Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
PLoS Pathog ; 17(12): e1010091, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34914813

RESUMEN

There are strong incentives for human populations to develop antiviral systems. Similarly, genomes that encode antiviral systems have had strong selective advantages. Protein-guided immune systems, which have been well studied in mammals, are necessary for survival in our virus-laden environments. Small RNA-directed antiviral immune systems suppress invasion of cells by non-self genetic material via complementary base pairing with target sequences. These RNA silencing-dependent systems operate in diverse organisms. In mammals, there is strong evidence that microRNAs (miRNAs) regulate endogenous genes important for antiviral immunity, and emerging evidence that virus-derived nucleic acids can be directly targeted by small interfering RNAs (siRNAs), PIWI-interacting RNAs (piRNAs), and transfer RNAs (tRNAs) for protection in some contexts. In this review, we summarize current knowledge of the antiviral functions of each of these small RNA types and consider their conceptual and mechanistic overlap with innate and adaptive protein-guided immunity, including mammalian antiviral cytokines, as well as the prokaryotic RNA-guided immune system, CRISPR. In light of recent successes in delivery of RNA for antiviral purposes, most notably for vaccination, we discuss the potential for development of small noncoding RNA-directed antiviral therapeutics and prophylactics.


Asunto(s)
ARN Pequeño no Traducido/inmunología , Virus/inmunología , Animales , Humanos
2.
Int J Mol Sci ; 22(16)2021 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-34445801

RESUMEN

The cytoplasmic retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs) initiate interferon (IFN) production and antiviral gene expression in response to RNA virus infection. Consequently, RLR signalling is tightly regulated by both host and viral factors. Tripartite motif protein 25 (TRIM25) is an E3 ligase that ubiquitinates multiple substrates within the RLR signalling cascade, playing both ubiquitination-dependent and -independent roles in RIG-I-mediated IFN induction. However, additional regulatory roles are emerging. Here, we show a novel interaction between TRIM25 and another protein in the RLR pathway that is essential for type I IFN induction, DEAD-box helicase 3X (DDX3X). In vitro assays and knockdown studies reveal that TRIM25 ubiquitinates DDX3X at lysine 55 (K55) and that TRIM25 and DDX3X cooperatively enhance IFNB1 induction following RIG-I activation, but the latter is independent of TRIM25's catalytic activity. Furthermore, we found that the influenza A virus non-structural protein 1 (NS1) disrupts the TRIM25:DDX3X interaction, abrogating both TRIM25-mediated ubiquitination of DDX3X and cooperative activation of the IFNB1 promoter. Thus, our results reveal a new interplay between two RLR-host proteins that cooperatively enhance IFN-ß production. We also uncover a new and further mechanism by which influenza A virus NS1 suppresses host antiviral defence.


Asunto(s)
Antivirales/inmunología , Proteína 58 DEAD Box/inmunología , ARN Helicasas DEAD-box/inmunología , Inmunidad/inmunología , Receptores Inmunológicos/inmunología , Factores de Transcripción/inmunología , Proteínas de Motivos Tripartitos/inmunología , Ubiquitina-Proteína Ligasas/inmunología , Línea Celular , Regulación de la Expresión Génica/inmunología , Células HEK293 , Humanos , Virus de la Influenza A/inmunología , Interferones/inmunología , Regiones Promotoras Genéticas/inmunología , Unión Proteica/inmunología , Transducción de Señal/inmunología , Ubiquitinación/inmunología
3.
J Virol ; 90(12): 5797-5807, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27076639

RESUMEN

UNLABELLED: The nucleolar subcompartment of the nucleus is increasingly recognized as an important target of RNA viruses. Here we document for the first time the ability of dengue virus (DENV) polymerase, nonstructural protein 5 (NS5), to accumulate within the nucleolus of infected cells and to target green fluorescent protein (GFP) to the nucleolus of live transfected cells. Intriguingly, NS5 exchange between the nucleus and nucleolus is dynamically modulated by extracellular pH, responding rapidly and reversibly to pH change, in contrast to GFP alone or other nucleolar and non-nucleolar targeted protein controls. The minimal pH-sensitive nucleolar targeting region (pHNTR), sufficient to target GFP to the nucleolus in a pH-sensitive fashion, was mapped to NS5 residues 1 to 244, with mutation of key hydrophobic residues, Leu-165, Leu-167, and Val-168, abolishing pHNTR function in NS5-transfected cells, and severely attenuating DENV growth in infected cells. This is the first report of a viral protein whose nucleolar targeting ability is rapidly modulated by extracellular stimuli, suggesting that DENV has the ability to detect and respond dynamically to the extracellular environment. IMPORTANCE: Infections by dengue virus (DENV) threaten 40% of the world's population yet there is no approved vaccine or antiviral therapeutic to treat infections. Understanding the molecular details that govern effective viral replication is key for the development of novel antiviral strategies. Here, we describe for the first time dynamic trafficking of DENV nonstructural protein 5 (NS5) to the subnuclear compartment, the nucleolus. We demonstrate that NS5's targeting to the nucleolus occurs in response to acidic pH, identify the key amino acid residues within NS5 that are responsible, and demonstrate that their mutation severely impairs production of infectious DENV. Overall, this study identifies a unique subcellular trafficking event and suggests that DENV is able to detect and respond dynamically to environmental changes.


Asunto(s)
Nucléolo Celular/metabolismo , Virus del Dengue/enzimología , Virus del Dengue/crecimiento & desarrollo , Espacio Extracelular/química , Proteínas no Estructurales Virales/metabolismo , Animales , Núcleo Celular/metabolismo , Chlorocebus aethiops , Virus del Dengue/química , Virus del Dengue/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Mutación , Transporte de Proteínas , Células Vero , Proteínas no Estructurales Virales/genética , Replicación Viral
4.
Biochem J ; 443(3): 851-6, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22417684

RESUMEN

The movement of proteins between the cytoplasm and nucleus mediated by the importin superfamily of proteins is essential to many cellular processes, including differentiation and development, and is critical to disease states such as viral disease and oncogenesis. We recently developed a high-throughput screen to identify specific and general inhibitors of protein nuclear import, from which ivermectin was identified as a potential inhibitor of importin α/ß-mediated transport. In the present study, we characterized in detail the nuclear transport inhibitory properties of ivermectin, demonstrating that it is a broad-spectrum inhibitor of importin α/ß nuclear import, with no effect on a range of other nuclear import pathways, including that mediated by importin ß1 alone. Importantly, we establish for the first time that ivermectin has potent antiviral activity towards both HIV-1 and dengue virus, both of which are strongly reliant on importin α/ß nuclear import, with respect to the HIV-1 integrase and NS5 (non-structural protein 5) polymerase proteins respectively. Ivermectin would appear to be an invaluable tool for the study of protein nuclear import, as well as the basis for future development of antiviral agents.


Asunto(s)
Virus del Dengue/efectos de los fármacos , VIH-1/efectos de los fármacos , Ivermectina/farmacología , Carioferinas/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos , Virus del Dengue/fisiología , VIH-1/fisiología , Células HeLa , Humanos , Carioferinas/fisiología
5.
Trends Microbiol ; 31(4): 393-404, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36463019

RESUMEN

Antiretroviral therapy (ART) reduces human immunodeficiency virus type 1 (HIV-1) infection, but selection of treatment-refractory variants remains a major challenge. HIV-1 encodes 16 canonical proteins, a small number of which are the singular targets of nearly all antiretrovirals developed to date. Cellular factors are increasingly being explored, which may present more therapeutic targets, more effectively target certain aspects of the viral replication cycle, and/or limit viral escape. Unlike most other positive-sense RNA viruses that encode at least one helicase, retroviruses are limited to the host repertoire. Accordingly, HIV-1 subverts DEAD-box helicase 3X (DDX3X) and numerous other cellular helicases of the Asp-Glu-x-Asp/His (DExD/H)-box family to service multiple aspects of its replication cycle. Here we review DDX3X and other DExD/H-box helicases in HIV-1 replication and their inhibition.


Asunto(s)
ARN Helicasas DEAD-box , Infecciones por VIH , VIH-1 , Humanos , Infecciones por VIH/tratamiento farmacológico , VIH-1/metabolismo , Replicación Viral/genética
6.
Nat Genet ; 55(6): 939-951, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37169872

RESUMEN

Mobile genetic elements (MEs) are heritable mutagens that recursively generate structural variants (SVs). ME variants (MEVs) are difficult to genotype and integrate in statistical genetics, obscuring their impact on genome diversification and traits. We developed a tool that accurately genotypes MEVs using short-read whole-genome sequencing (WGS) and applied it to global human populations. We find unexpected population-specific MEV differences, including an Alu insertion distribution distinguishing Japanese from other populations. Integrating MEVs with expression quantitative trait loci (eQTL) maps shows that MEV classes regulate tissue-specific gene expression by shared mechanisms, including creating or attenuating enhancers and recruiting post-transcriptional regulators, supporting class-wide interpretability. MEVs more often associate with gene expression changes than SNVs, thus plausibly impacting traits. Performing genome-wide association study (GWAS) with MEVs pinpoints potential causes of disease risk, including a LINE-1 insertion associated with keloid and fasciitis. This work implicates MEVs as drivers of human divergence and disease risk.


Asunto(s)
Estudio de Asociación del Genoma Completo , Polimorfismo de Nucleótido Simple , Humanos , Regulación de la Expresión Génica , Sitios de Carácter Cuantitativo , Fenotipo
7.
Cells ; 9(1)2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31936642

RESUMEN

Viral disease is one of the greatest burdens for human health worldwide, with an urgent need for efficacious antiviral strategies. While antiviral drugs are available, in many cases, they are prone to the development of drug resistance. A way to overcome drug resistance associated with common antiviral therapies is to develop antivirals targeting host cellular co-factors critical to viral replication, such as DEAD-box helicase 3 X-linked (DDX3X), which plays key roles in RNA metabolism and the antiviral response. Here, we use biochemical/biophysical approaches and infectious assays to show for the first time that the small molecule RK-33 has broad-spectrum antiviral action by inhibiting the enzymatic activities of DDX3X. Importantly, we show that RK-33 is efficacious at low micromolar concentrations in limiting infection by human parainfluenza virus type 3 (hPIV-3), respiratory syncytial virus (RSV), dengue virus (DENV), Zika virus (ZIKV) or West Nile virus (WNV)-for all of which, no Food and Drug Administration (FDA)-approved therapeutic is widely available. These findings establish for the first time that RK-33 is a broad-spectrum antiviral agent that blocks DDX3X's catalytic activities in vitro and limits viral replication in cells.


Asunto(s)
Antivirales/farmacología , Azepinas/farmacología , ARN Helicasas DEAD-box/antagonistas & inhibidores , Imidazoles/farmacología , Animales , Dominio Catalítico , Muerte Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , ARN Helicasas DEAD-box/metabolismo , Replicación Viral/efectos de los fármacos
8.
Clin Transl Immunology ; 8(7): e1067, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31312450

RESUMEN

Pathogen resistance and development costs are major challenges in current approaches to antiviral therapy. The high error rate of RNA synthesis and reverse-transcription confers genome plasticity, enabling the remarkable adaptability of RNA viruses to antiviral intervention. However, this property is coupled to fundamental constraints including limits on the size of information available to manipulate complex hosts into supporting viral replication. Accordingly, RNA viruses employ various means to extract maximum utility from their informationally limited genomes that, correspondingly, may be leveraged for effective host-oriented therapies. Host-oriented approaches are becoming increasingly feasible because of increased availability of bioactive compounds and recent advances in immunotherapy and precision medicine, particularly genome editing, targeted delivery methods and RNAi. In turn, one driving force behind these innovations is the increasingly detailed understanding of evolutionarily diverse host-virus interactions, which is the key concern of an emerging field, neo-virology. This review examines biotechnological solutions to disease and other sustainability issues of our time that leverage the properties of RNA and DNA viruses as developed through co-evolution with their hosts.

9.
Cells ; 8(10)2019 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-31575075

RESUMEN

DEAD-box helicase 3, X-linked (DDX3X) regulates the retinoic acid-inducible gene I (RIG-I)-like receptor (RLR)-mediated antiviral response, but can also be a host factor contributing to the replication of viruses of significance to human health, such as human immunodeficiency virus type 1 (HIV-1). These roles are mediated in part through its ability to actively shuttle between the nucleus and the cytoplasm to modulate gene expression, although the trafficking mechanisms, and impact thereof on immune signaling and viral infection, are incompletely defined. We confirm that DDX3X nuclear export is mediated by the nuclear transporter exportin-1/CRM1, dependent on an N-terminal, leucine-rich nuclear export signal (NES) and the monomeric guanine nucleotide binding protein Ran in activated GTP-bound form. Transcriptome profiling and ELISA show that exportin-1-dependent export of DDX3X to the cytoplasm strongly impacts IFN-ß production and the upregulation of immune genes in response to infection. That this is key to DDX3X's antiviral role was indicated by enhanced infection by human parainfluenza virus-3 (hPIV-3)/elevated virus production when the DDX3X NES was inactivated. Our results highlight a link between nucleocytoplasmic distribution of DDX3X and its role in antiviral immunity, with strong relevance to hPIV-3, as well as other viruses such as HIV-1.


Asunto(s)
Núcleo Celular/metabolismo , ARN Helicasas DEAD-box/metabolismo , Carioferinas/fisiología , Virus de la Parainfluenza 3 Humana/inmunología , Receptores Citoplasmáticos y Nucleares/fisiología , Infecciones por Respirovirus/inmunología , Células A549 , Transporte Activo de Núcleo Celular , Animales , Chlorocebus aethiops , Citoplasma/metabolismo , Células HEK293 , Células HeLa , Humanos , Carioferinas/genética , Receptores Citoplasmáticos y Nucleares/genética , Células Vero , Proteína Exportina 1
10.
Sci Rep ; 8(1): 358, 2018 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-29321677

RESUMEN

Hendra virus (HeV) is a paramyxovirus that causes lethal disease in humans, for which no vaccine or antiviral agent is available. HeV V protein is central to pathogenesis through its ability to interact with cytoplasmic host proteins, playing key antiviral roles. Here we use immunoprecipitation, siRNA knockdown and confocal laser scanning microscopy to show that HeV V shuttles to and from the nucleus through specific host nuclear transporters. Spectroscopic and small angle X-ray scattering studies reveal HeV V undergoes a disorder-to-order transition upon binding to either importin α/ß1 or exportin-1/Ran-GTP, dependent on the V N-terminus. Importantly, we show that specific inhibitors of nuclear transport prevent interaction with host transporters, and reduce HeV infection. These findings emphasize the critical role of host-virus interactions in HeV infection, and potential use of compounds targeting nuclear transport, such as the FDA-approved agent ivermectin, as anti-HeV agents.


Asunto(s)
Virus Hendra/fisiología , Infecciones por Henipavirus/metabolismo , Infecciones por Henipavirus/virología , Interacciones Huésped-Patógeno , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Antivirales/química , Antivirales/farmacología , Núcleo Celular/metabolismo , Descubrimiento de Drogas , Técnicas de Silenciamiento del Gen , Virus Hendra/efectos de los fármacos , Infecciones por Henipavirus/genética , Humanos , Carioferinas/química , Carioferinas/genética , Carioferinas/metabolismo , Modelos Moleculares , Conformación Molecular , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Relación Estructura-Actividad , Proteínas Virales/química , Proteínas Virales/metabolismo , Proteína Exportina 1
11.
J Exp Med ; 213(1): 1-13, 2016 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-26712804

RESUMEN

Viral infection activates danger signals that are transmitted via the retinoic acid-inducible gene 1-like receptor (RLR), nucleotide-binding oligomerization domain-like receptor (NLR), and Toll-like receptor (TLR) protein signaling cascades. This places host cells in an antiviral posture by up-regulating antiviral cytokines including type-I interferon (IFN-I). Ubiquitin modifications and cross-talk between proteins within these signaling cascades potentiate IFN-I expression, and inversely, a growing number of viruses are found to weaponize the ubiquitin modification system to suppress IFN-I. Here we review how host- and virus-directed ubiquitin modification of proteins in the RLR, NLR, and TLR antiviral signaling cascades modulate IFN-I expression.


Asunto(s)
Inmunidad Innata , Ubiquitina/metabolismo , Virosis/inmunología , Virosis/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Resistencia a la Enfermedad/inmunología , Interacciones Huésped-Patógeno , Humanos , Mitocondrias/metabolismo , Unión Proteica , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Ubiquitinación , Virosis/virología
12.
Clin Transl Immunology ; 9(2): e1115, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32099652
14.
Biomaterials ; 30(4): 682-9, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19000635

RESUMEN

Supported phospholipid bilayers are frequently used to establish a pseudo-physiological environment required for the study of protein function or the design of enzyme-based biosensors and biocatalytic reactors. These membranes are deposited from bilayer vesicles (liposomes) that rupture and fuse into a planar membrane upon adhesion to a surface. However, the morphology and homogeneity of the resulting layer is affected by the characteristics of the precursor liposome suspension and the substrate. Here we show that two distinct liposome populations contribute to membrane formation--equilibrium liposomes and small unilamellar vesicles. Liposome deposition onto carboxylic acid terminated self-assembled monolayers resulted in planar mono- and multilayer, vesicular and composite membranes, as a function of liposome size and composition. Quartz crystal microbalance data provided estimates for layer thicknesses and sheer moduli and were used for classification of the final structure. Finally, atomic force microscopy data illustrated the inherently inhomogeneous and dynamic nature of these membranes.


Asunto(s)
Ácidos Carboxílicos/química , Membrana Dobles de Lípidos/química , Fosfolípidos/química , 1,2-Dipalmitoilfosfatidilcolina/química , Liposomas/química , Microscopía de Fuerza Atómica , Modelos Químicos , Tamaño de la Partícula , Fosfatidilgliceroles/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA