Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.175
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Immunol ; 37: 73-95, 2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-31026414

RESUMEN

Neurotropic RNA viruses continue to emerge and are increasingly linked to diseases of the central nervous system (CNS) despite viral clearance. Indeed, the overall mortality of viral encephalitis in immunocompetent individuals is low, suggesting efficient mechanisms of virologic control within the CNS. Both immune and neural cells participate in this process, which requires extensive innate immune signaling between resident and infiltrating cells, including microglia and monocytes, that regulate the effector functions of antiviral T and B cells as they gain access to CNS compartments. While these interactions promote viral clearance via mainly neuroprotective mechanisms, they may also promote neuropathology and, in some cases, induce persistent alterations in CNS physiology and function that manifest as neurologic and psychiatric diseases. This review discusses mechanisms of RNA virus clearance and neurotoxicity during viral encephalitis with a focus on the cytokines essential for immune and neural cell inflammatory responses and interactions. Understanding neuroimmune communications in the setting of viral infections is essential for the development of treatments that augment neuroprotective processes while limiting ongoing immunopathological processes that cause ongoing CNS disease.


Asunto(s)
Encéfalo/inmunología , Inmunidad Innata , Microglía/fisiología , Infecciones por Virus ARN/inmunología , Virus ARN/fisiología , Animales , Barrera Hematoencefálica , Encéfalo/virología , Humanos , Inflamación Neurogénica , Neuroinmunomodulación
2.
Cell ; 186(22): 4834-4850.e23, 2023 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-37794589

RESUMEN

Regulation of viral RNA biogenesis is fundamental to productive SARS-CoV-2 infection. To characterize host RNA-binding proteins (RBPs) involved in this process, we biochemically identified proteins bound to genomic and subgenomic SARS-CoV-2 RNAs. We find that the host protein SND1 binds the 5' end of negative-sense viral RNA and is required for SARS-CoV-2 RNA synthesis. SND1-depleted cells form smaller replication organelles and display diminished virus growth kinetics. We discover that NSP9, a viral RBP and direct SND1 interaction partner, is covalently linked to the 5' ends of positive- and negative-sense RNAs produced during infection. These linkages occur at replication-transcription initiation sites, consistent with NSP9 priming viral RNA synthesis. Mechanistically, SND1 remodels NSP9 occupancy and alters the covalent linkage of NSP9 to initiating nucleotides in viral RNA. Our findings implicate NSP9 in the initiation of SARS-CoV-2 RNA synthesis and unravel an unsuspected role of a cellular protein in orchestrating viral RNA production.


Asunto(s)
COVID-19 , ARN Viral , Humanos , COVID-19/metabolismo , Endonucleasas/metabolismo , ARN Viral/metabolismo , SARS-CoV-2/genética , Replicación Viral
3.
Cell ; 185(21): 4023-4037.e18, 2022 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-36174579

RESUMEN

High-throughput RNA sequencing offers broad opportunities to explore the Earth RNA virome. Mining 5,150 diverse metatranscriptomes uncovered >2.5 million RNA virus contigs. Analysis of >330,000 RNA-dependent RNA polymerases (RdRPs) shows that this expansion corresponds to a 5-fold increase of the known RNA virus diversity. Gene content analysis revealed multiple protein domains previously not found in RNA viruses and implicated in virus-host interactions. Extended RdRP phylogeny supports the monophyly of the five established phyla and reveals two putative additional bacteriophage phyla and numerous putative additional classes and orders. The dramatically expanded phylum Lenarviricota, consisting of bacterial and related eukaryotic viruses, now accounts for a third of the RNA virome. Identification of CRISPR spacer matches and bacteriolytic proteins suggests that subsets of picobirnaviruses and partitiviruses, previously associated with eukaryotes, infect prokaryotic hosts.


Asunto(s)
Bacteriófagos , Virus ARN , Bacteriófagos/genética , ARN Polimerasas Dirigidas por ADN/genética , Genoma Viral , Filogenia , ARN , Virus ARN/genética , ARN Polimerasa Dependiente del ARN/genética , Viroma
4.
Cell ; 184(9): 2394-2411.e16, 2021 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-33743211

RESUMEN

SARS-CoV-2 is the cause of a pandemic with growing global mortality. Using comprehensive identification of RNA-binding proteins by mass spectrometry (ChIRP-MS), we identified 309 host proteins that bind the SARS-CoV-2 RNA during active infection. Integration of this data with ChIRP-MS data from three other RNA viruses defined viral specificity of RNA-host protein interactions. Targeted CRISPR screens revealed that the majority of functional RNA-binding proteins protect the host from virus-induced cell death, and comparative CRISPR screens across seven RNA viruses revealed shared and SARS-specific antiviral factors. Finally, by combining the RNA-centric approach and functional CRISPR screens, we demonstrated a physical and functional connection between SARS-CoV-2 and mitochondria, highlighting this organelle as a general platform for antiviral activity. Altogether, these data provide a comprehensive catalog of functional SARS-CoV-2 RNA-host protein interactions, which may inform studies to understand the host-virus interface and nominate host pathways that could be targeted for therapeutic benefit.


Asunto(s)
Interacciones Huésped-Patógeno , ARN Viral/genética , SARS-CoV-2/genética , Animales , COVID-19/virología , Sistemas CRISPR-Cas/genética , Línea Celular Tumoral , Chlorocebus aethiops , Femenino , Genoma Viral , Humanos , Pulmón/virología , Masculino , Espectrometría de Masas , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Proteoma/metabolismo , Proteínas de Unión al ARN/metabolismo , SARS-CoV-2/ultraestructura , Células Vero
5.
Cell ; 183(7): 1930-1945.e23, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33188777

RESUMEN

RNA viruses are among the most prevalent pathogens and are a major burden on society. Although RNA viruses have been studied extensively, little is known about the processes that occur during the first several hours of infection because of a lack of sensitive assays. Here we develop a single-molecule imaging assay, virus infection real-time imaging (VIRIM), to study translation and replication of individual RNA viruses in live cells. VIRIM uncovered a striking heterogeneity in replication dynamics between cells and revealed extensive coordination between translation and replication of single viral RNAs. Furthermore, using VIRIM, we identify the replication step of the incoming viral RNA as a major bottleneck of successful infection and identify host genes that are responsible for inhibition of early virus replication. Single-molecule imaging of virus infection is a powerful tool to study virus replication and virus-host interactions that may be broadly applicable to RNA viruses.


Asunto(s)
Biosíntesis de Proteínas , Virus ARN/fisiología , Replicación Viral/fisiología , Línea Celular Tumoral , Supervivencia Celular , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Interferones/metabolismo , Transporte de ARN , ARN Viral/genética , Reproducibilidad de los Resultados , Imagen Individual de Molécula , Factores de Tiempo
6.
Cell ; 167(4): 1088-1098.e6, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27814506

RESUMEN

The magnitude of the 2013-2016 Ebola virus disease (EVD) epidemic enabled an unprecedented number of viral mutations to occur over successive human-to-human transmission events, increasing the probability that adaptation to the human host occurred during the outbreak. We investigated one nonsynonymous mutation, Ebola virus (EBOV) glycoprotein (GP) mutant A82V, for its effect on viral infectivity. This mutation, located at the NPC1-binding site on EBOV GP, occurred early in the 2013-2016 outbreak and rose to high frequency. We found that GP-A82V had heightened ability to infect primate cells, including human dendritic cells. The increased infectivity was restricted to cells that have primate-specific NPC1 sequences at the EBOV interface, suggesting that this mutation was indeed an adaptation to the human host. GP-A82V was associated with increased mortality, consistent with the hypothesis that the heightened intrinsic infectivity of GP-A82V contributed to disease severity during the EVD epidemic.


Asunto(s)
Ebolavirus/genética , Ebolavirus/patogenicidad , Fiebre Hemorrágica Ebola/virología , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética , África Occidental/epidemiología , Sustitución de Aminoácidos , Animales , Callithrix , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Cheirogaleidae , Citoplasma/virología , Ebolavirus/fisiología , Fiebre Hemorrágica Ebola/epidemiología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Proteína Niemann-Pick C1 , Conformación Proteica en Hélice alfa , Proteínas del Envoltorio Viral/metabolismo , Virión/química , Virión/patogenicidad , Virulencia
7.
Mol Cell ; 81(3): 584-598.e5, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33444546

RESUMEN

Severe-acute-respiratory-syndrome-related coronavirus 2 (SARS-CoV-2) is the positive-sense RNA virus that causes coronavirus disease 2019 (COVID-19). The genome of SARS-CoV-2 is unique among viral RNAs in its vast potential to form RNA structures, yet as much as 97% of its 30 kilobases have not been structurally explored. Here, we apply a novel long amplicon strategy to determine the secondary structure of the SARS-CoV-2 RNA genome at single-nucleotide resolution in infected cells. Our in-depth structural analysis reveals networks of well-folded RNA structures throughout Orf1ab and reveals aspects of SARS-CoV-2 genome architecture that distinguish it from other RNA viruses. Evolutionary analysis shows that several features of the SARS-CoV-2 genomic structure are conserved across ß-coronaviruses, and we pinpoint regions of well-folded RNA structure that merit downstream functional analysis. The native, secondary structure of SARS-CoV-2 presented here is a roadmap that will facilitate focused studies on the viral life cycle, facilitate primer design, and guide the identification of RNA drug targets against COVID-19.


Asunto(s)
COVID-19 , Genoma Viral , Conformación de Ácido Nucleico , ARN Viral , Elementos de Respuesta , SARS-CoV-2 , COVID-19/genética , COVID-19/metabolismo , Línea Celular Tumoral , Humanos , ARN Viral/genética , ARN Viral/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/metabolismo
8.
Mol Cell ; 81(6): 1187-1199.e5, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33581076

RESUMEN

Type I interferons (IFNs) are critical cytokines in the host defense against invading pathogens. Sustained production of IFNs, however, is detrimental to the host, as it provokes autoimmune diseases. Thus, the expression of IFNs is tightly controlled. We report that the mRNA 5' cap-binding protein 4EHP plays a key role in regulating type I IFN concomitant with controlling virus replication, both in vitro and in vivo. Mechanistically, 4EHP suppresses IFN-ß production by effecting the miR-34a-induced translational silencing of Ifnb1 mRNA. miR-34a is upregulated by both RNA virus infection and IFN-ß induction, prompting a negative feedback regulatory mechanism that represses IFN-ß expression via 4EHP. These findings demonstrate the direct involvement of 4EHP in virus-induced host response, underscoring a critical translational silencing mechanism mediated by 4EHP and miR-34a to impede sustained IFN production. This study highlights an intrinsic regulatory function for miRNA and the translation machinery in maintaining host homeostasis.


Asunto(s)
Factor 4E Eucariótico de Iniciación/inmunología , Inmunidad Innata , MicroARNs/inmunología , Biosíntesis de Proteínas/inmunología , Infecciones por Virus ARN/inmunología , Virus ARN/inmunología , Animales , Factor 4E Eucariótico de Iniciación/genética , Células HEK293 , Humanos , Interferón beta/genética , Interferón beta/inmunología , Ratones , Ratones Transgénicos , MicroARNs/genética , Infecciones por Virus ARN/genética , Virus ARN/genética
9.
Mol Cell ; 78(4): 624-640.e7, 2020 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-32380061

RESUMEN

The primary interactions between incoming viral RNA genomes and host proteins are crucial to infection and immunity. Until now, the ability to study these events was lacking. We developed viral cross-linking and solid-phase purification (VIR-CLASP) to characterize the earliest interactions between viral RNA and cellular proteins. We investigated the infection of human cells using Chikungunya virus (CHIKV) and influenza A virus and identified hundreds of direct RNA-protein interactions. Here, we explore the biological impact of three protein classes that bind CHIKV RNA within minutes of infection. We find CHIKV RNA binds and hijacks the lipid-modifying enzyme fatty acid synthase (FASN) for pro-viral activity. We show that CHIKV genomes are N6-methyladenosine modified, and YTHDF1 binds and suppresses CHIKV replication. Finally, we find that the innate immune DNA sensor IFI16 associates with CHIKV RNA, reducing viral replication and maturation. Our findings have direct applicability to the investigation of potentially all RNA viruses.


Asunto(s)
Fiebre Chikungunya/virología , Virus Chikungunya/fisiología , Acido Graso Sintasa Tipo I/metabolismo , Genoma Viral , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Replicación Viral , Animales , Fiebre Chikungunya/genética , Fiebre Chikungunya/metabolismo , Chlorocebus aethiops , Acido Graso Sintasa Tipo I/genética , Células HEK293 , Humanos , Proteínas Nucleares/genética , Fosfoproteínas/genética , ARN Viral/genética , Proteínas de Unión al ARN/genética , Células Vero
10.
Trends Genet ; 40(8): 681-693, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38724328

RESUMEN

Positive-strand RNA [(+)RNA] viruses include pandemic SARS-CoV-2, tumor-inducing hepatitis C virus, debilitating chikungunya virus (CHIKV), lethal encephalitis viruses, and many other major pathogens. (+)RNA viruses replicate their RNA genomes in virus-induced replication organelles (ROs) that also evolve new viral species and variants by recombination and mutation and are crucial virus control targets. Recent cryo-electron microscopy (cryo-EM) reveals that viral RNA replication proteins form striking ringed 'crowns' at RO vesicle junctions with the cytosol. These crowns direct RO vesicle formation, viral (-)RNA and (+)RNA synthesis and capping, innate immune escape, and transfer of progeny (+)RNA genomes into translation and encapsidation. Ongoing studies are illuminating crown assembly, sequential functions, host factor interactions, etc., with significant implications for control and beneficial uses of viruses.


Asunto(s)
Genoma Viral , Orgánulos , ARN Viral , Replicación Viral , Replicación Viral/genética , Humanos , Genoma Viral/genética , Orgánulos/virología , Orgánulos/genética , Orgánulos/ultraestructura , ARN Viral/genética , Virus ARN Monocatenarios Positivos/genética , Microscopía por Crioelectrón , SARS-CoV-2/genética , SARS-CoV-2/fisiología , Ensamble de Virus/genética , Compartimentos de Replicación Viral , Animales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA