Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
PLoS One ; 17(3): e0265453, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35333910

RESUMEN

Several SARS-CoV-2 variants emerged that harbor mutations in the surface unit of the viral spike (S) protein that enhance infectivity and transmissibility. Here, we analyzed whether ten naturally-occurring mutations found within the extended loop harboring the S1/S2 cleavage site of the S protein, a determinant of SARS-CoV-2 cell tropism and pathogenicity, impact S protein processing and function. None of the mutations increased but several decreased S protein cleavage at the S1/S2 site, including S686G and P681H, the latter of which is found in variants of concern B.1.1.7 (Alpha variant) and B.1.1.529 (Omicron variant). None of the mutations reduced ACE2 binding and cell-cell fusion although several modulated the efficiency of host cell entry. The effects of mutation S686G on viral entry were cell-type dependent and could be linked to the availability of cathepsin L for S protein activation. These results show that polymorphisms at the S1/S2 site can modulate S protein processing and host cell entry.


Asunto(s)
Polimorfismo Genético/genética , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética , Animales , Chlorocebus aethiops , Células HEK293/virología , Humanos , Immunoblotting , Células Vero/virología
2.
Gut Microbes ; 13(1): 1955643, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34369301

RESUMEN

Rotavirus is the most common cause of severe diarrhea among infants and young children and is responsible for more than 200,000 pediatric deaths per year. There is currently no pharmacological treatment for rotavirus infection in clinical activity. Although cholesterol synthesis has been proven to play a key role in the infections of multiple viruses, little is known about the relationship between cholesterol biosynthesis and rotavirus replication. The models of rotavirus infected two cell lines and a human small intestinal organoid were used. We investigated the effects of cholesterol biosynthesis, including inhibition, enhancement, and their combinations on rotavirus replication on these models. The knockdown of 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) was built by small hairpin RNAs in Caco2 cells. In all these models, inhibition of cholesterol synthesis by statins or HMGCR knockdown had a significant inhibitory effect on rotavirus replication. The result was further confirmed by the other inhibitors: 6-fluoromevalonate, Zaragozic acid A and U18666A, in the cholesterol biosynthesis pathway. Conversely, enhancement of cholesterol production increased rotavirus replication, suggesting that cholesterol homeostasis is relevant for rotavirus replication. The effects of all these compounds toward rotavirus were further confirmed with a clinical rotavirus isolate. We concluded that rotavirus replication is dependent on cholesterol biosynthesis. To be specific, inhibition of cholesterol synthesis can downregulate rotavirus replication; on the contrary, rotavirus replication is upregulated. Statin treatment is potentially an effective novel clinical anti-rotavirus strategy.


Asunto(s)
Colesterol/biosíntesis , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Infecciones por Rotavirus/tratamiento farmacológico , Rotavirus/efectos de los fármacos , Rotavirus/crecimiento & desarrollo , Replicación Viral/efectos de los fármacos , Animales , Anticolesterolemiantes/farmacocinética , Anticolesterolemiantes/uso terapéutico , Células CACO-2/efectos de los fármacos , Células CACO-2/virología , Células Cultivadas/efectos de los fármacos , Células Cultivadas/virología , Chlorocebus aethiops/crecimiento & desarrollo , Chlorocebus aethiops/virología , Modelos Animales de Enfermedad , Células HEK293/efectos de los fármacos , Células HEK293/virología , Humanos
3.
Adv Mater ; 33(37): e2103221, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34302401

RESUMEN

Liquid-phase electron microscopy (LP-EM) is an exciting new area in the materials imaging field, providing unprecedented views of molecular processes. Time-resolved insights from LP-EM studies are a strong complement to the remarkable results achievable with other high-resolution techniques. Here, the opportunities to expand LP-EM technology beyond 2D temporal assessments and into the 3D regime are described. The results show new structures and dynamic insights of human viruses contained in minute volumes of liquid while acquired in a rapid timeframe. To develop this strategy, adeno-associated virus (AAV) is used as a model system. AAV is a well-known gene therapy vehicle with current applications involving drug delivery and vaccine development for COVID-19. Improving the understanding of the physical properties of biological entities in a liquid state, as maintained in the human body, has broad societal implications for human health and disease.


Asunto(s)
Microscopía por Crioelectrón/métodos , Dependovirus , Tamaño de la Partícula , COVID-19 , Vacunas contra la COVID-19 , Sistemas de Liberación de Medicamentos , Diseño de Equipo , Terapia Genética , Células HEK293/virología , Humanos , Concentración de Iones de Hidrógeno , Inmunoglobulina G/química , Ensayo de Materiales , SARS-CoV-2
4.
Mol Biol (Mosk) ; 54(4): 634-642, 2020.
Artículo en Ruso | MEDLINE | ID: mdl-32799226

RESUMEN

A test of the sensitivity of seven colon cancer cell lines to a panel of 12 nonpathogenic human enteroviruses revealed significant differences in the ability of tumor cells to become infected and replicate different viral strains. Among the factors that can affect the sensitivity of cells to viruses are differences in the state of the mechanisms of antiviral protection, associated with a reaction to type I interferons. Using the two colon cancer cell lines CaCo2 and LIM1215 as a model, significant differences were revealed in the ability of cells to defend themselves against virus infection after 16 hours of treatment with 1000 units/mL of interferon-alpha. To study the effect of the state of the interferon response system, represented by the Jak/STAT signaling pathway, on the sensitivity of cells to different strains of enteroviruses, HEK293T cell lines were used. These are capable of supporting replication of each of the tested enteroviruses, as well as maintaining the ability to protect against viral infection after the treatment with interferon. Using the CRISPR/Cas9 system, HEK293T sublines with knockouts of the IFNAR1 and STAT2 genes were obtained. The sensitivity of control and knockout cells to infection with five strains of enteroviruses and the vesicular stomatitis virus was analyzed. It was noted that knockout of the IFNAR1 and STAT2 genes resulted in an increased sensitivity to all tested viruses. In knockout cells, the levels of reproduction of the vaccine derived of poliovirus type 1, Echoviruses 7 and 30, and Coxsackie viruses B5 and A7 were also significantly increased in comparison with the control HEK293T cells. Thus, deficiencies in the Jak/STAT signaling pathway in tumor cells lead to an overall increase in the sensitivity to oncolytic viruses.


Asunto(s)
Enterovirus , Células HEK293/virología , Virus Oncolíticos , Transducción de Señal , Células CACO-2 , Línea Celular Tumoral , Humanos , Viroterapia Oncolítica , Replicación Viral
5.
Viruses ; 12(7)2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32674313

RESUMEN

Enterovirus 71 (EV71) is the main pathogen causing hand-foot-mouth disease (HFMD) in infants and children, which can also lead to severe neurological diseases and even death. Therefore, understanding the replication mechanism of EV71 is of great significance for the prevention and control of EV71-induced diseases. Beclin1 (BECN1, a mammalian homologue of ATG6 in yeast) is an important core protein for the initiation and the normal process of autophagy in cells. In addition to its involvement in autophagy, Beclin1 has also been reported to play an important role in cancer and innate immune signaling pathways. However, the role of Beclin1 in EV71 replication remains elusive. Here, we primarily found that Beclin1 facilitates EV71 replication in human rhabdomyosarcoma (RD) cells and the autophagy was actually induced, but Beclin1 was not significantly affected at either mRNA level or protein level during early EV71 infection. Further studies discovered that Beclin1 could interacts with EV71 non-structural protein 3D mainly through its evolutionary conserved domain (ECD) and coiled-coiled domain (CCD), thus promoting the replication of EV71 in human rhabdomyosarcoma (RD) cells and human astroglioma (U251) cells. Collectively, we reveal a novel regulatory mechanism associated with Beclin1 to promote EV71 replication, thus providing a potential therapeutic target for the prevention and control of EV71-associated diseases.


Asunto(s)
Beclina-1/metabolismo , Enterovirus Humano A/fisiología , Proteínas Virales/metabolismo , Replicación Viral , Beclina-1/fisiología , Western Blotting , Línea Celular Tumoral/virología , Enterovirus Humano A/metabolismo , Infecciones por Enterovirus/metabolismo , Infecciones por Enterovirus/virología , Técnica del Anticuerpo Fluorescente , Células HEK293/virología , Humanos , Inmunoprecipitación , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Virales/fisiología
6.
Nat Struct Mol Biol ; 27(6): 570-580, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32424346

RESUMEN

The ESCRT complexes drive membrane scission in HIV-1 release, autophagosome closure, multivesicular body biogenesis, cytokinesis, and other cell processes. ESCRT-I is the most upstream complex and bridges the system to HIV-1 Gag in virus release. The crystal structure of the headpiece of human ESCRT-I comprising TSG101-VPS28-VPS37B-MVB12A was determined, revealing an ESCRT-I helical assembly with a 12-molecule repeat. Electron microscopy confirmed that ESCRT-I subcomplexes form helical filaments in solution. Mutation of VPS28 helical interface residues blocks filament formation in vitro and autophagosome closure and HIV-1 release in human cells. Coarse-grained (CG) simulations of ESCRT assembly at HIV-1 budding sites suggest that formation of a 12-membered ring of ESCRT-I molecules is a geometry-dependent checkpoint during late stages of Gag assembly and HIV-1 budding and templates ESCRT-III assembly for membrane scission. These data show that ESCRT-I is not merely a bridging adaptor; it has an essential scaffolding and mechanical role in its own right.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/química , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , VIH-1/fisiología , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencias de Aminoácidos , Autofagosomas , Membrana Celular/metabolismo , Cristalización , Cristalografía por Rayos X , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Células HEK293/virología , VIH-1/metabolismo , Interacciones Huésped-Patógeno/fisiología , Humanos , Simulación de Dinámica Molecular , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Conformación Proteica , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Liberación del Virus , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/química , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo
7.
Med Microbiol Immunol ; 209(4): 499-514, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32322956

RESUMEN

An estimated number of 71 million people are living with chronic hepatitis C virus (HCV) infection worldwide and 400,000 annual deaths are related to the infection. HCV entry into the hepatocytes is complex and involves several host factors. The tetraspanin human CD81 (hCD81) is one of the four essential entry factors and is composed of one large extracellular loop, one small extracellular loop, four transmembrane domains, one intracellular loop and two intracellular tails. The large extracellular loop interacts with the E2 glycoprotein of HCV. Regions outside the large extracellular loop (backbone) of hCD81 have a critical role in post-binding entry steps and determine susceptibility of hepatocytes to HCV. Here, we investigated the effect of five non-synonymous single-nucleotide variants in the backbone of hCD81 on HCV susceptibility. We generated cell lines that stably express the hCD81 variants and infected the cells using HCV pseudoparticles and cell culture-derived HCV. Our results show that all the tested hCD81 variants support HCV pseudoparticle entry with similar efficiency as wild-type hCD81. In contrast, variants A54V, V211M and M220I are less supportive to cell culture-derived HCV infection. This altered susceptibility is HCV genotype dependent and specifically affected the cell entry step. Our findings identify three hCD81 genetic variants that are impaired in their function as HCV host factors for specific viral genotypes. This study provides additional evidence that genetic host variation contributes to inter-individual differences in HCV infection and outcome.


Asunto(s)
Hepatitis C Crónica/metabolismo , Tetraspanina 28/genética , Tetraspanina 28/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Línea Celular Tumoral/virología , Células HEK293/virología , Hepatocitos/metabolismo , Hepatocitos/virología , Humanos , Mutación Puntual , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus
8.
J Biotechnol ; 304: 1-9, 2019 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-31404563

RESUMEN

Clustered regularly interspaced short palindromic repeat (CRISPR)-CRISPR associated proteins (Cas) 9 system is a powerful tool for genome editing and still being aggressively improved. Cas12a, a recently discovered Cas9 ortholog, is expected to become complementary to Cas9 due to its unique characteristics. Previously we attempted to establish an adenovirus (Ad) vector-mediated delivery of CRISPR-Cas12a system since Ad vector is widely used for gene transfer in basic researches and medical applications. However, we found difficulties preparing of Ad vectors at an adequate titer. In this study, we have developed Ad vectors that conditionally express Cas12a either by a tetracycline-controlled promoter or a hepatocyte specific promoter to avoid putative inhibitory effects of Cas12a. These vectors successfully proliferated in packaging cells, HEK293 cells, and were recovered at high titers. We have also developed packaging cells that express shRNA for Cas12a to suppress expression of Cas12a. Using the cells, the Ad vector directing constitutive expression of Cas12a proliferated efficiently and was successfully recovered at a high titer. Overall, we improved recovery of Ad vectors carrying CRISPR-Cas12a system, thus provided them as a tool in genome editing researches.


Asunto(s)
Adenoviridae/fisiología , Proteínas Asociadas a CRISPR/genética , ARN Guía de Kinetoplastida/genética , Adenoviridae/genética , Sistemas CRISPR-Cas , Proliferación Celular , Edición Génica , Vectores Genéticos/fisiología , Células HEK293/citología , Células HEK293/virología , Humanos , Carga Viral
9.
Antiviral Res ; 167: 1-5, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30951731

RESUMEN

The antiviral drug T-705 (favipiravir) and its non-fluorinated analogue T-1105 inhibit the polymerases of RNA viruses after being converted to their ribonucleoside triphosphate (RTP) metabolite. We here compared the activation efficiency of T-705 and T-1105 in four cell lines that are commonly used for their antiviral evaluation. In MDCK cells, the levels of T-705-RTP were markedly lower than those of T-1105-RTP, while the opposite was seen in A549, Vero and HEK293T cells. In the latter three cell lines, T-1105 activation was hindered by inefficient conversion of the ribonucleoside monophosphate to the ribonucleoside diphosphate en route to forming the active triphosphate. Accordingly, T-1105 had better anti-RNA virus activity in MDCK cells, while T-705 was more potent in the other three cell lines. Additionally, we identified a fourth metabolite, the NAD analogue of T-705/T-1105, and showed that it can be formed by nicotinamide mononucleotide adenylyltransferase.


Asunto(s)
Amidas/farmacología , Antivirales/farmacología , Línea Celular , Pirazinas/farmacología , Virus ARN/efectos de los fármacos , Animales , Línea Celular/efectos de los fármacos , Línea Celular/metabolismo , Línea Celular/virología , Chlorocebus aethiops , Perros , Células HEK293/efectos de los fármacos , Células HEK293/metabolismo , Células HEK293/virología , Humanos , Células de Riñón Canino Madin Darby/efectos de los fármacos , Células de Riñón Canino Madin Darby/metabolismo , Células de Riñón Canino Madin Darby/virología , Ribonucleósidos/metabolismo , Células Vero/efectos de los fármacos , Células Vero/metabolismo , Células Vero/virología
11.
Hereditas ; 156: 10, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30774581

RESUMEN

BACKGROUND: Influenza A virus (IAV) belongs to the Orthomyxoviridae family. IAV causes a highly contagious respiratory disease in humans that exacts severe economic losses globally. The virus uses strategies developed to exploit and subvert cellular proteins and pathways to increase its own replication and to inhibit antiviral immune response. RESULTS: A/bar-headed goose/Qinghai/1/2005 (A/QH) was able to infect A549 and 293 T cells, with a high infection rate for A549 cells. To identify host cellular responses of human cells to influenza infection, differentially expressed genes (DEGs) between AIV-infected groups and uninfected controls were identified using RNA-sequencing. The DEGs were annotated by Gene Ontology and the Kyoto Encyclopedia of Genes and Genomes pathway analyses, which revealed that the DEGs were mainly linked to cellular function and metabolic processes, while the cellular function that is probably associated with host cellular response of human cells, including defense response to virus and protein modification. All the DEGs and pathways were possibly involved in the response to IAV invasion. CONCLUSIONS: The global transcriptome analysis results revealed that sensitive genes and pathways of the cells were infected with the influenza virus and provided further evidence to investigate the complicated relationship between IAV and host cells.


Asunto(s)
Células A549/metabolismo , Células HEK293/metabolismo , Subtipo H5N1 del Virus de la Influenza A/fisiología , Transcriptoma , Replicación Viral , Células A549/virología , Perfilación de la Expresión Génica , Células HEK293/virología , Humanos , Análisis de Secuencia de ARN
12.
PLoS Pathog ; 15(1): e1007569, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30677091

RESUMEN

Human Cytomegalovirus (HCMV) infection induces several metabolic activities that are essential for viral replication. Despite the important role that this metabolic modulation plays during infection, the viral mechanisms involved are largely unclear. We find that the HCMV UL38 protein is responsible for many aspects of HCMV-mediated metabolic activation, with UL38 being necessary and sufficient to drive glycolytic activation and induce the catabolism of specific amino acids. UL38's metabolic reprogramming role is dependent on its interaction with TSC2, a tumor suppressor that inhibits mTOR signaling. Further, shRNA-mediated knockdown of TSC2 recapitulates the metabolic phenotypes associated with UL38 expression. Notably, we find that in many cases the metabolic flux activation associated with UL38 expression is largely independent of mTOR activity, as broad spectrum mTOR inhibition does not impact UL38-mediated induction of glycolysis, glutamine consumption, or the secretion of proline or alanine. In contrast, the induction of metabolite concentrations observed with UL38 expression are largely dependent on active mTOR. Collectively, our results indicate that the HCMV UL38 protein induces a pro-viral metabolic environment via inhibition of TSC2.


Asunto(s)
Proteínas de la Cápside/metabolismo , Citomegalovirus/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo , Proteínas de la Cápside/genética , Línea Celular , Citomegalovirus/genética , Infecciones por Citomegalovirus/virología , Fibroblastos/virología , Glucólisis , Células HEK293/virología , Humanos , ARN Interferente Pequeño/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Replicación Viral
13.
Biotechnol Bioeng ; 114(11): 2539-2549, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28710851

RESUMEN

Apoptosis has important functions during pathophysiologic processes. However, from a biopharmaceutical point of view, active apoptosis of host cells is undesirable during viral packaging or protein expression, because it decreases the efficiency of viral or protein production. Here we used the CRISPR/Cas technique to knock out four pro-apoptotic genes, Caspase3, Caspase6, Caspase7 and AIF1, in HEK293 cells, and successfully produced an apoptosis-resistant cell line. Furthermore, this cell line showed higher expression levels of pro-apoptotic proteins and higher packaging efficiency for the virus carrying these proteins than control HEK293 cells. This study not only produced an apoptosis-resistant cell line that is useful in producing apoptosis-inducing proteins or viruses expressing these proteins, but also provides a methodology to build other apoptosis-resistant cell lines.


Asunto(s)
Apoptosis/genética , Sistemas CRISPR-Cas/genética , Mejoramiento Genético/métodos , Células HEK293/fisiología , Células HEK293/virología , Lentivirus/crecimiento & desarrollo , Proteínas Recombinantes/biosíntesis , Técnicas de Inactivación de Genes/métodos , Células HEK293/citología , Humanos , Lentivirus/aislamiento & purificación , Ingeniería de Proteínas/métodos , Proteínas Recombinantes/aislamiento & purificación
14.
Vaccine ; 35(26): 3423-3430, 2017 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-28495315

RESUMEN

Despite major advances in developing capacities and alternative technologies to egg-based production of influenza vaccines, responsiveness to an influenza pandemic threat is limited by the time it takes to generate a Candidate Vaccine Virus (CVV) as reported by the 2015 WHO Informal Consultation report titled "Influenza Vaccine Response during the Start of a Pandemic". In previous work, we have shown that HEK-293 cell culture in suspension and serum free medium is an efficient production platform for cell culture manufacturing of influenza candidate vaccines. This report, took advantage of, recombinant DNA technology using Reverse Genetics of influenza strains, and advances in the large-scale transfection of suspension cultured HEK-293 cells. We demonstrate the efficient generation of H1N1 with the PR8 backbone reassortant under controlled bioreactor conditions in two sequential steps (transfection/rescue and infection/production). This approach could deliver a CVV for influenza vaccine manufacturing within two-weeks, starting from HA and NA pandemic sequences. Furthermore, the scalability of the transfection technology combined with the HEK-293 platform has been extensively demonstrated at >100L scale for several biologics, including recombinant viruses. Thus, this innovative approach is better suited to rationally engineer and mass produce influenza CVV within significantly shorter timelines to enable an effective global response in pandemic situations.


Asunto(s)
Células HEK293/virología , Subtipo H1N1 del Virus de la Influenza A/crecimiento & desarrollo , Genética Inversa , Cultivo de Virus , Reactores Biológicos , Pruebas de Inhibición de Hemaglutinación , Humanos , Vacunas contra la Influenza , Virus Reordenados/crecimiento & desarrollo , Transfección
15.
J Mol Biol ; 429(8): 1171-1191, 2017 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-28315663

RESUMEN

The retroviral restriction factors of the APOBEC3 (A3) cytidine deaminase family catalyze the deamination of cytidines in single-stranded viral DNA. APOBEC3C (A3C) is a strong antiviral factor against viral infectivity factor (vif)-deficient simian immunodeficiency virus Δvif, which is, however, a weak inhibitor against human immunodeficiency virus (HIV)-1 for reasons unknown. The precise link between the antiretroviral effect of A3C and its catalytic activity is incompletely understood. Here, we show that the S61P mutation in human A3C (A3C.S61P) boosted hypermutation in the viral genomes of simian immunodeficiency virus Δvif and murine leukemia virus but not in human immunodeficiency virus HIV-1Δvif. The enhanced antiviral activity of A3C.S61P correlated with enhanced in vitro cytidine deamination. Furthermore, the S61P mutation did not change the substrate specificity of A3C, ribonucleoprotein complex formation, self-association, Zinc coordination, or viral incorporation features. We propose that local structural changes induced by the serine-to-proline substitution are responsible for the gain of catalytic activity of A3C.S61P. Our results are a first step toward an understanding of A3C's DNA binding capacity, deamination-dependent editing, and antiviral functions at the molecular level. We conclude that the enhanced enzymatic activity of A3C is insufficient to restrict HIV-1, indicating an unknown escape mechanism of HIV-1.


Asunto(s)
Citidina Desaminasa/química , Citidina Desaminasa/metabolismo , VIH-1/patogenicidad , Sustitución de Aminoácidos , Animales , Citidina Desaminasa/genética , Citosina/metabolismo , ADN de Cadena Simple/química , ADN de Cadena Simple/metabolismo , ADN Viral/metabolismo , Células HEK293/virología , VIH-1/genética , Interacciones Huésped-Patógeno , Humanos , Virus de la Leucemia Murina/metabolismo , Virus de la Leucemia Murina/patogenicidad , Pan troglodytes , Conformación Proteica , Virus de la Inmunodeficiencia de los Simios/metabolismo , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Zinc/metabolismo , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo
16.
Proteomics ; 17(5)2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28067018

RESUMEN

Sendai virus (SeV) is an enveloped nonsegmented negative-strand RNA virus that belongs to the genus Respirovirus of the Paramyxoviridae family. As a model pathogen, SeV has been extensively studied to define the basic biochemical and molecular biologic properties of the paramyxoviruses. In addition, SeV-infected host cells were widely employed to uncover the mechanism of innate immune response. To identify proteins involved in the SeV infection process or the SeV-induced innate immune response process, system-wide evaluations of SeV-host interactions have been performed. cDNA microarray, siRNA screening and phosphoproteomic analysis suggested that multiple signaling pathways are involved in SeV infection process. Here, to study SeV-host interaction, a global quantitative proteomic analysis was performed on SeV-infected HEK 293T cells. A total of 4699 host proteins were quantified, with 742 proteins being differentially regulated. Bioinformatics analysis indicated that regulated proteins were mainly involved in "interferon type I (IFN-I) signaling pathway" and "defense response to virus," suggesting that these processes play roles in SeV infection. Further RNAi-based functional studies indicated that the regulated proteins, tripartite motif (TRIM24) and TRIM27, affect SeV-induced IFN-I production. Our data provided a comprehensive view of host cell response to SeV and identified host proteins involved in the SeV infection process or the SeV-induced innate immune response process.


Asunto(s)
Interacciones Huésped-Patógeno/fisiología , Proteoma/análisis , Infecciones por Respirovirus/metabolismo , Virus Sendai/patogenicidad , Citoplasma/química , Citoplasma/metabolismo , Citoplasma/virología , Células HEK293/virología , Humanos , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Proteínas Nucleares/análisis , Proteínas Nucleares/metabolismo , Reacción en Cadena de la Polimerasa/métodos , Proteoma/genética , Proteoma/metabolismo , Proteómica/métodos , Reproducibilidad de los Resultados , Infecciones por Respirovirus/virología , Factores de Transcripción/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Replicación Viral
17.
Cell Host Microbe ; 20(6): 798-809, 2016 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-27866901

RESUMEN

Chromosomal structure of nuclear DNA is usually maintained by insertion of nucleosomes into preexisting chromatin, both on newly synthesized DNA at replication forks and at sites of DNA damage. But during retrovirus infection, a histone-free DNA copy of the viral genome is synthesized that must be loaded with nucleosomes de novo. Here, we show that core histones are rapidly loaded onto unintegrated Moloney murine leukemia virus DNAs. Loading of nucleosomes requires nuclear entry, but does not require viral DNA integration. The histones associated with unintegrated DNAs become marked by covalent modifications, with a delay relative to the time of core histone loading. Expression from unintegrated DNA can be enhanced by modulation of the histone-modifying machinery. The data show that histone loading onto unintegrated DNAs occurs very rapidly after nuclear entry and does not require prior establishment of an integrated provirus.


Asunto(s)
ADN Viral , Código de Histonas , Histonas/metabolismo , Retroviridae/genética , Internalización del Virus , Animales , Ciclo Celular , Cromatina , Inmunoprecipitación de Cromatina , Daño del ADN , Replicación del ADN/genética , ADN Viral/análisis , Epigenómica , Células HEK293/virología , Humanos , Ratones , Virus de la Leucemia Murina de Moloney/genética , Células 3T3 NIH/virología , Nucleosomas/genética , Infecciones por Retroviridae/virología , Integración Viral
18.
Cell Host Microbe ; 20(6): 770-784, 2016 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-27866900

RESUMEN

RIG-I detects double-stranded RNA (dsRNA) to trigger antiviral cytokine production. Protein deamidation is emerging as a post-translational modification that chiefly regulates protein function. We report here that UL37 of herpes simplex virus 1 (HSV-1) is a protein deamidase that targets RIG-I to block RNA-induced activation. Mass spectrometry analysis identified two asparagine residues in the helicase 2i domain of RIG-I that were deamidated upon UL37 expression or HSV-1 infection. Deamidation rendered RIG-I unable to sense viral dsRNA, thus blocking its ability to trigger antiviral immune responses and restrict viral replication. Purified full-length UL37 and its carboxyl-terminal fragment were sufficient to deamidate RIG-I in vitro. Uncoupling RIG-I deamidation from HSV-1 infection, by engineering deamidation-resistant RIG-I or introducing deamidase-deficient UL37 into the HSV-1 genome, restored RIG-I activation and antiviral immune signaling. Our work identifies a viral deamidase and extends the paradigm of deamidation-mediated suppression of innate immunity by microbial pathogens.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , ADN Helicasas/metabolismo , Herpesvirus Humano 1/genética , Proteínas Virales/metabolismo , Adenosina Trifosfatasas , Adenosina Trifosfato/metabolismo , Antivirales/inmunología , Asparagina , Línea Celular/virología , Citocinas/metabolismo , Proteína 58 DEAD Box/efectos de los fármacos , ADN Recombinante , Escherichia coli/genética , Células HEK293/virología , Herpes Simple , Herpesvirus Humano 1/enzimología , Humanos , Evasión Inmune , Inmunidad Innata , Espectrometría de Masas , Mutación , Procesamiento Proteico-Postraduccional , ARN Bicatenario , ARN Viral/metabolismo , Transducción de Señal , Proteínas Estructurales Virales/análisis , Proteínas Estructurales Virales/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos
19.
FEBS Lett ; 590(16): 2797-810, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27423063

RESUMEN

NF90 is a novel host antiviral factor that regulates PKR activation and stress granule formation in influenza A virus (IAV)-infected cells, but the precise mechanisms by which it operates remain unclear. We identified NF90 as a novel interacting protein of IAV nonstructural protein 1 (NS1). The interaction was dependent on the RNA-binding properties of NS1. NS1 associated with NF90 and PKR simultaneously; however, the interaction between NF90 and PKR was restricted by NS1. Knockdown of NF90 promoted inhibition of PKR phosphorylation induced by NS1, while coexpression of NF90 impeded reduction of PKR phosphorylation and stress granule formation triggered by NS1. In summary, NF90 exerts its antiviral activity by antagonizing the inhibitory role of NS1 on PKR phosphorylation.


Asunto(s)
Virus de la Influenza A/metabolismo , Gripe Humana/virología , Proteínas del Factor Nuclear 90/metabolismo , Proteínas no Estructurales Virales/metabolismo , eIF-2 Quinasa/metabolismo , Células HEK293/virología , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/patogenicidad , Gripe Humana/genética , Gripe Humana/metabolismo , Proteínas del Factor Nuclear 90/genética , Fosforilación/genética , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Proteínas no Estructurales Virales/química , Replicación Viral/genética , eIF-2 Quinasa/química
20.
J Virol ; 90(14): 6502-14, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27147747

RESUMEN

UNLABELLED: Enveloped viruses utilize transmembrane surface glycoproteins to gain entry into target cells. Glycoproteins from diverse viral families can be incorporated into nonnative viral particles in a process termed pseudotyping; however, the molecular mechanisms governing acquisition of these glycoproteins are poorly understood. For murine leukemia virus envelope (MLV Env) glycoprotein, incorporation into foreign viral particles has been shown to be an active process, but it does not appear to be caused by direct interactions among viral proteins. In this study, we coupled in vivo selection systems with Illumina next-generation sequencing (NGS) to test hundreds of thousands of MLV Env mutants for the ability to be enriched in viral particles and to perform other glycoprotein functions. NGS analyses on a subset of these mutants predicted that the residues important for incorporation are in the membrane-proximal external region (MPER), particularly W127 and W137, and the residues in the membrane-spanning domain (MSD) and also immediately flanking it (T140 to L163). These predictions were validated by directly measuring the impact of mutations in these regions on fusogenicity, infectivity, and incorporation. We suggest that these two regions dictate pseudotyping through interactions with specific lipid environments formed during viral assembly. IMPORTANCE: Researchers from numerous fields routinely exploit the ability to manipulate viral tropism by swapping viral surface proteins. However, this process, termed pseudotyping, is poorly understood at the molecular level. For murine leukemia virus envelope (MLV Env) glycoprotein, incorporation into foreign viral particles is an active process, but it does not appear to occur through direct viral protein-protein interactions. In this study, we tested hundreds of thousands of MLV Env mutants for the ability to be enriched in viral particles as well as perform other glycoprotein functions. Our analyses on a subset of these mutants predict that the glycoprotein regions embedded in and immediately flanking the viral membrane dictate active incorporation into viral particles. We suggest that pseudotyping occurs through specific lipid-protein interactions at the viral assembly site.


Asunto(s)
Células HEK293/virología , Virus de la Leucemia Murina/genética , Infecciones por Retroviridae/virología , Proteínas del Envoltorio Viral/metabolismo , Ensamble de Virus , Internalización del Virus , Secuencia de Aminoácidos , Animales , Fusión Celular , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mutagénesis , Mutación/genética , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA