Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Virol ; 97(12): e0092823, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-38047713

RESUMEN

IMPORTANCE: Most protease-targeted antiviral development evaluates the ability of small molecules to inhibit the cleavage of artificial substrates. However, before they can cleave any other substrates, viral proteases need to cleave themselves out of the viral polyprotein in which they have been translated. This can occur either intra- or inter-molecularly. Whether this process occurs intra- or inter-molecularly has implications for the potential for precursors to accumulate and for the effectiveness of antiviral drugs. We argue that evaluating candidate antivirals for their ability to block these cleavages is vital to drug development because the buildup of uncleaved precursors can be inhibitory to the virus and potentially suppress the selection of drug-resistant variants.


Asunto(s)
Antivirales , Enterovirus , Inhibidores de Proteasa Viral , Proteasas Virales , Antivirales/farmacología , Antivirales/química , Proteolisis , Proteasas Virales/metabolismo , Inhibidores de Proteasa Viral/farmacología , Enterovirus/efectos de los fármacos , Enterovirus/fisiología , Poliproteínas/metabolismo
2.
J Infect Dis ; 228(Suppl 6): S427-S445, 2023 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-37849401

RESUMEN

Picornaviruses are nonenveloped particles with a single-stranded RNA genome of positive polarity. This virus family includes poliovirus, hepatitis A virus, rhinoviruses, and Coxsackieviruses. Picornaviruses are common human pathogens, and infection can result in a spectrum of serious illnesses, including acute flaccid myelitis, severe respiratory complications, and hand-foot-mouth disease. Despite research on poliovirus establishing many fundamental principles of RNA virus biology and the first transgenic animal model of disease for infection by a human virus, picornaviruses are understudied. Existing knowledge gaps include, identification of molecules required for virus entry, understanding cellular and humoral immune responses elicited during virus infection, and establishment of immune-competent animal models of virus pathogenesis. Such knowledge is necessary for development of pan-picornavirus countermeasures. Defining enterovirus A71 and D68, human rhinovirus C, and echoviruses 29 as prototype pathogens of this virus family may provide insight into picornavirus biology needed to establish public health strategies necessary for pandemic preparedness.


Asunto(s)
Infecciones por Enterovirus , Picornaviridae , Poliovirus , Animales , Humanos , Picornaviridae/genética , Poliovirus/fisiología , Rhinovirus , Enterovirus Humano B/fisiología
3.
Cell Rep ; 42(4): 112389, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37058406

RESUMEN

Enterovirus A71 (EV-A71) causes hand, foot, and mouth disease outbreaks with neurological complications and deaths. We previously isolated an EV-A71 variant in the stool, cerebrospinal fluid, and blood of an immunocompromised patient who had a leucine-to-arginine substitution on the VP1 capsid protein, resulting in increased heparin sulfate binding. We show here that this mutation increases the virus's pathogenicity in orally infected mice with depleted B cells, which mimics the patient's immune status, and increases susceptibility to neutralizing antibodies. However, a double mutant with even greater heparin sulfate affinity is not pathogenic, suggesting that increased heparin sulfate affinity may trap virions in peripheral tissues and reduce neurovirulence. This research sheds light on the increased pathogenicity of variant with heparin sulfate (HS)-binding ability in individuals with decreased B cell immunity.


Asunto(s)
Enterovirus Humano A , Infecciones por Enterovirus , Enterovirus , Humanos , Animales , Ratones , Enterovirus/genética , Enterovirus Humano A/genética , Antígenos Virales/metabolismo , Heparitina Sulfato/metabolismo , Heparina/metabolismo
4.
Nat Microbiol ; 8(4): 629-639, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36914754

RESUMEN

Enterovirus A71 causes severe disease upon systemic infection, sometimes leading to life-threatening neurological dysfunction. However, in most cases infection is asymptomatic and limited to the gastrointestinal tract, where virus is amplified for transmission. Picornaviruses have previously been shown to exit infected cells via either cell lysis or secretion of vesicles. Here we report that entire Enterovirus A71-infected cells are specifically extruded from the apical surface of differentiated human colon organoids, as observed by confocal microscopy. Differential sensitivity to chemical and peptide inhibitors demonstrated that extrusion of virus-infected cells is dependent on force sensing via mechanosensitive ion channels rather than apoptotic cell death. When isolated and used as inoculum, intact virus-containing extruded cells can initiate new infections. In contrast, when mechanical force sensing is inhibited, large amounts of free virus are released. Thus, extrusion of live, virus-infected cells from intact epithelial tissue is likely to benefit both the integrity of host tissues and the protected spread of this faecal-oral pathogen within and between hosts.


Asunto(s)
Enterovirus Humano A , Infecciones por Enterovirus , Enterovirus , Humanos , Enterovirus Humano A/fisiología , Replicación Viral/fisiología , Antígenos Virales
5.
Cell ; 184(10): 2633-2648.e19, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33864768

RESUMEN

Long non-coding RNA (lncRNA) genes have well-established and important impacts on molecular and cellular functions. However, among the thousands of lncRNA genes, it is still a major challenge to identify the subset with disease or trait relevance. To systematically characterize these lncRNA genes, we used Genotype Tissue Expression (GTEx) project v8 genetic and multi-tissue transcriptomic data to profile the expression, genetic regulation, cellular contexts, and trait associations of 14,100 lncRNA genes across 49 tissues for 101 distinct complex genetic traits. Using these approaches, we identified 1,432 lncRNA gene-trait associations, 800 of which were not explained by stronger effects of neighboring protein-coding genes. This included associations between lncRNA quantitative trait loci and inflammatory bowel disease, type 1 and type 2 diabetes, and coronary artery disease, as well as rare variant associations to body mass index.


Asunto(s)
Enfermedad/genética , Herencia Multifactorial/genética , Población/genética , ARN Largo no Codificante/genética , Transcriptoma , Enfermedad de la Arteria Coronaria/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 2/genética , Perfilación de la Expresión Génica , Variación Genética , Humanos , Enfermedades Inflamatorias del Intestino/genética , Especificidad de Órganos/genética , Sitios de Carácter Cuantitativo
6.
mBio ; 11(6)2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33173007

RESUMEN

Affordable and effective antiviral therapies are needed worldwide, especially against agents such as dengue virus that are endemic in underserved regions. Many antiviral compounds have been studied in cultured cells but are unsuitable for clinical applications due to pharmacokinetic profiles, side effects, or inconsistent efficacy across dengue serotypes. Such tool compounds can, however, aid in identifying clinically useful treatments. Here, computational screening (Rapid Overlay of Chemical Structures) was used to identify entries in an in silico database of safe-in-human compounds (SWEETLEAD) that display high chemical similarities to known inhibitors of dengue virus. Inhibitors of the dengue proteinase NS2B/3, the dengue capsid, and the host autophagy pathway were used as query compounds. Three FDA-approved compounds that resemble the tool molecules structurally, cause little toxicity, and display strong antiviral activity in cultured cells were selected for further analysis. Pyrimethamine (50% inhibitory concentration [IC50] = 1.2 µM), like the dengue proteinase inhibitor ARDP0006 to which it shows structural similarity, inhibited intramolecular NS2B/3 cleavage. Lack of toxicity early in infection allowed testing in mice, in which pyrimethamine also reduced viral loads. Niclosamide (IC50 = 0.28 µM), like dengue core inhibitor ST-148, affected structural components of the virion and inhibited early processes during infection. Vandetanib (IC50 = 1.6 µM), like cellular autophagy inhibitor spautin-1, blocked viral exit from cells and could be shown to extend survival in vivo Thus, three FDA-approved compounds with promising utility for repurposing to treat dengue virus infections and their potential mechanisms were identified using computational tools and minimal phenotypic screening.IMPORTANCE No antiviral therapeutics are currently available for dengue virus infections. By computationally overlaying the three-dimensional (3D) chemical structures of compounds known to inhibit dengue virus over those of compounds known to be safe in humans, we identified three FDA-approved compounds that are attractive candidates for repurposing as antivirals. We identified targets for two previously identified antiviral compounds and revealed a previously unknown potential anti-dengue compound, vandetanib. This computational approach to analyze a highly curated library of structures has the benefits of speed and cost efficiency. It also leverages mechanistic work with query compounds used in biomedical research to provide strong hypotheses for the antiviral mechanisms of the safer hit compounds. This workflow to identify compounds with known safety profiles can be expanded to any biological activity for which a small-molecule query compound has been identified, potentially expediting the translation of basic research to clinical interventions.


Asunto(s)
Antivirales/farmacología , Virus del Dengue/efectos de los fármacos , Dengue/virología , Animales , Bases de Datos Farmacéuticas , Dengue/tratamiento farmacológico , Virus del Dengue/genética , Virus del Dengue/fisiología , Evaluación Preclínica de Medicamentos , Reposicionamiento de Medicamentos , Humanos , Ratones , Ratones Endogámicos C57BL , Carga Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
7.
PLoS Biol ; 18(9): e3000827, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32997652

RESUMEN

Matrix proteins are encoded by many enveloped viruses, including influenza viruses, herpes viruses, and coronaviruses. Underneath the viral envelope of influenza virus, matrix protein 1 (M1) forms an oligomeric layer critical for particle stability and pH-dependent RNA genome release. However, high-resolution structures of full-length monomeric M1 and the matrix layer have not been available, impeding antiviral targeting and understanding of the pH-dependent transitions involved in cell entry. Here, purification and extensive mutagenesis revealed protein-protein interfaces required for the formation of multilayered helical M1 oligomers similar to those observed in virions exposed to the low pH of cell entry. However, single-layered helical oligomers with biochemical and ultrastructural similarity to those found in infectious virions before cell entry were observed upon mutation of a single amino acid. The highly ordered structure of the single-layered oligomers and their likeness to the matrix layer of intact virions prompted structural analysis by cryo-electron microscopy (cryo-EM). The resulting 3.4-Å-resolution structure revealed the molecular details of M1 folding and its organization within the single-shelled matrix. The solution of the full-length M1 structure, the identification of critical assembly interfaces, and the development of M1 assembly assays with purified proteins are crucial advances for antiviral targeting of influenza viruses.


Asunto(s)
Imagenología Tridimensional , Proteínas de la Matriz Viral/química , Secuencia de Aminoácidos , Reactivos de Enlaces Cruzados/química , Concentración de Iones de Hidrógeno , Modelos Moleculares , Mutación/genética , Multimerización de Proteína , Estructura Secundaria de Proteína , Subunidades de Proteína/química , Recombinación Genética/genética , Proteínas de la Matriz Viral/genética , Virión/ultraestructura
8.
Sci Adv ; 6(5): eaax9318, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-32064341

RESUMEN

Viral infections kill millions of people and new antivirals are needed. Nontoxic drugs that irreversibly inhibit viruses (virucidal) are postulated to be ideal. Unfortunately, all virucidal molecules described to date are cytotoxic. We recently developed nontoxic, broad-spectrum virucidal gold nanoparticles. Here, we develop further the concept and describe cyclodextrins, modified with mercaptoundecane sulfonic acids, to mimic heparan sulfates and to provide the key nontoxic virucidal action. We show that the resulting macromolecules are broad-spectrum, biocompatible, and virucidal at micromolar concentrations in vitro against many viruses [including herpes simplex virus (HSV), respiratory syncytial virus (RSV), dengue virus, and Zika virus]. They are effective ex vivo against both laboratory and clinical strains of RSV and HSV-2 in respiratory and vaginal tissue culture models, respectively. Additionally, they are effective when administrated in mice before intravaginal HSV-2 inoculation. Lastly, they pass a mutation resistance test that the currently available anti-HSV drug (acyclovir) fails.


Asunto(s)
Ciclodextrinas/farmacología , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 2/efectos de los fármacos , Virosis/tratamiento farmacológico , Aciclovir/química , Aciclovir/farmacología , Animales , Antivirales/síntesis química , Antivirales/química , Antivirales/farmacología , Ciclodextrinas/síntesis química , Ciclodextrinas/química , Femenino , Oro/química , Heparitina Sulfato/química , Heparitina Sulfato/farmacología , Herpesvirus Humano 1/patogenicidad , Herpesvirus Humano 2/patogenicidad , Humanos , Nanopartículas del Metal/química , Ratones , Simplexvirus/efectos de los fármacos , Simplexvirus/patogenicidad , Virosis/virología , Virus Zika/efectos de los fármacos , Virus Zika/patogenicidad
9.
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
10.
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
11.
Bio Protoc ; 8(20)2018 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-30505886

RESUMEN

Due to the exceptionally high mutation rates of RNA-dependent RNA polymerases, infectious RNA viruses generate extensive sequence diversity, leading to some of the lowest barriers to the development of antiviral drug resistance in the microbial world. We have previously discovered that higher barriers to the development of drug resistance can be achieved through dominant suppression of drug-resistant viruses by their drug-susceptible parents. We have explored the existence of dominant drug targets in poliovirus, dengue virus and hepatitis C virus (HCV). The low replication capacity of HCV required the development of novel strategies for identifying cells co-infected with drug-susceptible and drug-resistant strains. To monitor co-infected cell populations, we generated codon-altered versions of the JFH1 strain of HCV. Then, we could differentiate the codon-altered and wild-type strains using a novel type of RNA fluorescent in situ hybridization (FISH) coupled with flow cytometry or confocal microscopy. Both of these techniques can be used in conjunction with standard antibody-protein detection methods. Here, we describe a detailed protocol for both RNA FISH flow cytometry and confocal microscopy.

12.
Proc Natl Acad Sci U S A ; 115(40): 10136-10141, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30228122

RESUMEN

Many positive-strand RNA viruses translate their genomes as single polyproteins that are processed by host and viral proteinases to generate all viral protein products. Among these is dengue virus, which encodes the serine proteinase NS2B/3 responsible for seven different cleavages in the polyprotein. NS2B/3 has been the subject of many directed screens to find chemical inhibitors, of which the compound ARDP0006 is among the most effective at inhibiting viral growth. We show that at least three cleavages in the dengue polyprotein are exclusively intramolecular. By definition, such a cis-acting defect cannot be rescued in trans This creates the possibility that a drug-susceptible or inhibited proteinase can be genetically dominant, inhibiting the outgrowth of drug-resistant virus via precursor accumulation. Indeed, an NS3-G459L variant that is incapable of cleavage at the internal NS3 junction dominantly inhibited negative-strand RNA synthesis of wild-type virus present in the same cell. This internal NS3 cleavage site is the junction most inhibited by ARDP0006, making it likely that the accumulation of toxic precursors, not inhibition of proteolytic activity per se, explains the antiviral efficacy of this compound in restraining viral growth. We argue that intramolecularly cleaving proteinases are promising drug targets for viruses that encode polyproteins. The most effective inhibitors will specifically target cleavage sites required for processing precursors that exert trans-dominant inhibition.


Asunto(s)
Virus del Dengue/metabolismo , Proteolisis , ARN Viral/biosíntesis , Serina Endopeptidasas/metabolismo , Proteínas no Estructurales Virales/metabolismo , Animales , Línea Celular , Cricetinae , Virus del Dengue/genética , ARN Viral/genética , Serina Endopeptidasas/genética , Proteínas no Estructurales Virales/genética
13.
Elife ; 72018 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-29589830

RESUMEN

Antiviral development is plagued by drug resistance and genetic barriers to resistance are needed. For HIV and hepatitis C virus (HCV), combination therapy has proved life-saving. The targets of direct-acting antivirals for HCV infection are NS3/4A protease, NS5A phosphoprotein and NS5B polymerase. Differential visualization of drug-resistant and -susceptible RNA genomes within cells revealed that resistant variants of NS3/4A protease and NS5A phosphoprotein are cis-dominant, ensuring their direct selection from complex environments. Confocal microscopy revealed that RNA replication complexes are genome-specific, rationalizing the non-interaction of wild-type and variant products. No HCV antivirals yet display the dominance of drug susceptibility shown for capsid proteins of other viruses. However, effective inhibitors of HCV polymerase exact such high fitness costs for drug resistance that stable genome selection is not observed. Barriers to drug resistance vary with target biochemistry and detailed analysis of these barriers should lead to the use of fewer drugs.


Asunto(s)
Antivirales/farmacología , Farmacorresistencia Viral , Aptitud Genética , Hepacivirus/efectos de los fármacos , Proteínas Mutantes/genética , Selección Genética , Proteínas no Estructurales Virales/genética , Hepacivirus/genética , Hepacivirus/crecimiento & desarrollo , Proteínas Mutantes/metabolismo , Proteínas no Estructurales Virales/metabolismo
14.
Autophagy ; 14(5): 898-912, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29465287

RESUMEN

Macroautophagy/autophagy is a conserved catabolic process that promotes survival during stress. Autophagic dysfunction is associated with pathologies such as cancer and neurodegenerative diseases. Thus, autophagy must be strictly modulated at multiple levels (transcriptional, post-transcriptional, translational and post-translational) to prevent deregulation. Relatively little is known about the post-transcriptional control of autophagy. Here we report that the exoribonuclease Xrn1/XRN1 functions as a negative autophagy factor in the yeast Saccharomyces cerevisiae and in mammalian cells. In yeast, chromosomal deletion of XRN1 enhances autophagy and the frequency of autophagosome formation. Loss of Xrn1 results in the upregulation of autophagy-related (ATG) transcripts under nutrient-replete conditions, and this effect is dependent on the ribonuclease activity of Xrn1. Xrn1 expression is regulated by the yeast transcription factor Ash1 in rich conditions. In mammalian cells, siRNA depletion of XRN1 enhances autophagy and the replication of 2 picornaviruses. This work provides insight into the role of the RNA decay factor Xrn1/XRN1 as a post-transcriptional regulator of autophagy.


Asunto(s)
Autofagia , Exorribonucleasas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/genética , Transcripción Genética , Autofagosomas/metabolismo , Autofagosomas/ultraestructura , Células HeLa , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/metabolismo , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/ultraestructura
15.
Proc Natl Acad Sci U S A ; 114(26): 6653-6655, 2017 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-28607085
16.
Curr Opin Virol ; 20: 106-111, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27764731

RESUMEN

If a freshly minted genome contains a mutation that confers drug resistance, will it be selected in the presence of the drug? Not necessarily. During viral infections, newly synthesized viral genomes occupy the same cells as parent and other progeny genomes. If the antiviral target is chosen so that the drug-resistant progeny's growth is dominantly inhibited by the drug-susceptible members of its intracellular family, its outgrowth can be suppressed. Precedent for 'dominant drug targeting' as a deliberate approach to suppress the outgrowth of inhibitor-resistant viruses has been established for envelope variants of vesicular stomatitis virus and for capsid variants of poliovirus and dengue virus. Small molecules that stabilize oligomeric assemblages are a promising means to an unfit family to destroy the effectiveness of a newborn drug-resistant relative due to the co-assembly of drug-susceptible and drug-resistant monomers.


Asunto(s)
Antivirales/farmacología , Virus del Dengue/fisiología , Farmacorresistencia Viral , Poliovirus/fisiología , Selección Genética , Vesiculovirus/fisiología , Replicación Viral , Virus del Dengue/efectos de los fármacos , Genética de Población , Humanos , Poliovirus/efectos de los fármacos , Vesiculovirus/efectos de los fármacos
17.
PLoS Genet ; 12(5): e1005986, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27149616

RESUMEN

Rapidly evolving viruses are a major threat to human health. Such viruses are often highly pathogenic (e.g., influenza virus, HIV, Ebola virus) and routinely circumvent therapeutic intervention through mutational escape. Error-prone genome replication generates heterogeneous viral populations that rapidly adapt to new selection pressures, leading to resistance that emerges with treatment. However, population heterogeneity bears a cost: when multiple viral variants replicate within a cell, they can potentially interfere with each other, lowering viral fitness. This genetic interference can be exploited for antiviral strategies, either by taking advantage of a virus's inherent genetic diversity or through generating de novo interference by engineering a competing genome. Here, we discuss two such antiviral strategies, dominant drug targeting and therapeutic interfering particles. Both strategies harness the power of genetic interference to surmount two particularly vexing obstacles-the evolution of drug resistance and targeting therapy to high-risk populations-both of which impede treatment in resource-poor settings.


Asunto(s)
Antivirales/uso terapéutico , Farmacorresistencia Viral/genética , Ebolavirus/genética , VIH-1/genética , Subtipo H1N1 del Virus de la Influenza A/genética , Ebolavirus/efectos de los fármacos , Ebolavirus/patogenicidad , Variación Genética , Genoma Viral , VIH-1/efectos de los fármacos , VIH-1/patogenicidad , Humanos , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Selección Genética/genética , Replicación Viral/genética
18.
PLoS Pathog ; 12(2): e1005428, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26863439

RESUMEN

Hepatitis C virus (HCV) is a positive-strand RNA virus of the Flaviviridae family and a major cause of liver disease worldwide. HCV replicates in the cytoplasm, and the synthesis of viral proteins induces extensive rearrangements of host cell membranes producing structures, collectively termed the membranous web (MW). The MW contains the sites of viral replication and assembly, and we have identified distinct membrane fractions derived from HCV-infected cells that contain replication and assembly complexes enriched for viral RNA and infectious virus, respectively. The complex membrane structure of the MW is thought to protect the viral genome limiting its interactions with cytoplasmic pattern recognition receptors (PRRs) and thereby preventing activation of cellular innate immune responses. Here we show that PRRs, including RIG-I and MDA5, and ribosomes are excluded from viral replication and assembly centers within the MW. Furthermore, we present evidence that components of the nuclear transport machinery regulate access of proteins to MW compartments. We show that the restricted assess of RIG-I to the MW can be overcome by the addition of a nuclear localization signal sequence, and that expression of a NLS-RIG-I construct leads to increased immune activation and the inhibition of viral replication.


Asunto(s)
Hepacivirus/fisiología , Replicación Viral/genética , Transporte Activo de Núcleo Celular , Línea Celular , Membrana Celular/virología , Humanos , Señales de Localización Nuclear/metabolismo , ARN Viral/aislamiento & purificación , Receptores de Reconocimiento de Patrones/inmunología , Proteínas Virales/genética
19.
mBio ; 6(6): e01960-15, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26670386

RESUMEN

UNLABELLED: Dengue virus is a major human pathogen responsible for 400 million infections yearly. As with other RNA viruses, daunting challenges to antiviral design exist due to the high error rates of RNA-dependent RNA synthesis. Indeed, treatment of dengue virus infection with a nucleoside analog resulted in the expected genetic selection of resistant viruses in tissue culture and in mice. However, when the function of the oligomeric core protein was inhibited, no detectable selection of drug resistance in tissue culture or in mice was detected, despite the presence of drug-resistant variants in the population. Suppressed selection of drug-resistant virus correlated with cooligomerization of the targeted drug-susceptible and drug-resistant core proteins. The concept of "dominant drug targets," in which inhibition of oligomeric viral assemblages leads to the formation of drug-susceptible chimeras, can therefore be used to prevent the outgrowth of drug resistance during dengue virus infection. IMPORTANCE: Drug resistance is a major hurdle in the development of effective antivirals, especially those directed at RNA viruses. We have found that one can use the concept of the genetic dominance of defective subunits to "turn cousins into enemies," i.e., to thwart the outgrowth of drug-resistant viral genomes as soon as they are generated. This requires deliberate targeting of larger assemblages, which would otherwise rarely be considered by antiviral researchers.


Asunto(s)
Virus del Dengue/efectos de los fármacos , Virus del Dengue/crecimiento & desarrollo , Farmacorresistencia Viral , Supresión Genética , Animales , Antivirales/administración & dosificación , Antivirales/farmacología , Ratones , Selección Genética
20.
Virology ; 479-480: 444-9, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25890822

RESUMEN

How do viruses spread from cell to cell? Enveloped viruses acquire their surrounding membranes by budding. If a newly enveloped virus has budded through the plasma membrane, it finds itself outside the cell immediately. If it has budded through the bounding membrane of an internal compartment such as the ER, the virus finds itself in the lumen, from which it can exit the cell via the conventional secretion pathway. Thus, although some enveloped viruses destroy the cells they infect, there is no topological need to do so. On the other hand, naked viruses such as poliovirus lack an external membrane. They are protein-nucleic acid complexes within the cytoplasm or nucleus of the infected cell, like a ribosome, a spliceosome or an aggregate of Huntingtin protein. The simplest way for such a particle to pass through the single lipid bilayer that separates it from the outside of the cell would be to violate the integrity of that bilayer. Thus, it is not surprising that the primary mode of exit for non-enveloped viruses is cell lysis. However, more complex exit strategies are possible, such as the creation of new compartments whose complex topologies allow the exit of cytoplasm and its contents without violating the integrity of the cell. Here we will discuss the non-lytic spread of poliovirus and recent observations of such compartments during viral infection with several different picornaviruses.


Asunto(s)
Poliovirus/fisiología , Liberación del Virus , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA