Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell ; 184(12): 3109-3124.e22, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34004145

RESUMEN

Glycans modify lipids and proteins to mediate inter- and intramolecular interactions across all domains of life. RNA is not thought to be a major target of glycosylation. Here, we challenge this view with evidence that mammals use RNA as a third scaffold for glycosylation. Using a battery of chemical and biochemical approaches, we found that conserved small noncoding RNAs bear sialylated glycans. These "glycoRNAs" were present in multiple cell types and mammalian species, in cultured cells, and in vivo. GlycoRNA assembly depends on canonical N-glycan biosynthetic machinery and results in structures enriched in sialic acid and fucose. Analysis of living cells revealed that the majority of glycoRNAs were present on the cell surface and can interact with anti-dsRNA antibodies and members of the Siglec receptor family. Collectively, these findings suggest the existence of a direct interface between RNA biology and glycobiology, and an expanded role for RNA in extracellular biology.


Asunto(s)
Membrana Celular/metabolismo , Polisacáridos/metabolismo , ARN/metabolismo , Animales , Anticuerpos/metabolismo , Secuencia de Bases , Vías Biosintéticas , Línea Celular , Supervivencia Celular , Humanos , Espectrometría de Masas , Ácido N-Acetilneuramínico/metabolismo , Poliadenilación , Polisacáridos/química , ARN/química , ARN/genética , ARN no Traducido/metabolismo , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo , Coloración y Etiquetado
2.
Cell ; 159(5): 1086-1095, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25416947

RESUMEN

Fighting viral infections is hampered by the scarcity of viral targets and their variability, resulting in development of resistance. Viruses depend on cellular molecules-which are attractive alternative targets-for their life cycle, provided that they are dispensable for normal cell functions. Using the model organism Drosophila melanogaster, we identify the ribosomal protein RACK1 as a cellular factor required for infection by internal ribosome entry site (IRES)-containing viruses. We further show that RACK1 is an essential determinant for hepatitis C virus translation and infection, indicating that its function is conserved for distantly related human and fly viruses. Inhibition of RACK1 does not affect Drosophila or human cell viability and proliferation, and RACK1-silenced adult flies are viable, indicating that this protein is not essential for general translation. Our findings demonstrate a specific function for RACK1 in selective mRNA translation and uncover a target for the development of broad antiviral intervention.


Asunto(s)
Dicistroviridae/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/virología , Proteínas de Unión al GTP/metabolismo , Hepatocitos/virología , Virus de Insectos/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Línea Celular Tumoral , Drosophila melanogaster/metabolismo , Hepacivirus/metabolismo , Hepatocitos/metabolismo , Humanos , Modelos Moleculares , Factores de Iniciación de Péptidos/metabolismo , Biosíntesis de Proteínas , Receptores de Cinasa C Activada , Secuencias Reguladoras de Ácido Ribonucleico , Replicación Viral
3.
PLoS Pathog ; 20(3): e1012060, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38442126

RESUMEN

The recent discovery of Hepatitis D (HDV)-like viruses across a wide range of taxa led to the establishment of the Kolmioviridae family. Recent studies suggest that kolmiovirids can be satellites of viruses other than Hepatitis B virus (HBV), challenging the strict HBV/HDV-association dogma. Studying whether kolmiovirids are able to replicate in any animal cell they enter is essential to assess their zoonotic potential. Here, we compared replication of three kolmiovirids: HDV, rodent (RDeV) and snake (SDeV) deltavirus in vitro and in vivo. We show that SDeV has the narrowest and RDeV the broadest host cell range. High resolution imaging of cells persistently replicating these viruses revealed nuclear viral hubs with a peculiar RNA-protein organization. Finally, in vivo hydrodynamic delivery of viral replicons showed that both HDV and RDeV, but not SDeV, efficiently replicate in mouse liver, forming massive nuclear viral hubs. Our comparative analysis lays the foundation for the discovery of specific host factors controlling Kolmioviridae host-shifting.


Asunto(s)
Hepatitis D , Virus de la Hepatitis Delta , Ratones , Animales , Humanos , Roedores , Virus de la Hepatitis B/genética , Serpientes , Replicación Viral , ARN Viral/genética
4.
Semin Cell Dev Biol ; 111: 86-100, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32847707

RESUMEN

As obligate intracellular parasites with limited coding capacity, RNA viruses rely on host cells to complete their multiplication cycle. Viral RNAs (vRNAs) are central to infection. They carry all the necessary information for a virus to synthesize its proteins, replicate and spread and could also play essential non-coding roles. Regardless of its origin or tropism, vRNA has by definition evolved in the presence of host RNA Binding Proteins (RBPs), which resulted in intricate and complicated interactions with these factors. While on one hand some host RBPs recognize vRNA as non-self and mobilize host antiviral defenses, vRNA must also co-opt other host RBPs to promote viral infection. Focusing on pathogenic RNA viruses, we will review important scenarios of RBP-vRNA interactions during which host RBPs recognize, modify or degrade vRNAs. We will then focus on how vRNA hijacks the largest ribonucleoprotein complex (RNP) in the cell, the ribosome, to selectively promote the synthesis of its proteins. We will finally reflect on how novel technologies are helping in deepening our understanding of vRNA-host RBPs interactions, which can be ultimately leveraged to combat everlasting viral threats.


Asunto(s)
Virus ARN/genética , ARN Mensajero/genética , ARN Viral/genética , Proteínas de Unión al ARN/genética , Proteínas Virales/genética , Virosis/genética , Factores Eucarióticos de Iniciación/genética , Factores Eucarióticos de Iniciación/inmunología , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/genética , Unión Proteica , Biosíntesis de Proteínas , Virus ARN/crecimiento & desarrollo , Virus ARN/patogenicidad , ARN Mensajero/inmunología , ARN Viral/inmunología , Proteínas de Unión al ARN/inmunología , Ribosomas/genética , Ribosomas/metabolismo , Transducción de Señal , Proteínas Virales/metabolismo , Ensamble de Virus/genética , Virosis/inmunología , Virosis/patología , Virosis/virología
5.
PLoS Biol ; 17(1): e2006926, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30608919

RESUMEN

Many viruses interface with the autophagy pathway, a highly conserved process for recycling cellular components. For three viral infections in which autophagy constituents are proviral (poliovirus, dengue, and Zika), we developed a panel of knockouts (KOs) of autophagy-related genes to test which components of the canonical pathway are utilized. We discovered that each virus uses a distinct set of initiation components; however, all three viruses utilize autophagy-related gene 9 (ATG9), a lipid scavenging protein, and LC3 (light-chain 3), which is involved in membrane curvature. These results show that viruses use noncanonical routes for membrane sculpting and LC3 recruitment. By measuring viral RNA abundance, we also found that poliovirus utilizes these autophagy components for intracellular growth, while dengue and Zika virus only use autophagy components for post-RNA replication processes. Comparing how RNA viruses manipulate the autophagy pathway reveals new noncanonical autophagy routes, explains the exacerbation of disease by starvation, and uncovers common targets for antiviral drugs.


Asunto(s)
Autofagia/genética , Virus ARN/genética , Virus ARN/fisiología , Proteínas Relacionadas con la Autofagia/metabolismo , Línea Celular , Dengue/virología , Virus del Dengue/genética , Virus del Dengue/fisiología , Células HeLa , Humanos , Poliomielitis/virología , Poliovirus/genética , Poliovirus/fisiología , Virus ARN/metabolismo , ARN Viral , Virosis/genética , Replicación Viral , Virus Zika/genética , Virus Zika/fisiología , Infección por el Virus Zika/virología
6.
Nature ; 535(7610): 159-63, 2016 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-27383987

RESUMEN

The Flaviviridae are a family of viruses that cause severe human diseases. For example, dengue virus (DENV) is a rapidly emerging pathogen causing an estimated 100 million symptomatic infections annually worldwide. No approved antivirals are available to date and clinical trials with a tetravalent dengue vaccine showed disappointingly low protection rates. Hepatitis C virus (HCV) also remains a major medical problem, with 160 million chronically infected patients worldwide and only expensive treatments available. Despite distinct differences in their pathogenesis and modes of transmission, the two viruses share common replication strategies. A detailed understanding of the host functions that determine viral infection is lacking. Here we use a pooled CRISPR genetic screening strategy to comprehensively dissect host factors required for these two highly important Flaviviridae members. For DENV, we identified endoplasmic-reticulum (ER)-associated multi-protein complexes involved in signal sequence recognition, N-linked glycosylation and ER-associated degradation. DENV replication was nearly completely abrogated in cells deficient in the oligosaccharyltransferase (OST) complex. Mechanistic studies pinpointed viral RNA replication and not entry or translation as the crucial step requiring the OST complex. Moreover, we show that viral non-structural proteins bind to the OST complex. The identified ER-associated protein complexes were also important for infection by other mosquito-borne flaviviruses including Zika virus, an emerging pathogen causing severe birth defects. By contrast, the most significant genes identified in the HCV screen were distinct and included viral receptors, RNA-binding proteins and enzymes involved in metabolism. We found an unexpected link between intracellular flavin adenine dinucleotide (FAD) levels and HCV replication. This study shows notable divergence in host-depenency factors between DENV and HCV, and illuminates new host targets for antiviral therapy.


Asunto(s)
Sistemas CRISPR-Cas/genética , Virus del Dengue/fisiología , Genoma Humano/genética , Hepacivirus/fisiología , Factores Celulares Derivados del Huésped/genética , Interacciones Huésped-Patógeno/genética , Virus del Dengue/genética , Virus del Dengue/crecimiento & desarrollo , Descubrimiento de Drogas , Retículo Endoplásmico/metabolismo , Degradación Asociada con el Retículo Endoplásmico , Flavina-Adenina Dinucleótido/biosíntesis , Flavina-Adenina Dinucleótido/metabolismo , Infecciones por Flavivirus/genética , Infecciones por Flavivirus/virología , Glicosilación , Hexosiltransferasas/deficiencia , Hexosiltransferasas/genética , Hexosiltransferasas/metabolismo , Humanos , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Terapia Molecular Dirigida , Unión Proteica , Señales de Clasificación de Proteína , Proteínas de Unión al ARN/genética , Receptores Virales/genética , Proteínas no Estructurales Virales/metabolismo , Replicación Viral , Virus Zika/metabolismo
7.
Mol Ther ; 29(3): 1016-1027, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33678249

RESUMEN

Recombinant adeno-associated virus (rAAV) vectors have the unique property of being able to perform genomic targeted integration (TI) without inducing a double-strand break (DSB). In order to improve our understanding of the mechanism behind TI mediated by AAV and improve its efficiency, we performed an unbiased genetic screen in human cells using a promoterless AAV-homologous recombination (AAV-HR) vector system. We identified that the inhibition of the Fanconi anemia complementation group M (FANCM) protein enhanced AAV-HR-mediated TI efficiencies in different cultured human cells by ∼6- to 9-fold. The combined knockdown of the FANCM and two proteins also associated with the FANCM complex, RecQ-mediated genome instability 1 (RMI1) and Bloom DNA helicase (BLM) from the BLM-topoisomerase IIIα (TOP3A)-RMI (BTR) dissolvase complex (RMI1, having also been identified in our screen), led to the enhancement of AAV-HR-mediated TI up to ∼17 times. AAV-HR-mediated TI in the presence of a nuclease (CRISPR-Cas9) was also increased by ∼1.5- to 2-fold in FANCM and RMI1 knockout cells, respectively. Furthermore, knockdown of FANCM in human CD34+ hematopoietic stem and progenitor cells (HSPCs) increased AAV-HR-mediated TI by ∼3.5-fold. This study expands our knowledge on the mechanisms related to AAV-mediated TI, and it highlights new pathways that might be manipulated for future improvements in AAV-HR-mediated TI.


Asunto(s)
Sistemas CRISPR-Cas , ADN Helicasas/antagonistas & inhibidores , Proteínas de Unión al ADN/antagonistas & inhibidores , Dependovirus/genética , Edición Génica , Células Madre Hematopoyéticas/metabolismo , RecQ Helicasas/antagonistas & inhibidores , ADN Helicasas/genética , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Vectores Genéticos , Células HeLa , Células Madre Hematopoyéticas/citología , Recombinación Homóloga , Humanos , RecQ Helicasas/genética , RecQ Helicasas/metabolismo
8.
Nat Methods ; 15(3): 207-212, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29400715

RESUMEN

RNA-protein interactions play numerous roles in cellular function and disease. Here we describe RNA-protein interaction detection (RaPID), which uses proximity-dependent protein labeling, based on the BirA* biotin ligase, to rapidly identify the proteins that bind RNA sequences of interest in living cells. RaPID displays utility in multiple applications, including in evaluating protein binding to mutant RNA motifs in human genetic disorders, in uncovering potential post-transcriptional networks in breast cancer, and in discovering essential host proteins that interact with Zika virus RNA. To improve the BirA*-labeling component of RaPID, moreover, a new mutant BirA* was engineered from Bacillus subtilis, termed BASU, that enables >1,000-fold faster kinetics and >30-fold increased signal-to-noise ratio over the prior standard Escherichia coli BirA*, thereby enabling direct study of RNA-protein interactions in living cells on a timescale as short as 1 min.


Asunto(s)
Biotina/química , Proteínas de Unión al ARN/metabolismo , ARN/metabolismo , Proteínas Virales/metabolismo , Virus Zika/metabolismo , Bacillus subtilis/metabolismo , Ligasas de Carbono-Nitrógeno/genética , Ligasas de Carbono-Nitrógeno/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Células HEK293 , Humanos , Neuronas/citología , Neuronas/metabolismo , ARN/química , ARN/genética , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas Virales/química , Proteínas Virales/genética , Virus Zika/genética
9.
PLoS Pathog ; 15(5): e1007467, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31075158

RESUMEN

Hepatitis C virus (HCV) depends on liver-specific microRNA miR-122 for efficient viral RNA amplification in liver cells. This microRNA interacts with two different conserved sites at the very 5' end of the viral RNA, enhancing miR-122 stability and promoting replication of the viral RNA. Treatment of HCV patients with oligonucleotides that sequester miR-122 resulted in profound loss of viral RNA in phase II clinical trials. However, some patients accumulated in their sera a viral RNA genome that contained a single cytidine to uridine mutation at the third nucleotide from the 5' genomic end. It is shown here that this C3U variant indeed displayed higher rates of replication than that of wild-type HCV when miR-122 abundance is low in liver cells. However, when miR-122 abundance is high, binding of miR-122 to site 1, most proximal to the 5' end in the C3U variant RNA, is impaired without disrupting the binding of miR-122 to site 2. As a result, C3U RNA displays a much lower rate of replication than wild-type mRNA when miR-122 abundance is high in the liver. This phenotype was accompanied by binding of a different set of cellular proteins to the 5' end of the C3U RNA genome. In particular, binding of RNA helicase DDX6 was important for displaying the C3U RNA replication phenotype in liver cells. These findings suggest that sequestration of miR-122 leads to a resistance-associated mutation that has only been observed in treated patients so far, and raises the question about the function of the C3U variant in the peripheral blood.


Asunto(s)
Nucleótidos de Citosina/genética , Genoma Viral , Hepacivirus/genética , Hepatitis C/virología , MicroARNs/metabolismo , Mutación , ARN Viral/genética , Sitios de Unión , Hepatitis C/genética , Hepatitis C/metabolismo , Interacciones Huésped-Patógeno , Humanos , MicroARNs/genética , Replicación Viral
10.
Nucleic Acids Res ; 46(2): e8, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29136179

RESUMEN

Human translation initiation relies on the combined activities of numerous ribosome-associated eukaryotic initiation factors (eIFs). The largest factor, eIF3, is an ∼800 kDa multiprotein complex that orchestrates a network of interactions with the small 40S ribosomal subunit, other eIFs, and mRNA, while participating in nearly every step of initiation. How these interactions take place during the time course of translation initiation remains unclear. Here, we describe a method for the expression and affinity purification of a fluorescently-tagged eIF3 from human cells. The tagged eIF3 dodecamer is structurally intact, functions in cell-based assays, and interacts with the HCV IRES mRNA and the 40S-IRES complex in vitro. By tracking the binding of single eIF3 molecules to the HCV IRES RNA with a zero-mode waveguides-based instrument, we show that eIF3 samples both wild-type IRES and an IRES that lacks the eIF3-binding region, and that the high-affinity eIF3-IRES interaction is largely determined by slow dissociation kinetics. The application of single-molecule methods to more complex systems involving eIF3 may unveil dynamics underlying mRNA selection and ribosome loading during human translation initiation.


Asunto(s)
Factor 3 de Iniciación Eucariótica/metabolismo , Colorantes Fluorescentes/química , Imagen Individual de Molécula/métodos , Análisis Espectral/métodos , Factor 3 de Iniciación Eucariótica/química , Factor 3 de Iniciación Eucariótica/genética , Hepacivirus/genética , Humanos , Sitios Internos de Entrada al Ribosoma/genética , Biosíntesis de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Reproducibilidad de los Resultados , Subunidades Ribosómicas Pequeñas de Eucariotas/genética , Subunidades Ribosómicas Pequeñas de Eucariotas/metabolismo
11.
Gastroenterology ; 157(5): 1431-1432, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31586567
12.
EMBO Mol Med ; 16(4): 1004-1026, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38472366

RESUMEN

Viral neuroinfections represent a major health burden for which the development of antivirals is needed. Antiviral compounds that target the consequences of a brain infection (symptomatic treatment) rather than the cause (direct-acting antivirals) constitute a promising mitigation strategy that requires to be investigated in relevant models. However, physiological surrogates mimicking an adult human cortex are lacking, limiting our understanding of the mechanisms associated with viro-induced neurological disorders. Here, we optimized the Organotypic culture of Post-mortem Adult human cortical Brain explants (OPAB) as a preclinical platform for Artificial Intelligence (AI)-driven antiviral studies. OPAB shows robust viability over weeks, well-preserved 3D cytoarchitecture, viral permissiveness, and spontaneous local field potential (LFP). Using LFP as a surrogate for neurohealth, we developed a machine learning framework to predict with high confidence the infection status of OPAB. As a proof-of-concept, we showed that antiviral-treated OPAB could partially restore LFP-based electrical activity of infected OPAB in a donor-dependent manner. Together, we propose OPAB as a physiologically relevant and versatile model to study neuroinfections and beyond, providing a platform for preclinical drug discovery.


Asunto(s)
Antivirales , Hepatitis C Crónica , Humanos , Antivirales/farmacología , Inteligencia Artificial , Sistemas Microfisiológicos , Encéfalo
13.
Antiviral Res ; 209: 105461, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36396025

RESUMEN

Hepatitis delta virus (HDV), a satellite of hepatitis B virus (HBV), possesses the smallest viral genome known to infect animals. HDV needs HBV surface protein for secretion and entry into target liver cells. However, HBV is dispensable for HDV genome amplification, as it relies almost exclusively on cellular host factors for replication. HBV/HDV co-infections affect over 12 million people worldwide and constitute the most severe form of viral hepatitis. Co-infected individuals are at higher risk of developing liver cirrhosis and hepatocellular carcinoma compared to HBV mono-infected patients. Bulevirtide, an entry inhibitor, was conditionally approved in July 2020 in the European Union for adult patients with chronic hepatitis delta (CHD) and compensated liver disease. There are several drugs in development, including lonafarnib and interferon lambda, with different modes of action. In this review, we detail our current fundamental knowledge of HDV lifecycle and review antiviral treatments under development against this virus, outlining their respective mechanisms-of-action. Finally, we describe the antiviral effect these compounds are showing in ongoing clinical trials, discussing their promise and potential pitfalls for managing HDV infected patients.


Asunto(s)
Hepatitis B , Hepatitis D , Animales , Virus de la Hepatitis Delta , Hepatitis D/tratamiento farmacológico , Antivirales/farmacología , Antivirales/uso terapéutico , Virus de la Hepatitis B/genética
14.
Nat Commun ; 11(1): 2707, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32483149

RESUMEN

Chronic HBV infection is a major cause of liver disease and cancer worldwide. Approaches for cure are lacking, and the knowledge of virus-host interactions is still limited. Here, we perform a genome-wide gain-of-function screen using a poorly permissive hepatoma cell line to uncover host factors enhancing HBV infection. Validation studies in primary human hepatocytes identified CDKN2C as an important host factor for HBV replication. CDKN2C is overexpressed in highly permissive cells and HBV-infected patients. Mechanistic studies show a role for CDKN2C in inducing cell cycle G1 arrest through inhibition of CDK4/6 associated with the upregulation of HBV transcription enhancers. A correlation between CDKN2C expression and disease progression in HBV-infected patients suggests a role in HBV-induced liver disease. Taken together, we identify a previously undiscovered clinically relevant HBV host factor, allowing the development of improved infectious model systems for drug discovery and the study of the HBV life cycle.


Asunto(s)
Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Mutación con Ganancia de Función , Pruebas Genéticas/métodos , Estudio de Asociación del Genoma Completo/métodos , Hepatitis B/genética , Línea Celular Tumoral , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/metabolismo , Perfilación de la Expresión Génica/métodos , Células HEK293 , Células Hep G2 , Hepatitis B/metabolismo , Hepatitis B/virología , Virus de la Hepatitis B/fisiología , Interacciones Microbiota-Huesped , Humanos , Estimación de Kaplan-Meier , Hígado/metabolismo , Hígado/patología , Hígado/virología , Interferencia de ARN , Replicación Viral/fisiología
15.
Viruses ; 11(8)2019 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-31426357

RESUMEN

Animal cells have evolved dedicated molecular systems for sensing and delivering a coordinated response to viral threats. Our understanding of these pathways is almost entirely defined by studies in humans or model organisms like mice, fruit flies and worms. However, new genomic and functional data from organisms such as sponges, anemones and mollusks are helping redefine our understanding of these immune systems and their evolution. In this review, we will discuss our current knowledge of the innate immune pathways involved in sensing, signaling and inducing genes to counter viral infections in vertebrate animals. We will then focus on some central conserved players of this response including Toll-like receptors (TLRs), RIG-I-like receptors (RLRs) and cGAS-STING, attempting to put their evolution into perspective. To conclude, we will reflect on the arms race that exists between viruses and their animal hosts, illustrated by the dynamic evolution and diversification of innate immune pathways. These concepts are not only important to understand virus-host interactions in general but may also be relevant for the development of novel curative approaches against human disease.


Asunto(s)
Evolución Biológica , Inmunidad Innata , Virosis/inmunología , Animales , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/inmunología , Humanos , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Virosis/genética , Fenómenos Fisiológicos de los Virus , Virus/genética
16.
Nat Microbiol ; 4(12): 2369-2382, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31384002

RESUMEN

Flaviviruses, including dengue virus (DENV) and Zika virus (ZIKV), cause severe human disease. Co-opting cellular factors for viral translation and viral genome replication at the endoplasmic reticulum is a shared replication strategy, despite different clinical outcomes. Although the protein products of these viruses have been studied in depth, how the RNA genomes operate inside human cells is poorly understood. Using comprehensive identification of RNA-binding proteins by mass spectrometry (ChIRP-MS), we took an RNA-centric viewpoint of flaviviral infection and identified several hundred proteins associated with both DENV and ZIKV genomic RNA in human cells. Genome-scale knockout screens assigned putative functional relevance to the RNA-protein interactions observed by ChIRP-MS. The endoplasmic-reticulum-localized RNA-binding proteins vigilin and ribosome-binding protein 1 directly bound viral RNA and each acted at distinct stages in the life cycle of flaviviruses. Thus, this versatile strategy can elucidate features of human biology that control the pathogenesis of clinically relevant viruses.


Asunto(s)
Infecciones por Flavivirus/virología , Flavivirus/genética , Flavivirus/fisiología , ARN Viral/genética , Sistemas CRISPR-Cas , Proteínas Portadoras , Línea Celular , Virus del Dengue/genética , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Flavivirus/patogenicidad , Técnicas de Inactivación de Genes , Interacciones Huésped-Patógeno/genética , Humanos , ARN Viral/metabolismo , Proteínas de Unión al ARN/genética , Replicación Viral , Virus Zika/genética
17.
Nat Rev Microbiol ; 15(6): 351-364, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28420884

RESUMEN

Viruses depend on their hosts to complete their replication cycles; they exploit cellular receptors for entry and hijack cellular functions to replicate their genome, assemble progeny virions and spread. Recently, genome-scale CRISPR-Cas screens have been used to identify host factors that are required for virus replication, including the replication of clinically relevant viruses such as Zika virus, West Nile virus, dengue virus and hepatitis C virus. In this Review, we discuss the technical aspects of genome-scale knockout screens using CRISPR-Cas technology, and we compare these screens with alternative genetic screening technologies. The relative ease of use and reproducibility of CRISPR-Cas make it a powerful tool for probing virus-host interactions and for identifying new antiviral targets.


Asunto(s)
Sistemas CRISPR-Cas/genética , Virus del Dengue/genética , Hepacivirus/genética , Ensayos Analíticos de Alto Rendimiento/métodos , Virus del Nilo Occidental/genética , Virus Zika/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Técnicas de Inactivación de Genes , Humanos , Interferencia de ARN , Replicación Viral/genética
18.
G3 (Bethesda) ; 7(7): 2249-2258, 2017 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-28522639

RESUMEN

Receptor for Activated protein C kinase 1 (RACK1) is a scaffold protein that has been found in association with several signaling complexes, and with the 40S subunit of the ribosome. Using the model organism Drosophila melanogaster, we recently showed that RACK1 is required at the ribosome for internal ribosome entry site (IRES)-mediated translation of viruses. Here, we report a proteomic characterization of the interactome of RACK1 in Drosophila S2 cells. We carried out Label-Free quantitation using both Data-Dependent and Data-Independent Acquisition (DDA and DIA, respectively) and observed a significant advantage for the Sequential Window Acquisition of all THeoretical fragment-ion spectra (SWATH) method, both in terms of identification of interactants and quantification of low abundance proteins. These data represent the first SWATH spectral library available for Drosophila and will be a useful resource for the community. A total of 52 interacting proteins were identified, including several molecules involved in translation such as structural components of the ribosome, factors regulating translation initiation or elongation, and RNA binding proteins. Among these 52 proteins, 15 were identified as partners by the SWATH strategy only. Interestingly, these 15 proteins are significantly enriched for the functions translation and nucleic acid binding. This enrichment reflects the engagement of RACK1 at the ribosome and highlights the added value of SWATH analysis. A functional screen did not reveal any protein sharing the interesting properties of RACK1, which is required for IRES-dependent translation and not essential for cell viability. Intriguingly however, 10 of the RACK1 partners identified restrict replication of Cricket paralysis virus (CrPV), an IRES-containing virus.


Asunto(s)
Dicistroviridae , Proteínas de Drosophila , Redes Reguladoras de Genes , Sitios Internos de Entrada al Ribosoma , Modelos Genéticos , Receptores de Cinasa C Activada , Proteínas Virales , Animales , Línea Celular , Dicistroviridae/genética , Dicistroviridae/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Biosíntesis de Proteínas/genética , Receptores de Cinasa C Activada/genética , Receptores de Cinasa C Activada/metabolismo , Proteínas Virales/biosíntesis , Proteínas Virales/genética
19.
Cell Rep ; 21(11): 3032-3039, 2017 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-29241533

RESUMEN

The mosquito-borne flaviviruses include important human pathogens such as dengue, Zika, West Nile, and yellow fever viruses, which pose a serious threat for global health. Recent genetic screens identified endoplasmic reticulum (ER)-membrane multiprotein complexes, including the oligosaccharyltransferase (OST) complex, as critical flavivirus host factors. Here, we show that a chemical modulator of the OST complex termed NGI-1 has promising antiviral activity against flavivirus infections. We demonstrate that NGI-1 blocks viral RNA replication and that antiviral activity does not depend on inhibition of the N-glycosylation function of the OST. Viral mutants adapted to replicate in cells deficient of the OST complex showed resistance to NGI-1 treatment, reinforcing the on-target activity of NGI-1. Lastly, we show that NGI-1 also has strong antiviral activity in primary and disease-relevant cell types. This study provides an example for advancing from the identification of genetic determinants of infection to a host-directed antiviral compound with broad activity against flaviviruses.


Asunto(s)
Antivirales/farmacología , Benzamidas/farmacología , Virus del Dengue/efectos de los fármacos , Hexosiltransferasas/genética , Interacciones Huésped-Patógeno/efectos de los fármacos , Proteínas de la Membrana/genética , Sulfonamidas/farmacología , Replicación Viral/efectos de los fármacos , Virus del Dengue/genética , Virus del Dengue/crecimiento & desarrollo , Expresión Génica , Regulación de la Expresión Génica , Genes Reporteros , Células HEK293 , Hexosiltransferasas/antagonistas & inhibidores , Hexosiltransferasas/deficiencia , Humanos , Luciferasas , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/deficiencia , Pruebas de Sensibilidad Microbiana , Transducción de Señal , Virus del Nilo Occidental/efectos de los fármacos , Virus del Nilo Occidental/genética , Virus del Nilo Occidental/crecimiento & desarrollo , Virus de la Fiebre Amarilla/efectos de los fármacos , Virus de la Fiebre Amarilla/genética , Virus de la Fiebre Amarilla/crecimiento & desarrollo , Virus Zika/efectos de los fármacos , Virus Zika/genética , Virus Zika/crecimiento & desarrollo
20.
Cancer Res ; 69(18): 7459-65, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19738052

RESUMEN

MicroRNAs regulate diverse cellular processes and play an integral role in cancer pathogenesis. Genomic variation within miRNA target sites may therefore be important sources for genetic differences in cancer risk. To investigate this possibility, we mapped HapMap single nucleotide polymorphisms (SNP) to putative miRNA recognition sites within genes dysregulated in estrogen receptor-stratified breast tumors and used local linkage disequilibrium patterns to identify high-ranking SNPs in the Cancer Genetic Markers of Susceptibility (CGEMS) breast cancer genome-wide association study for further testing. Two SNPs, rs1970801 and rs11097457, scoring in the top 100 from the CGEMS study, were in strong linkage disequilibrium with rs1434536, an SNP that resides within a miR-125b target site in the 3' untranslated region of the bone morphogenic receptor type 1B (BMPR1B) gene encoding a transmembrane serine/threonine kinase. We validated the CGEMS association findings for rs1970801 in an independent cohort of admixture-corrected cases identified from families with multiple case histories. Subsequent association testing of rs1434536 for these cases and CGEMS controls with imputed genotypes supported the association. Furthermore, luciferase reporter assays and overexpression of miR-125b-mimics combined with quantitative reverse transcription-PCR showed that BMPR1B transcript is a direct target of miR-125b and that miR-125b differentially regulates the C and T alleles of rs1434536. These results suggest that allele-specific regulation of BMPR1B by miR-125b explains the observed disease risk. Our approach is general and can help identify and explain the mechanisms behind disease association for alleles that affect miRNA regulation.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Neoplasias de la Mama/genética , Transformación Celular Neoplásica/genética , MicroARNs/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Sitios de Unión , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/biosíntesis , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/metabolismo , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Receptores de Estrógenos/genética , Transfección , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA