Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 211
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 74(1): 196-211.e11, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30799147

RESUMEN

The compendium of RNA-binding proteins (RBPs) has been greatly expanded by the development of RNA-interactome capture (RIC). However, it remained unknown if the complement of RBPs changes in response to environmental perturbations and whether these rearrangements are important. To answer these questions, we developed "comparative RIC" and applied it to cells challenged with an RNA virus called sindbis (SINV). Over 200 RBPs display differential interaction with RNA upon SINV infection. These alterations are mainly driven by the loss of cellular mRNAs and the emergence of viral RNA. RBPs stimulated by the infection redistribute to viral replication factories and regulate the capacity of the virus to infect. For example, ablation of XRN1 causes cells to be refractory to SINV, while GEMIN5 moonlights as a regulator of SINV gene expression. In summary, RNA availability controls RBP localization and function in SINV-infected cells.


Asunto(s)
Células Epiteliales/virología , Perfilación de la Expresión Génica/métodos , ARN Viral/genética , Proteínas de Unión al ARN/genética , Virus Sindbis/genética , Transcriptoma , Neoplasias del Cuello Uterino/virología , Regiones no Traducidas 5' , Sitios de Unión , Células Epiteliales/metabolismo , Exorribonucleasas/genética , Exorribonucleasas/metabolismo , Femenino , Regulación Viral de la Expresión Génica , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Unión Proteica , ARN Viral/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/genética , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Proteínas del Complejo SMN , Virus Sindbis/crecimiento & desarrollo , Virus Sindbis/metabolismo , Virus Sindbis/patogenicidad , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo , Replicación Viral
2.
PLoS Pathog ; 15(5): e1007798, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31116799

RESUMEN

Cellular antiviral programs encode molecules capable of targeting multiple steps in the virus lifecycle. Zinc-finger antiviral protein (ZAP) is a central and general regulator of antiviral activity that targets pathogen mRNA stability and translation. ZAP is diffusely cytoplasmic, but upon infection ZAP is targeted to particular cytoplasmic structures, termed stress granules (SGs). However, it remains unclear if ZAP's antiviral activity correlates with SG localization, and what molecular cues are required to induce this localization event. Here, we use Sindbis virus (SINV) as a model infection and find that ZAP's localization to SGs can be transient. Sometimes no apparent viral infection follows ZAP SG localization but ZAP SG localization always precedes accumulation of SINV non-structural protein, suggesting virus replication processes trigger SG formation and ZAP recruitment. Data from single-molecule RNA FISH corroborates this finding as the majority of cells with ZAP localization in SGs contain low levels of viral RNA. Furthermore, ZAP recruitment to SGs occurred in ZAP-expressing cells when co-cultured with cells replicating full-length SINV, but not when co-cultured with cells replicating a SINV replicon. ZAP recruitment to SGs is functionally important as a panel of alanine ZAP mutants indicate that the anti-SINV activity is correlated with ZAP's ability to localize to SGs. As ZAP is a central component of the cellular antiviral programs, these data provide further evidence that SGs are an important cytoplasmic antiviral hub. These findings provide insight into how antiviral components are regulated upon virus infection to inhibit virus spread.


Asunto(s)
Infecciones por Alphavirus/prevención & control , Antivirales/farmacología , Gránulos Citoplasmáticos/metabolismo , Proteínas de Unión al ARN/farmacología , Virus Sindbis/patogenicidad , Estrés Fisiológico , Replicación Viral/efectos de los fármacos , Infecciones por Alphavirus/metabolismo , Infecciones por Alphavirus/virología , Antivirales/metabolismo , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/metabolismo , Neoplasias Óseas/virología , Humanos , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/metabolismo , Osteosarcoma/virología , Transporte de Proteínas , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Células Tumorales Cultivadas
3.
Nucleic Acids Res ; 46(7): 3657-3670, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29361131

RESUMEN

Alphaviruses are mosquito-borne pathogens that cause human diseases ranging from debilitating arthritis to lethal encephalitis. Studies with Sindbis virus (SINV), which causes fever, rash, and arthralgia in humans, and Venezuelan equine encephalitis virus (VEEV), which causes encephalitis, have identified RNA structural elements that play key roles in replication and pathogenesis. However, a complete genomic structural profile has not been established for these viruses. We used the structural probing technique SHAPE-MaP to identify structured elements within the SINV and VEEV genomes. Our SHAPE-directed structural models recapitulate known RNA structures, while also identifying novel structural elements, including a new functional element in the nsP1 region of SINV whose disruption causes a defect in infectivity. Although RNA structural elements are important for multiple aspects of alphavirus biology, we found the majority of RNA structures were not conserved between SINV and VEEV. Our data suggest that alphavirus RNA genomes are highly divergent structurally despite similar genomic architecture and sequence conservation; still, RNA structural elements are critical to the viral life cycle. These findings reframe traditional assumptions about RNA structure and evolution: rather than structures being conserved, alphaviruses frequently evolve new structures that may shape interactions with host immune systems or co-evolve with viral proteins.


Asunto(s)
Virus de la Encefalitis Equina Venezolana/genética , ARN/genética , Virus Sindbis/genética , Replicación Viral/genética , Alphavirus/química , Alphavirus/genética , Alphavirus/patogenicidad , Animales , Encefalitis/genética , Encefalitis/virología , Virus de la Encefalitis Equina Venezolana/química , Virus de la Encefalitis Equina Venezolana/patogenicidad , Genoma Viral/genética , Caballos/virología , Humanos , Conformación de Ácido Nucleico , ARN/química , Virus Sindbis/química , Virus Sindbis/patogenicidad
4.
Proc Natl Acad Sci U S A ; 114(10): 2681-2686, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28213497

RESUMEN

The innate immune system senses RNA viruses by pattern recognition receptors (PRRs) and protects the host from virus infection. PRRs mediate the production of immune modulatory factors and direct the elimination of RNA viruses. Here, we show a unique PRR that mediates antiviral response. Tetrachlorodibenzo-p-dioxin (TCDD)-inducible poly(ADP ribose) polymerase (TIPARP), a Cysteine3 Histidine (CCCH)-type zinc finger-containing protein, binds to Sindbis virus (SINV) RNA via its zinc finger domain and recruits an exosome to induce viral RNA degradation. TIPARP typically localizes in the nucleus, but it accumulates in the cytoplasm after SINV infection, allowing targeting of cytoplasmic SINV RNA. Redistribution of TIPARP is induced by reactive oxygen species (ROS)-dependent oxidization of the nuclear pore that affects cytoplasmic-nuclear transport. BCL2-associated X protein (BAX) and BCL2 antagonist/killer 1 (BAK1), B-cell leukemia/lymphoma 2 (BCL2) family members, mediate mitochondrial damage to generate ROS after SINV infection. Thus, TIPARP is a viral RNA-sensing PRR that mediates antiviral responses triggered by BAX- and BAK1-dependent mitochondrial damage.


Asunto(s)
Inmunidad Innata/genética , Poli(ADP-Ribosa) Polimerasas/genética , Virus ARN/genética , Receptores de Reconocimiento de Patrones/genética , Transporte Activo de Núcleo Celular/genética , Transporte Activo de Núcleo Celular/inmunología , Citoplasma/genética , Citoplasma/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Mitocondrias/genética , Mitocondrias/patología , Mitocondrias/virología , Proteínas de Transporte de Nucleósidos , Poli(ADP-Ribosa) Polimerasas/inmunología , Virus ARN/inmunología , Especies Reactivas de Oxígeno/metabolismo , Receptores de Reconocimiento de Patrones/inmunología , Virus Sindbis/genética , Virus Sindbis/inmunología , Virus Sindbis/patogenicidad , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína Destructora del Antagonista Homólogo bcl-2/inmunología , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/inmunología
5.
J Virol ; 92(7)2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29321325

RESUMEN

Arthropod-borne viruses, such as the members of the genus Alphavirus, are a significant concern to global public health. As obligate intracellular pathogens, RNA viruses must interact with the host cell machinery to establish and complete their life cycles. Despite considerable efforts to define the host-pathogen interactions essential for alphaviral replication, an unbiased and inclusive assessment of alphaviral RNA-protein interactions has not been undertaken. Moreover, the biological and molecular importance of these interactions, in the full context of their molecular function as RNA-binding proteins, has not been fully realized. The data presented here introduce a robust viral RNA-protein discovery method to elucidate the Sindbis virus (SINV) RNA-protein host interface. Cross-link-assisted mRNP purification (CLAMP) assessment revealed an extensive array of host-pathogen interactions centered on the viral RNAs (vRNAs). After prioritization of the host proteins associated with the vRNAs, we identified the site of protein-vRNA interaction by a UV cross-linking and immunoprecipitation sequencing (CLIP-seq) approach and assessed the consequences of the RNA-protein binding event of hnRNP K, hnRNP I, and hnRNP M in regard to viral infection. Here, we demonstrate that mutation of the prioritized hnRNP-vRNA interaction sites effectively disrupts hnRNP-vRNA interaction. Correlating with disrupted hnRNP-vRNA binding, SINV growth kinetics were reduced relative to wild-type parental viral infections in vertebrate and invertebrate tissue culture models of infection. The molecular mechanism leading to reduced viral growth kinetics was found to be dysregulated structural-gene expression. Collectively, this study further defines the scope and importance of the alphavirus host-pathogen vRNA-protein interactions.IMPORTANCE Members of the genus Alphavirus are widely recognized for their potential to cause severe disease. Despite this recognition, there are no antiviral therapeutics, or safe and effective vaccines, currently available to treat alphaviral infection. Alphaviruses utilize the host cell machinery to efficiently establish and complete their life cycle. However, the extent and importance of host-pathogen RNA-protein interactions are woefully undercharacterized. The efforts detailed in this study fill this critical gap, and the significance of this research is 3-fold. First, the data presented here fundamentally expand the scope and understanding of alphavirus host-pathogen interactions. Second, this study identifies the sites of interaction for several prioritized interactions and defines the contribution of the RNA-protein interaction at the molecular level. Finally, these studies build a strategy by which the importance of the given host-pathogen interactions may be assessed in the future, using a mouse model of infection.


Asunto(s)
Infecciones por Alphavirus/virología , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Interacciones Huésped-Patógeno , ARN Viral/metabolismo , Virus Sindbis/patogenicidad , Replicación Viral , Infecciones por Alphavirus/metabolismo , Células Cultivadas , Ribonucleoproteínas Nucleares Heterogéneas/genética , Humanos , ARN Viral/genética , Virus Sindbis/genética , Ensamble de Virus
6.
J Virol ; 92(6)2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29263262

RESUMEN

Alphaviruses are an important cause of mosquito-borne outbreaks of arthritis, rash, and encephalomyelitis. Previous studies in mice with a virulent strain (neuroadapted SINV [NSV]) of the alphavirus Sindbis virus (SINV) identified a role for Th17 cells and regulation by interleukin-10 (IL-10) in the pathogenesis of fatal encephalomyelitis (K. A. Kulcsar, V. K. Baxter, I. P. Greene, and D. E. Griffin, Proc Natl Acad Sci U S A 111:16053-16058, 2014, https://doi.org/10.1073/pnas.1418966111). To determine the role of virus virulence in generation of immune responses, we analyzed the modulatory effects of IL-10 on disease severity, virus clearance, and the CD4+ T cell response to infection with a recombinant strain of SINV of intermediate virulence (TE12). The absence of IL-10 during TE12 infection led to longer morbidity, more weight loss, higher mortality, and slower viral clearance than in wild-type mice. More severe disease and impaired virus clearance in IL-10-/- mice were associated with more Th1 cells, fewer Th2 cells, innate lymphoid type 2 cells, regulatory cells, and B cells, and delayed production of antiviral antibody in the central nervous system (CNS) without an effect on Th17 cells. Therefore, IL-10 deficiency led to more severe disease in TE12-infected mice by increasing Th1 cells and by hampering development of the local B cell responses necessary for rapid production of antiviral antibody and virus clearance from the CNS. In addition, the shift from Th17 to Th1 responses with decreased virus virulence indicates that the effects of IL-10 deficiency on immunopathologic responses in the CNS during alphavirus infection are influenced by virus strain.IMPORTANCE Alphaviruses cause mosquito-borne outbreaks of encephalomyelitis, but determinants of outcome are incompletely understood. We analyzed the effects of the anti-inflammatory cytokine IL-10 on disease severity and virus clearance after infection with an alphavirus strain of intermediate virulence. The absence of IL-10 led to longer illness, more weight loss, more death, and slower viral clearance than in mice that produced IL-10. IL-10 influenced development of disease-causing T cells and entry into the brain of B cells producing antiviral antibody. The Th1 pathogenic cell subtype that developed in IL-10-deficient mice infected with a less virulent virus was distinct from the Th17 subtype that developed in response to a more virulent virus, indicating a role for virus strain in determining the immune response. Slow production of antibody in the nervous system led to delayed virus clearance. Therefore, both the virus strain and the host response to infection are important determinants of outcome.


Asunto(s)
Infecciones por Alphavirus/inmunología , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Encefalomielitis/inmunología , Interleucina-10/inmunología , Virus Sindbis/inmunología , Infecciones por Alphavirus/genética , Infecciones por Alphavirus/patología , Animales , Anticuerpos Antivirales/inmunología , Formación de Anticuerpos/genética , Linfocitos B/patología , Linfocitos T CD4-Positivos/patología , Línea Celular , Cricetinae , Encefalomielitis/genética , Encefalomielitis/patología , Interleucina-10/genética , Ratones , Ratones Noqueados , Virus Sindbis/genética , Virus Sindbis/patogenicidad
7.
PLoS Pathog ; 13(6): e1006473, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28662211

RESUMEN

Alphaviruses are arthropod-borne viruses that represent a significant threat to public health at a global level. While the formation of alphaviral nucleocapsid cores, consisting of cargo nucleic acid and the viral capsid protein, is an essential molecular process of infection, the precise interactions between the two partners are ill-defined. A CLIP-seq approach was used to screen for candidate sites of interaction between the viral Capsid protein and genomic RNA of Sindbis virus (SINV), a model alphavirus. The data presented in this report indicates that the SINV capsid protein binds to specific viral RNA sequences in the cytoplasm of infected cells, but its interaction with genomic RNA in mature extracellular viral particles is largely non-specific in terms of nucleotide sequence. Mutational analyses of the cytoplasmic viral RNA-capsid interaction sites revealed a functional role for capsid binding early in infection. Interaction site mutants exhibited decreased viral growth kinetics; however, this defect was not a function of decreased particle production. Rather mutation of the cytoplasmic capsid-RNA interaction sites negatively affected the functional capacity of the incoming viral genomic RNAs leading to decreased infectivity. Furthermore, cytoplasmic capsid interaction site mutants are attenuated in a murine model of neurotropic alphavirus infection. Collectively, the findings of this study indicate that the identified cytoplasmic interactions of the viral capsid protein and genomic RNA, while not essential for particle formation, are necessary for genomic RNA function early during infection. This previously unappreciated role of capsid protein during the alphaviral replication cycle also constitutes a novel virulence determinant.


Asunto(s)
Proteínas de la Cápside/metabolismo , ARN Viral/metabolismo , Virus Sindbis/metabolismo , Animales , Cápside/metabolismo , Citoplasma/metabolismo , Genoma Viral/genética , Virus Sindbis/genética , Virus Sindbis/patogenicidad , Proteínas del Envoltorio Viral/metabolismo , Virión/metabolismo , Virulencia/fisiología , Ensamble de Virus/fisiología
8.
Rev Med Virol ; 26(4): 221-41, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26990827

RESUMEN

Sindbis virus (SINV; family Togaviridae, genus Alphavirus) is an enveloped RNA virus widely distributed in Eurasia, Africa, Oceania and Australia. SINV is transmitted among its natural bird hosts via mosquitoes. Human disease caused by SINV infection has been reported mainly in South Africa and in Northern Europe. Vector mosquito abundance affects the annual incidence of SINV infections with occasional outbreaks of up to 1500 patients. Symptoms include fever, malaise, rash and musculoskeletal pain. In a significant portion of patients the debilitating musculoskeletal symptoms persist for years. Chronic disease after SINV infection shares many features with autoimmune diseases. Currently there is no specific treatment available. Recently SINV infections have been detected outside the previously known distribution range. In this article we will summarize the current knowledge on epidemiology, clinical disease and pathogenesis of SINV infection in man. Copyright © 2016 John Wiley & Sons, Ltd.


Asunto(s)
Infecciones por Alphavirus/epidemiología , Infecciones por Alphavirus/patología , Virus Sindbis/patogenicidad , Zoonosis/epidemiología , Zoonosis/virología , Infecciones por Alphavirus/veterinaria , Infecciones por Alphavirus/virología , Animales , Enfermedades de las Aves/epidemiología , Enfermedades de las Aves/virología , Aves , Brotes de Enfermedades , Salud Global , Humanos , Incidencia , Topografía Médica
9.
Emerg Infect Dis ; 21(12): 2225-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26583836

RESUMEN

Old World alphaviruses were identified in 52 of 623 horses with febrile or neurologic disease in South Africa. Five of 8 Sindbis virus infections were mild; 2 of 3 fatal cases involved co-infections. Of 44 Middelburg virus infections, 28 caused neurologic disease; 12 were fatal. Middelburg virus likely has zoonotic potential.


Asunto(s)
Caballos/virología , Virus Sindbis/patogenicidad , Zoonosis/epidemiología , Alphavirus/genética , Animales , Caballos/genética , Humanos , Filogenia , Virus Sindbis/genética , Sudáfrica/epidemiología
10.
Biochem Biophys Res Commun ; 462(4): 426-32, 2015 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-25976675

RESUMEN

We examined the impact of Sindbis virus (SINV) infection on cell cycle progression in a cancer cell line, HeLa, and a non-cancerous cell line, Vero. Cell cycle analyses showed that SINV infection is able to alter the cell cycle progression in both HeLa and Vero cells, but differently, especially during the early stage of infection. SINV infection affected the expression of several cell cycle regulators (CDK4, CDK6, cyclin E, p21, cyclin A and cyclin B) in HeLa cells and caused HeLa cells to accumulate in S phase during the early stage of infection. Monitoring SINV replication in HeLa and Vero cells expressing cell cycle indicators revealed that SINV which infected HeLa cells during G1 phase preferred to proliferate during S/G2 phase, and the average time interval for viral replication was significantly shorter in both HeLa and Vero cells infected during G1 phase than in cells infected during S/G2 phase.


Asunto(s)
Ciclo Celular , Virus Sindbis/patogenicidad , Células HeLa , Humanos , Virus Sindbis/fisiología , Replicación Viral
11.
J Virol ; 87(22): 12216-26, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24006438

RESUMEN

The genus Alphavirus consists of a group of enveloped, single-stranded RNA viruses, many of which are transmitted by arthropods to a wide range of vertebrate host species. Here we report that Sindbis virus (SINV) produced from a representative mammalian cell line consists of at least two unique particle subpopulations, separable on the basis of virion density. In contrast, mosquito-derived SINV consists of a homogeneous population of particles. Our findings indicate that the denser particle subpopulation, SINV(Heavy), is more infectious on a per-particle basis than SINV(Light). SINV produced in mosquito cell lines (SINV(C6/36)) exhibited particle-to-PFU ratios similar to those observed for SINV(Heavy). In mammalian cells, viral RNA was synthesized and accumulated more rapidly following infection with SINV(Heavy) or SINV(C6/36) than following infection with SINV(Light), due partly to enhanced translation of viral genomic RNA early in infection. Analysis of the individual particle subpopulations indicated that SINV(Heavy) and SINV(C6/36) contain host-derived factors whose presence correlates with the enhanced translation, RNA synthesis, and infectivity observed for these particles.


Asunto(s)
Infecciones por Alphavirus/transmisión , Culicidae/virología , Fibroblastos/virología , Interacciones Huésped-Patógeno , Riñón/virología , Virus Sindbis/patogenicidad , Infecciones por Alphavirus/virología , Animales , Células Cultivadas , Cricetinae , Reactivos de Enlaces Cruzados , Fibroblastos/patología , Células HEK293 , Humanos , Inmunoprecipitación , Riñón/patología , Ratones , Reacción en Cadena de la Polimerasa , ARN Viral/genética , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Internalización del Virus , Replicación Viral
12.
J Virol ; 87(17): 9579-89, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23785213

RESUMEN

Alphaviruses are enveloped viruses with highly organized structures. The nucleocapsid (NC) core contains a capsid protein lattice enclosing the plus-sense RNA genome, and it is surrounded by a lipid bilayer containing a lattice of the E1 and E2 envelope glycoproteins. Capsid protein is synthesized in the cytoplasm and particle budding occurs at the plasma membrane (PM), but the traffic and assembly of viral components and the exit of virions from host cells are not well understood. To visualize the dynamics of capsid protein during infection, we developed a Sindbis virus infectious clone tagged with a tetracysteine motif. Tagged capsid protein could be fluorescently labeled with biarsenical dyes in living cells without effects on virus growth, morphology, or protein distribution. Live cell imaging and colocalization experiments defined distinct groups of capsid foci in infected cells. We observed highly motile internal puncta that colocalized with E2 protein, which may represent the transport machinery that capsid protein uses to reach the PM. Capsid was also found in larger nonmotile internal structures that colocalized with cellular G3BP and viral nsP3. Thus, capsid may play an unforeseen role in these previously observed G3BP-positive foci, such as regulation of cellular stress granules. Capsid puncta were also observed at the PM. These puncta colocalized with E2 and recruited newly synthesized capsid protein; thus, they may be sites of virus assembly and egress. Together, our studies provide the first dynamic views of the alphavirus capsid protein in living cells and a system to define detailed mechanisms during alphavirus infection.


Asunto(s)
Proteínas de la Cápside/fisiología , Virus Sindbis/fisiología , Infecciones por Alphavirus/virología , Animales , Transporte Biológico Activo , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Línea Celular , Membrana Celular/virología , Chlorocebus aethiops , Cricetinae , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Interacciones Huésped-Patógeno/fisiología , Proteínas Luminiscentes/química , Proteínas Luminiscentes/genética , Virus Sindbis/genética , Virus Sindbis/patogenicidad , Células Vero , Proteínas del Envoltorio Viral/fisiología , Proteínas no Estructurales Virales/fisiología , Ensamble de Virus/fisiología , Liberación del Virus/fisiología , Replicación Viral/fisiología , Proteína Fluorescente Roja
13.
J Virol ; 87(15): 8317-26, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23740988

RESUMEN

Due to inactivation of the α1,3-galactosyltransferase gene (GGTA1, or the α1,3GT gene) approximately 28 million years ago, the carbohydrate αGal (Galα1,3Galß1,4GlcNAc) is not expressed on the cells of Old World monkeys and apes (including humans) but is expressed in all other mammals. The proposed selective advantage of this mutation for these primates is the ability to produce anti-Gal antibodies, which may be an effective immune component in neutralizing αGal-expressing pathogens. However, loss of α1,3GT expression may have been advantageous by providing natural resistance against viral pathogens that exploited the α1,3GT pathway or cell surface αGal for infection. Infections of paired cell lines with differential expression of α1,3GT showed that Sindbis viruses (SINV) preferentially replicate in α1,3GT-positive cells, whereas herpes simplex viruses type 1 and type 2 (HSV-1 and HSV-2) preferentially grow in cells lacking α1,3GT. Viral growth and spread correlated with the ability of the different viruses to successfully initiate infection in the presence or absence of α1,3GT expression. GT knockout (KO) suckling mice infected with SINV strains (AR339 and S.A.AR86) experienced significant delay in onset of disease symptoms and mortality compared to wild-type (WT) B6 suckling mice. In contrast, HSV-2-infected GT KO mice had higher viral titers in spleen and liver and exhibited significantly more focal hepatic necrosis than WT B6 mice. This study demonstrates that α1,3GT activity plays a role in the course of infections for certain viruses. Furthermore, this study has implications for the evolution of resistance to viral infections in primates.


Asunto(s)
Resistencia a la Enfermedad , Evolución Molecular , Galactosiltransferasas/genética , Receptores Virales/genética , Virosis/inmunología , Internalización del Virus , Fenómenos Fisiológicos de los Virus , Infecciones por Alphavirus/patología , Infecciones por Alphavirus/virología , Animales , Línea Celular , Cercopithecidae , Modelos Animales de Enfermedad , Femenino , Galactosiltransferasas/metabolismo , Eliminación de Gen , Herpes Simple/patología , Herpes Simple/virología , Herpesvirus Humano 1/crecimiento & desarrollo , Herpesvirus Humano 1/fisiología , Herpesvirus Humano 2/crecimiento & desarrollo , Herpesvirus Humano 2/fisiología , Humanos , Hígado/patología , Hígado/virología , Masculino , Ratones , Ratones Noqueados , Receptores Virales/metabolismo , Selección Genética , Virus Sindbis/crecimiento & desarrollo , Virus Sindbis/patogenicidad , Virus Sindbis/fisiología , Bazo/patología , Bazo/virología
14.
PLoS Pathog ; 8(7): e1002783, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22792064

RESUMEN

Ubiquitin (Ub) is a vital regulatory component in various cellular processes, including cellular responses to viral infection. As obligate intracellular pathogens, viruses have the capacity to manipulate the ubiquitin (Ub) cycle to their advantage by encoding Ub-modifying proteins including deubiquitinases (DUBs). However, how cellular DUBs modulate specific viral infections, such as norovirus, is poorly understood. To examine the role of DUBs during norovirus infection, we used WP1130, a small molecule inhibitor of a subset of cellular DUBs. Replication of murine norovirus in murine macrophages and the human norovirus Norwalk virus in a replicon system were significantly inhibited by WP1130. Chemical proteomics identified the cellular DUB USP14 as a target of WP1130 in murine macrophages, and pharmacologic inhibition or siRNA-mediated knockdown of USP14 inhibited murine norovirus infection. USP14 is a proteasome-associated DUB that also binds to inositol-requiring enzyme 1 (IRE1), a critical mediator of the unfolded protein response (UPR). WP1130 treatment of murine macrophages did not alter proteasome activity but activated the X-box binding protein-1 (XBP-1) through an IRE1-dependent mechanism. In addition, WP1130 treatment or induction of the UPR also reduced infection of other RNA viruses including encephalomyocarditis virus, Sindbis virus, and La Crosse virus but not vesicular stomatitis virus. Pharmacologic inhibition of the IRE1 endonuclease activity partially rescued the antiviral effect of WP1130. Taken together, our studies support a model whereby induction of the UPR through cellular DUB inhibition blocks specific viral infections, and suggest that cellular DUBs and the UPR represent novel targets for future development of broad spectrum antiviral therapies.


Asunto(s)
Antivirales/farmacología , Nitrilos/farmacología , Norovirus/efectos de los fármacos , Piridinas/farmacología , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos , Animales , Infecciones por Caliciviridae/tratamiento farmacológico , Infecciones por Caliciviridae/virología , Línea Celular , Línea Celular Tumoral , Cianoacrilatos , Proteínas de Unión al ADN/metabolismo , Virus de la Encefalomiocarditis/efectos de los fármacos , Virus de la Encefalomiocarditis/patogenicidad , Inhibidores Enzimáticos/farmacología , Humanos , Virus La Crosse/efectos de los fármacos , Virus La Crosse/patogenicidad , Macrófagos/virología , Proteínas de la Membrana/metabolismo , Ratones , Norovirus/fisiología , Virus Norwalk/efectos de los fármacos , Virus Norwalk/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Factores de Transcripción del Factor Regulador X , Virus Sindbis/efectos de los fármacos , Virus Sindbis/patogenicidad , Factores de Transcripción/metabolismo , Ubiquitina/metabolismo , Ubiquitina Tiolesterasa/genética , Replicación Viral/efectos de los fármacos , Proteína 1 de Unión a la X-Box
15.
J Virol ; 86(13): 7180-91, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22514352

RESUMEN

The Old World alphaviruses are emerging human pathogens with an ability to cause widespread epidemics. The latest epidemic of Chikungunya virus, from 2005 to 2007, affected over 40 countries in Africa, Asia, and Europe. The Old World alphaviruses are highly cytopathic and known to evade the cellular antiviral response by inducing global inhibition of transcription in vertebrate cells. This function was shown to be mediated by their nonstructural nsP2 protein; however, the detailed mechanism of this phenomenon has remained unknown. Here, we report that nsP2 proteins of Sindbis, Semliki Forest, and Chikungunya viruses inhibit cellular transcription by inducing rapid degradation of Rpb1, a catalytic subunit of the RNAPII complex. This degradation of Rpb1 is independent of the nsP2-associated protease activity, but, instead, it proceeds through nsP2-mediated Rpb1 ubiquitination. This function of nsP2 depends on the integrity of the helicase and S-adenosylmethionine (SAM)-dependent methyltransferase-like domains, and point mutations in either of these domains abolish Rpb1 degradation. We go on to show that complete degradation of Rpb1 in alphavirus-infected cells occurs within 6 h postinfection, before other previously described virus-induced changes in cell physiology, such as apoptosis, autophagy, and inhibition of STAT1 phosphorylation, are detected. Since Rpb1 is a subunit that catalyzes the polymerase reaction during RNA transcription, degradation of Rpb1 plays an indispensable role in blocking the activation of cellular genes and downregulating cellular antiviral response. This indicates that the nsP2-induced degradation of Rpb1 is a critical mechanism utilized by the Old World alphaviruses to subvert the cellular antiviral response.


Asunto(s)
Virus Chikungunya/enzimología , Cisteína Endopeptidasas/metabolismo , Evasión Inmune , Proteolisis , ARN Polimerasa II/antagonistas & inhibidores , Virus de los Bosques Semliki/enzimología , Virus Sindbis/enzimología , Animales , Dominio Catalítico , Línea Celular , Virus Chikungunya/patogenicidad , Cricetinae , Inmunidad Innata , Ratones , ARN Polimerasa II/metabolismo , Virus de los Bosques Semliki/patogenicidad , Virus Sindbis/patogenicidad , Ubiquitinación
16.
Insect Mol Biol ; 22(1): 104-14, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23331493

RESUMEN

RNA interference (RNAi) is the major innate antiviral pathway in Aedes aegypti that responds to replicating arboviruses such as dengue virus (DENV) and Sindbis virus (SINV). On the one hand, the mosquito's RNAi machinery is capable of completely eliminating DENV2 from Ae. aegypti. On the other, transient silencing of key genes of the RNAi pathway increases replication of SINV and DENV2, allowing the viruses to temporally overcome dose-dependent midgut infection and midgut escape barriers (MEB) more efficiently. Here we expressed Flock house virus B2 (FHV-B2) from the poly-ubiquitin (PUb) promoter in Ae. aegypti using the ΦC31 site-directed recombination system to investigate the impact of transgene-mediated RNAi pathway suppression on infections with SINV-TR339eGFP and DENV2-QR94, the latter of which has been shown to be confronted with a strong MEB in Ae. aegypti. FHV-B2 was constitutively expressed in midguts of sugar- and blood-fed mosquitoes of transgenic line PUbB2 P61. B2 over-expression suppressed RNA silencing of carboxypeptidase A-1 (AeCPA-1) in midgut tissue of PUbB2 P61 mosquitoes. Following oral challenge with SINV-TR339eGFP or DENV2-QR94, mean titres in midguts of PUbB2 P61 females were significantly higher at 7 days post-bloodmeal (pbm) than in those of nontransgenic control mosquitoes. At 14 days pbm, infection rates of carcasses were significantly increased in PubB2 P61 mosquitoes infected with SINV-TR339eGFP. Following infection with DENV2-QR94, midgut infection rates were significantly increased in the B2-expressing mosquitoes at 14 days pbm. However, B2 expression in PUbB2 P61 did not increase the DENV2-QR94 dissemination rate, indicating that the infection phenotype was not primarily controlled by RNAi.


Asunto(s)
Aedes/genética , Aedes/virología , Animales Modificados Genéticamente , Virus del Dengue/fisiología , Virus Sindbis/fisiología , Replicación Viral/genética , Animales , Virus del Dengue/patogenicidad , Femenino , Tracto Gastrointestinal/fisiología , Expresión Génica , Silenciador del Gen , Nodaviridae/genética , Poliubiquitina/genética , Regiones Promotoras Genéticas , Interferencia de ARN , Virus Sindbis/patogenicidad , Transgenes
17.
J Virol ; 85(21): 11490-501, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21865385

RESUMEN

Sindbis virus (SINV) infection of the central nervous system (CNS) provides a model for understanding the role of the immune response in recovery from alphavirus infection of neurons. Virus clearance occurred in three phases: clearance of infectious virus (days 3 to 7), clearance of viral RNA (days 8 to 60), and maintenance of low levels of viral RNA (>day 60). The antiviral immune response was initiated in the cervical lymph nodes with rapid extrafollicular production of plasmablasts secreting IgM, followed by germinal center production of IgG-secreting and memory B cells. The earliest inflammatory cells to enter the brain were CD8(+) T cells, followed by CD4(+) T cells and CD19(+) B cells. During the clearance of infectious virus, effector lymphocytes in the CNS were primarily CD8(+) T cells and IgM antibody-secreting cells (ASCs). During the clearance of viral RNA, there were more CD4(+) than CD8(+) T cells, and B cells included IgG and IgA ASCs. At late times after infection, ASCs in the CNS were primarily CD19(+) CD38(+) CD138(-) Blimp-1(+) plasmablasts, with few fully differentiated CD38(-) CD138(+) Blimp-1(+) plasma cells. CD19(+) CD38(+) surface Ig(+) memory B cells were also present. The level of antibody to SINV increased in the brain over time, and the proportion of SINV-specific ASCs increased from 15% of total ASCs at day 14 to 90% at 4 to 6 months, suggesting specific retention in the CNS during viral RNA persistence. B cells in the CNS continued to differentiate, as evidenced by accumulation of IgA ASCs not present in peripheral lymphoid tissue and downregulation of major histocompatibility complex (MHC) class II expression on plasmablasts. However, there was no evidence of germinal center activity or IgG avidity maturation within the CNS.


Asunto(s)
Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/patología , Células Productoras de Anticuerpos/inmunología , Encefalomielitis/inmunología , Encefalomielitis/patología , Virus Sindbis/inmunología , Virus Sindbis/patogenicidad , Infecciones por Alphavirus/virología , Animales , Antígenos CD/análisis , Linfocitos B/química , Linfocitos B/inmunología , Encéfalo/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Encefalomielitis/virología , Femenino , Centro Germinal/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Ganglios Linfáticos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Sistema Nervioso/inmunología , Sistema Nervioso/patología , Sistema Nervioso/virología
18.
J Virol ; 84(21): 11429-39, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20739538

RESUMEN

Previous studies with Venezuelan equine encephalitis virus and Sindbis virus (SINV) indicate that alphaviruses are capable of suppressing the cellular response to type I and type II interferons (IFNs) by disrupting Jak/STAT signaling; however, the relevance of this signaling inhibition toward pathogenesis has not been investigated. The relative abilities of neurovirulent and nonneurovirulent SINV strains to downregulate Jak/STAT signaling were compared to determine whether the ability to inhibit IFN signaling correlates with virulence potential. The adult mouse neurovirulent strain AR86 was found to rapidly and robustly inhibit tyrosine phosphorylation of STAT1 and STAT2 in response to IFN-γ and/or IFN-ß. In contrast, the closely related SINV strains Girdwood and TR339, which do not cause detectable disease in adult mice, were relatively inefficient inhibitors of STAT1/2 activation. Decreased STAT activation in AR86-infected cells was associated with decreased activation of the IFN receptor-associated tyrosine kinases Tyk2, Jak1, and Jak2. To identify the viral factor(s) involved, we infected cells with several panels of AR86/Girdwood chimeric viruses. Surprisingly, we found that a single amino acid determinant, threonine at nsP1 position 538, which is required for AR86 virulence, was also required for efficient disruption of STAT1 activation, and this determinant fully restored STAT1 inhibition when it was introduced into the avirulent Girdwood background. These data indicate that a key virulence determinant plays a critical role in downregulating the response to type I and type II IFNs, which suggests that the ability of alphaviruses to inhibit Jak/STAT signaling relates to their in vivo virulence potential.


Asunto(s)
Quinasas Janus/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Virus Sindbis/patogenicidad , Animales , Chlorocebus aethiops , Regulación hacia Abajo , Interferón Tipo I , Interferón gamma , Ratones , Fosforilación , Especificidad de la Especie , Células Vero , Virulencia
19.
J Virol ; 84(19): 10004-15, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20668087

RESUMEN

Venezuelan equine encephalitis virus (VEEV) is a significant human and animal pathogen. The highlight of VEEV replication in vitro, in cells of vertebrate origin, is the rapid development of cytopathic effect (CPE), which is strongly dependent upon the expression of viral capsid protein. Besides being an integral part of virions, the latter protein is capable of (i) binding both the nuclear import and nuclear export receptors, (ii) accumulating in the nuclear pore complexes, (iii) inhibiting nucleocytoplasmic trafficking, and (iv) inhibiting transcription of cellular ribosomal and messenger RNAs. Using our knowledge of the mechanism of VEEV capsid protein function in these processes, we designed VEEV variants containing combinations of mutations in the capsid-coding sequences. These mutations made VEEV dramatically less cytopathic but had no effect on infectious virus production. In cell lines that have defects in type I interferon (IFN) signaling, the capsid mutants demonstrated very efficient persistent replication. In other cells, which have no defects in IFN production or signaling, the same mutants were capable of inducing a long-term antiviral state, downregulating virus replication to an almost undetectable level. However, ultimately, these cells also developed a persistent infection, characterized by continuous virus replication and beta IFN (IFN-beta) release. The results of this study demonstrate that the long-term cellular antiviral state is determined by the synergistic effects of type I IFN signaling and the antiviral reaction induced by replicating viral RNA and/or the expression of VEEV-specific proteins. The designed mutants represent an important model for studying the mechanisms of cell interference with VEEV replication and development of persistent infection.


Asunto(s)
Proteínas de la Cápside/genética , Virus de la Encefalitis Equina Venezolana/genética , Virus de la Encefalitis Equina Venezolana/patogenicidad , Encefalomielitis Equina Venezolana/virología , Enfermedad Aguda , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas de la Cápside/fisiología , Células Cultivadas , Cricetinae , Efecto Citopatogénico Viral/genética , Efecto Citopatogénico Viral/fisiología , ADN Viral/genética , Virus de la Encefalitis Equina Venezolana/inmunología , Virus de la Encefalitis Equina Venezolana/fisiología , Encefalomielitis Equina Venezolana/inmunología , Genes Virales , Enfermedades de los Caballos/inmunología , Enfermedades de los Caballos/virología , Caballos , Humanos , Interferón Tipo I/inmunología , Ratones , Datos de Secuencia Molecular , Mutación , Células 3T3 NIH , Homología de Secuencia de Aminoácido , Transducción de Señal/inmunología , Virus Sindbis/genética , Virus Sindbis/patogenicidad , Virus Sindbis/fisiología , Replicación Viral
20.
Cell Microbiol ; 12(8): 1144-57, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20331640

RESUMEN

Poliovirus 2B protein is a well-known viroporin implicated in plasma membrane permeabilization to ions and low-molecular-weight compounds during infection. Translation in mammalian cells expressing 2B protein is inhibited by hygromycin B (HB) but remains unaffected in mock cells, which are not permeable to the inhibitor. Here we describe a previously unreported bystander effect in which healthy baby hamster kidney (BHK) cells become sensitive to HB when co-cultured with a low proportion of cells expressing poliovirus 2B. Viroporins E from mouse hepatitis virus, 6K from Sindbis virus and NS4A protein from hepatitis C virus were also able to permeabilize neighbouring cells to different extents. Expression of 2B induced permeabilization of neighbouring cell lines other than BHK. We found that gap junctions are responsible mediating the observed bystander permeabilization. Gap junctional communication was confirmed in 2B-expressing co-cultures by fluorescent dye transfer. Moreover, the presence of connexin 43 was confirmed in both mock and 2B-transfected cells. Finally, inhibition of HB entry to neighbouring cells was observed with 18alpha-glycyrrhethinic acid, an inhibitor of gap junctions. Taken together, these findings support a mechanism involving gap junctional intercellular communication in the bystander permeabilization effect observed in healthy cells co-cultured with poliovirus 2B-expressing cells.


Asunto(s)
Permeabilidad de la Membrana Celular/efectos de los fármacos , Uniones Comunicantes/efectos de los fármacos , Proteínas no Estructurales Virales/metabolismo , Animales , Proteínas Portadoras/metabolismo , Línea Celular , Técnicas de Cocultivo , Cricetinae , Hepacivirus/patogenicidad , Humanos , Péptidos y Proteínas de Señalización Intracelular , Virus de la Hepatitis Murina/patogenicidad , Poliovirus/patogenicidad , Virus Sindbis/patogenicidad , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA