunto(s)
COVID-19 , ARN Viral , COVID-19/genética , Humanos , Pandemias , ARN Viral/genética , SARS-CoV-2/genética , Replicación Viral/genética
13.
J Exp Med ; 218(4)2021 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-33533916

RESUMEN

Several studies have analyzed antiviral immune pathways in late-stage severe COVID-19. However, the initial steps of SARS-CoV-2 antiviral immunity are poorly understood. Here we have isolated primary SARS-CoV-2 viral strains and studied their interaction with human plasmacytoid predendritic cells (pDCs), a key player in antiviral immunity. We show that pDCs are not productively infected by SARS-CoV-2. However, they efficiently diversified into activated P1-, P2-, and P3-pDC effector subsets in response to viral stimulation. They expressed CD80, CD86, CCR7, and OX40 ligand at levels similar to influenza virus-induced activation. They rapidly produced high levels of interferon-α, interferon-λ1, IL-6, IP-10, and IL-8. All major aspects of SARS-CoV-2-induced pDC activation were inhibited by hydroxychloroquine. Mechanistically, SARS-CoV-2-induced pDC activation critically depended on IRAK4 and UNC93B1, as established using pDC from genetically deficient patients. Overall, our data indicate that human pDC are efficiently activated by SARS-CoV-2 particles and may thus contribute to type I IFN-dependent immunity against SARS-CoV-2 infection.


Asunto(s)
COVID-19/inmunología , COVID-19/metabolismo , Plasticidad de la Célula/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Proteínas de Transporte de Membrana/metabolismo , SARS-CoV-2/inmunología , Biomarcadores , COVID-19/virología , Citocinas/metabolismo , Células Dendríticas/virología , Interacciones Huésped-Patógeno/inmunología , Humanos , Hidroxicloroquina/farmacología , Hidroxicloroquina/uso terapéutico , Inmunomodulación , Inmunofenotipificación , Mediadores de Inflamación/metabolismo , Interferón Tipo I/metabolismo , Interferones/metabolismo , Interferón lambda , Tratamiento Farmacológico de COVID-19
14.
bioRxiv ; 2021 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-33442685

RESUMEN

Several studies have analyzed antiviral immune pathways in late-stage severe COVID-19. However, the initial steps of SARS-CoV-2 antiviral immunity are poorly understood. Here, we have isolated primary SARS-CoV-2 viral strains, and studied their interaction with human plasmacytoid pre-dendritic cells (pDC), a key player in antiviral immunity. We show that pDC are not productively infected by SARS-CoV-2. However, they efficiently diversified into activated P1-, P2-, and P3-pDC effector subsets in response to viral stimulation. They expressed CD80, CD86, CCR7, and OX40 ligand at levels similar to influenza virus-induced activation. They rapidly produced high levels of interferon-α, interferon-λ1, IL-6, IP-10, and IL-8. All major aspects of SARS-CoV-2-induced pDC activation were inhibited by hydroxychloroquine. Mechanistically, SARS-CoV-2-induced pDC activation critically depended on IRAK4 and UNC93B1, as established using pDC from genetically deficient patients. Overall, our data indicate that human pDC are efficiently activated by SARS-CoV-2 particles and may thus contribute to type I IFN-dependent immunity against SARS-CoV-2 infection.

15.
C R Biol ; 343(4): 79-89, 2021 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-33988325

RESUMEN

Chikungunya is an infectious disease caused by the chikungunya virus (CHIKV), an alphavirus transmitted to humans by Aedes mosquitoes, and for which there is no licensed vaccine nor antiviral treatments. By using a loss-of-function genetic screen, we have recently identified the FHL1 protein as an essential host factor for CHIKV tropism and pathogenesis. FHL1 is highly expressed in muscles cells and fibroblasts, the main CHIKV-target cells. FHL1 interacts with the viral protein nsP3 and plays a critical role in CHIKV genome amplification. Experiments in vivo performed in FHL1-deficient mice have shown that these animals are resistant to infection and do not develop muscular lesions. Altogether these observations, published in the journal Nature [1], show that FHL1 is a key host factor for CHIKV pathogenesis and identify the interaction between FHL1 and nsP3 as a promising target for the development of new antiviral strategies.


Le chikungunya est une maladie infectieuse causée par le virus chikungunya (CHIKV), un alphavirus transmis à l'Homme par les moustiques Aedes et contre lequel il n'existe ni vaccin, ni traitements antiviraux. En utilisant une approche de crible génétique par perte de fonction, nous avons récemment identifié la protéine FHL1 comme un facteur cellulaire essentiel pour le tropisme et la pathogénèse du CHIKV. FHL1 est une molécule présente majoritairement dans les cellules musculaires et les fibroblastes, les cibles privilégiées de CHIKV. FHL1 interagit avec la protéine virale nsP3 et joue un rôle décisif dans le mécanisme d'amplification du génome de CHIKV. Des expériences in vivo chez des souris déficientes pour FHL1 ont montré que ces animaux sont résistants à l'infection et ne développent pas de lésions musculaires. L'ensemble de ces observations publiées dans la revue Nature [1] montrent que FHL1 est un facteur cellulaire clé pour la pathogénèse de CHIKV et identifient l'interaction entre FHL1 et nsp3 comme une cible prometteuse pour le développement de nouvelles stratégies antivirales.


Asunto(s)
Fiebre Chikungunya , Virus Chikungunya , Animales , Virus Chikungunya/genética , Péptidos y Proteínas de Señalización Intracelular , Proteínas con Dominio LIM , Ratones , Proteínas Musculares , Tropismo , Proteínas no Estructurales Virales , Replicación Viral
16.
Sci Immunol ; 6(62)2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34413140

RESUMEN

Autosomal inborn errors of type I IFN immunity and autoantibodies against these cytokines underlie at least 10% of critical COVID-19 pneumonia cases. We report very rare, biochemically deleterious X-linked TLR7 variants in 16 unrelated male individuals aged 7 to 71 years (mean: 36.7 years) from a cohort of 1,202 male patients aged 0.5 to 99 years (mean: 52.9 years) with unexplained critical COVID-19 pneumonia. None of the 331 asymptomatically or mildly infected male individuals aged 1.3 to 102 years (mean: 38.7 years) tested carry such TLR7 variants (p = 3.5 × 10-5). The phenotypes of five hemizygous relatives of index cases infected with SARS-CoV-2 include asymptomatic or mild infection (n=2, 5 and 38 years), or moderate (n=1, 5 years), severe (n=1, 27 years), or critical (n=1, 29 years) pneumonia. Two boys (aged 7 and 12 years) from a cohort of 262 male patients with severe COVID-19 pneumonia (mean: 51.0 years) are hemizygous for a deleterious TLR7 variant. The cumulative allele frequency for deleterious TLR7 variants in the male general population is < 6.5x10-4 We also show that blood B cell lines and myeloid cell subsets from the patients do not respond to TLR7 stimulation, a phenotype rescued by wild-type TLR7 The patients' blood plasmacytoid dendritic cells (pDCs) produce low levels of type I IFNs in response to SARS-CoV-2. Overall, X-linked recessive TLR7 deficiency is a highly penetrant genetic etiology of critical COVID-19 pneumonia, in about 1.8% of male patients below the age of 60 years. Human TLR7 and pDCs are essential for protective type I IFN immunity against SARS-CoV-2 in the respiratory tract.


Asunto(s)
COVID-19/complicaciones , Enfermedades Genéticas Ligadas al Cromosoma X/complicaciones , Enfermedades del Sistema Inmune/complicaciones , Receptor Toll-Like 7/deficiencia , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Niño , Preescolar , Humanos , Lactante , Masculino , Persona de Mediana Edad , Linaje , Penetrancia , Receptor Toll-Like 7/genética , Adulto Joven
17.
J Virol ; 83(11): 5477-84, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19297469

RESUMEN

Claudin-1, a component of tight junctions between liver hepatocytes, is a hepatitis C virus (HCV) late-stage entry cofactor. To investigate the structural and functional roles of various claudin-1 domains in HCV entry, we applied a mutagenesis strategy. Putative functional intracellular claudin-1 domains were not important. However, we identified seven novel residues in the first extracellular loop that are critical for entry of HCV isolates drawn from six different subtypes. Most of the critical residues belong to the highly conserved claudin motif W(30)-GLW(51)-C(54)-C(64). Alanine substitutions of these residues did not impair claudin-1 cell surface expression or lateral protein interactions within the plasma membrane, including claudin-1-claudin-1 and claudin-1-CD81 interactions. However, these mutants no longer localized to cell-cell contacts. Based on our observations, we propose that cell-cell contacts formed by claudin-1 may generate specialized membrane domains that are amenable to HCV entry.


Asunto(s)
Comunicación Celular , Hepacivirus/fisiología , Proteínas de la Membrana/metabolismo , Internalización del Virus , Secuencias de Aminoácidos , Línea Celular , Membrana Celular/metabolismo , Claudina-1 , Espacio Extracelular/metabolismo , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Mutación/genética
18.
J Virol ; 82(7): 3555-60, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18234789

RESUMEN

Hepatitis C virus (HCV) is a major cause of liver disease in humans. The CD81 tetraspanin is necessary but not sufficient for HCV penetration into hepatocytes, and it was recently reported that the tight junction protein claudin-1 is a critical HCV entry cofactor. Here, we confirm the role of claudin-1 in HCV entry. In addition, we show that claudin-6 and claudin-9 expressed in CD81(+) cells also enable the entry of HCV pseudoparticles derived from six of the major genotypes. Whereas claudin-1, -6, and -9 function equally well as entry cofactors in endothelial cells, claudin-1 is more efficient in hepatoma cells. This suggests that additional cellular factors modulate the ability of claudins to function as HCV entry cofactors. Our work has generated novel and essential means to investigate the mechanism of HCV penetration into hepatocytes and the role of the claudin protein family in HCV dissemination, replication, and pathogenesis.


Asunto(s)
Hepacivirus/fisiología , Proteínas de la Membrana/fisiología , Receptores Virales/fisiología , Internalización del Virus , Línea Celular , Claudina-1 , Claudinas , Células Endoteliales/virología , Silenciador del Gen , Hepatocitos/virología , Humanos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
19.
Cell Rep ; 23(6): 1779-1793, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29742433

RESUMEN

Dengue virus (DENV) is a major human pathogen causing millions of infections yearly. Despite intensive investigations, a DENV receptor that directly participates in virus internalization has not yet been characterized. Here, we report that the phosphatidylserine receptor TIM-1 is an authentic DENV entry receptor that plays an active role in virus endocytosis. Genetic ablation of TIM-1 strongly impaired DENV infection. Total internal reflection fluorescence microscopy analyses of live infected cells show that TIM-1 is mostly confined in clathrin-coated pits and is co-internalized with DENV during viral entry. TIM-1 is ubiquitinated at two lysine residues of its cytoplasmic domain, and this modification is required for DENV endocytosis. Furthermore, STAM-1, a component of the ESCRT-0 complex involved in intracellular trafficking of ubiquitinated cargos, interacts with TIM-1 and is required for DENV infection. Overall, our results show that TIM-1 is the first bona fide receptor identified for DENV.


Asunto(s)
Virus del Dengue/fisiología , Dengue/virología , Receptor Celular 1 del Virus de la Hepatitis A/metabolismo , Ubiquitinación , Internalización del Virus , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Línea Celular Tumoral , Virus del Dengue/ultraestructura , Endocitosis , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Eliminación de Gen , Receptor Celular 1 del Virus de la Hepatitis A/química , Receptor Celular 1 del Virus de la Hepatitis A/genética , Humanos , Fosfoproteínas/metabolismo , Unión Proteica , Dominios Proteicos , Proteómica
20.
Cell Rep ; 21(13): 3900-3913, 2017 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-29281836

RESUMEN

Dengue virus (DENV) infections cause the most prevalent mosquito-borne viral disease worldwide, for which no therapies are available. DENV encodes seven non-structural (NS) proteins that co-assemble and recruit poorly characterized host factors to form the DENV replication complex essential for viral infection. Here, we provide a global proteomic analysis of the human host factors that interact with the DENV NS1 protein. Combined with a functional RNAi screen, this study reveals a comprehensive network of host cellular processes involved in DENV infection and identifies DENV host restriction and dependency factors. We highlight an important role of RACK1 and the chaperonin TRiC (CCT) and oligosaccharyltransferase (OST) complexes during DENV replication. We further show that the OST complex mediates NS1 and NS4B glycosylation, and pharmacological inhibition of its N-glycosylation function strongly impairs DENV infection. In conclusion, our study provides a global interactome of the DENV NS1 and identifies host factors targetable for antiviral therapies.


Asunto(s)
Virus del Dengue/metabolismo , Interacciones Huésped-Patógeno , Mapas de Interacción de Proteínas , Proteínas no Estructurales Virales/metabolismo , Dengue/virología , Glicosilación , Células HEK293 , Células HeLa , Humanos , Complejos Multiproteicos/metabolismo , Proteínas de Neoplasias/metabolismo , ARN Interferente Pequeño/metabolismo , Receptores de Cinasa C Activada/metabolismo , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA