Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 18(6): e1010228, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35675358

RESUMEN

Influenza A virus (IAV) preferentially infects conducting airway and alveolar epithelial cells in the lung. The outcome of these infections is impacted by the host response, including the production of various cytokines, chemokines, and growth factors. Fibroblast growth factor-9 (FGF9) is required for lung development, can display antiviral activity in vitro, and is upregulated in asymptomatic patients during early IAV infection. We therefore hypothesized that FGF9 would protect the lungs from respiratory virus infection and evaluated IAV pathogenesis in mice that overexpress FGF9 in club cells in the conducting airway epithelium (FGF9-OE mice). However, we found that FGF9-OE mice were highly susceptible to IAV and Sendai virus infection compared to control mice. FGF9-OE mice displayed elevated and persistent viral loads, increased expression of cytokines and chemokines, and increased numbers of infiltrating immune cells as early as 1 day post-infection (dpi). Gene expression analysis showed an elevated type I interferon (IFN) signature in the conducting airway epithelium and analysis of IAV tropism uncovered a dramatic shift in infection from the conducting airway epithelium to the alveolar epithelium in FGF9-OE lungs. These results demonstrate that FGF9 signaling primes the conducting airway epithelium to rapidly induce a localized IFN and proinflammatory cytokine response during viral infection. Although this response protects the airway epithelial cells from IAV infection, it allows for early and enhanced infection of the alveolar epithelium, ultimately leading to increased morbidity and mortality. Our study illuminates a novel role for FGF9 in regulating respiratory virus infection and pathogenesis.


Asunto(s)
Factor 9 de Crecimiento de Fibroblastos , Virus de la Influenza A , Gripe Humana , Interferón Tipo I , Infecciones por Orthomyxoviridae , Animales , Citocinas/metabolismo , Células Epiteliales/metabolismo , Factor 9 de Crecimiento de Fibroblastos/biosíntesis , Humanos , Virus de la Influenza A/metabolismo , Gripe Humana/metabolismo , Gripe Humana/virología , Interferón Tipo I/metabolismo , Ratones , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología
2.
J Virol ; 96(1): e0114321, 2022 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-34668781

RESUMEN

Chikungunya virus (CHIKV) is an arthritogenic alphavirus that causes both debilitating acute and chronic disease. Previous work has shown that type I interferons (IFNs) play a critical role in limiting CHIKV pathogenesis and that interferon alpha (IFN-α) and interferon beta (IFN-ß) control acute CHIKV infection by distinct mechanisms. However, the role of type I IFNs, especially specific subtypes, during chronic CHIKV disease is unclear. To address this gap in knowledge, we evaluated chronic CHIKV pathogenesis in mice lacking IFN-α or IFN-ß. We found that IFN-α was the dominant subtype that controls chronic disease. Despite detecting a varying type I IFN response throughout the course of disease, IFN-α acts within the first few days of infection to control the levels of persistent CHIKV RNA. In addition, using a novel CHIKV-3'-Cre tdTomato reporter system that fate maps CHIKV-infected cells, we showed that IFN-α limits the number of cells that survive CHIKV at sites of dissemination, particularly dermal fibroblasts and immune cells. Though myofibers play a significant role in CHIKV disease, they were not impacted by the loss of IFN-α. Our studies highlight that IFN-α and IFN-ß play divergent roles during chronic CHIKV disease through events that occur early in infection and that not all cell types are equally dependent on type I IFNs for restricting viral persistence. IMPORTANCE Chikungunya virus (CHIKV) is a reemerging global pathogen with no effective vaccine or antiviral treatment for acute or chronic disease, and the mechanisms underlying chronic disease manifestations remain poorly defined. The significance of our research is in defining IFN-α, but not IFN-ß, as an important host regulator of chronic CHIKV pathogenesis that acts within the first 48 hours of infection to limit persistent viral RNA and the number of cells that survive CHIKV infection 1 month post-infection. Loss of IFN-α had a greater impact on immune cells and dermal fibroblasts than myofibers, highlighting the need to delineate cell-specific responses to type I IFNs. Altogether, our work demonstrates that very early events of acute CHIKV infection influence chronic disease. Continued efforts to delineate early host-pathogen interactions may help stratify patients who are at risk for developing chronic CHIKV symptoms and identify therapeutics that may prevent progression to chronic disease altogether.


Asunto(s)
Fiebre Chikungunya/metabolismo , Fiebre Chikungunya/virología , Virus Chikungunya/fisiología , Interacciones Huésped-Patógeno , Interferón-alfa/metabolismo , Interferón beta/metabolismo , Animales , Supervivencia Celular , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Ratones , Ratones Noqueados , ARN Viral , Replicación Viral
3.
PLoS Pathog ; 15(8): e1007993, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31465513

RESUMEN

Chikungunya virus (CHIKV) is an arthritogenic alphavirus that acutely causes fever as well as severe joint and muscle pain. Chronic musculoskeletal pain persists in a substantial fraction of patients for months to years after the initial infection, yet we still have a poor understanding of what promotes chronic disease. While replicating virus has not been detected in joint-associated tissues of patients with persistent arthritis nor in various animal models at convalescent time points, viral RNA is detected months after acute infection. To identify the cells that might contribute to pathogenesis during this chronic phase, we developed a recombinant CHIKV that expresses Cre recombinase (CHIKV-3'-Cre). CHIKV-3'-Cre replicated in myoblasts and fibroblasts, and it induced arthritis during the acute phase in mice. Importantly, it also induced chronic disease, including persistent viral RNA and chronic myositis and synovitis similar to wild-type virus. CHIKV-3'-Cre infection of tdTomato reporter mice resulted in a population of tdTomato+ cells that persisted for at least 112 days. Immunofluorescence and flow cytometric profiling revealed that these tdTomato+ cells predominantly were myofibers and dermal and muscle fibroblasts. Treatment with an antibody against Mxra8, a recently defined host receptor for CHIKV, reduced the number of tdTomato+ cells in the chronic phase and diminished the levels of chronic viral RNA, implicating these tdTomato+ cells as the reservoir of chronic viral RNA. Finally, isolation and flow cytometry-based sorting of the tdTomato+ fibroblasts from the skin and ankle and analysis for viral RNA revealed that the tdTomato+ cells harbor most of the persistent CHIKV RNA at chronic time points. Therefore, this CHIKV-3'-Cre and tdTomato reporter mouse system identifies the cells that survive CHIKV infection in vivo and are enriched for persistent CHIKV RNA. This model represents a useful tool for studying CHIKV pathogenesis in the acute and chronic stages of disease.


Asunto(s)
Artritis Experimental/virología , Fiebre Chikungunya/virología , Virus Chikungunya/patogenicidad , Dermis/patología , Fibroblastos/patología , Músculo Esquelético/patología , ARN Viral/metabolismo , Animales , Artritis Experimental/metabolismo , Artritis Experimental/patología , Fiebre Chikungunya/metabolismo , Virus Chikungunya/genética , Dermis/metabolismo , Dermis/virología , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibroblastos/virología , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Fibras Musculares Esqueléticas/virología , Músculo Esquelético/metabolismo , Músculo Esquelético/virología , ARN Viral/genética , Replicación Viral
4.
J Virol ; 94(1)2019 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-31619554

RESUMEN

Type I interferons (IFNs) are key mediators of the innate immune response. Although members of this family of cytokines signal through a single shared receptor, biochemical and functional variation exists in response to different IFN subtypes. While previous work has demonstrated that type I IFNs are essential to control infection by chikungunya virus (CHIKV), a globally emerging alphavirus, the contributions of individual IFN subtypes remain undefined. To address this question, we evaluated CHIKV pathogenesis in mice lacking IFN-ß (IFN-ß knockout [IFN-ß-KO] mice or mice treated with an IFN-ß-blocking antibody) or IFN-α (IFN regulatory factor 7 knockout [IRF7-KO] mice or mice treated with a pan-IFN-α-blocking antibody). Mice lacking either IFN-α or IFN-ß developed severe clinical disease following infection with CHIKV, with a marked increase in foot swelling compared to wild-type mice. Virological analysis revealed that mice lacking IFN-α sustained elevated infection in the infected ankle and in distant tissues. In contrast, IFN-ß-KO mice displayed minimal differences in viral burdens within the ankle or at distal sites and instead had an altered cellular immune response. Mice lacking IFN-ß had increased neutrophil infiltration into musculoskeletal tissues, and depletion of neutrophils in IFN-ß-KO but not IRF7-KO mice mitigated musculoskeletal disease caused by CHIKV. Our findings suggest disparate roles for the IFN subtypes during CHIKV infection, with IFN-α limiting early viral replication and dissemination and IFN-ß modulating neutrophil-mediated inflammation.IMPORTANCE Type I interferons (IFNs) possess a range of biological activity and protect against a number of viruses, including alphaviruses. Despite signaling through a shared receptor, there are established biochemical and functional differences among the IFN subtypes. The significance of our research is in demonstrating that IFN-α and IFN-ß both have protective roles during acute chikungunya virus (CHIKV) infection but do so by distinct mechanisms. IFN-α limits CHIKV replication and dissemination, whereas IFN-ß protects from CHIKV pathogenesis by limiting inflammation mediated by neutrophils. Our findings support the premise that the IFN subtypes have distinct biological activities in the antiviral response.


Asunto(s)
Fiebre Chikungunya/genética , Virus Chikungunya/patogenicidad , Factor 7 Regulador del Interferón/genética , Interferón-alfa/genética , Interferón beta/genética , Neutrófilos/inmunología , Animales , Anticuerpos Neutralizantes/farmacología , Huesos/inmunología , Huesos/patología , Huesos/virología , Fiebre Chikungunya/inmunología , Fiebre Chikungunya/patología , Fiebre Chikungunya/virología , Virus Chikungunya/inmunología , Femenino , Expresión Génica , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Inflamación , Factor 7 Regulador del Interferón/deficiencia , Factor 7 Regulador del Interferón/inmunología , Interferón-alfa/antagonistas & inhibidores , Interferón-alfa/deficiencia , Interferón-alfa/inmunología , Interferón beta/antagonistas & inhibidores , Interferón beta/deficiencia , Interferón beta/inmunología , Masculino , Ratones , Ratones Noqueados , Músculo Esquelético/inmunología , Músculo Esquelético/patología , Músculo Esquelético/virología , Infiltración Neutrófila , Neutrófilos/patología , Neutrófilos/virología , Tarso Animal/inmunología , Tarso Animal/patología , Tarso Animal/virología , Replicación Viral
5.
Am J Respir Cell Mol Biol ; 60(2): 144-157, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30156437

RESUMEN

The complement system provides host defense against pathogens and environmental stress. C3, the central component of complement, is present in the blood and increases in BAL fluid after injury. We recently discovered that C3 is taken up by certain cell types and cleaved intracellularly to C3a and C3b. C3a is required for CD4+ T-cell survival. These observations made us question whether complement operates at environmental interfaces, particularly in the respiratory tract. We found that airway epithelial cells (AECs, represented by both primary human tracheobronchial cells and BEAS-2B [cell line]) cultured in C3-free media were unique from other cell types in that they contained large intracellular stores of de novo synthesized C3. A fraction of this protein reduced ("storage form") but the remainder did not, consistent with it being pro-C3 ("precursor form"). These two forms of intracellular C3 were absent in CRISPR knockout-induced C3-deficient AECs and decreased with the use of C3 siRNA, indicating endogenous generation. Proinflammatory cytokine exposure increased both stored and secreted forms of C3. Furthermore, AECs took up C3 from exogenous sources, which mitigated stress-associated cell death (e.g., from oxidative stress or starvation). C3 stores were notably increased within AECs in lung tissues from individuals with different end-stage lung diseases. Thus, at-risk cells furnish C3 through biosynthesis and/or uptake to increase locally available C3 during inflammation, while intracellularly, these stores protect against certain inducers of cell death. These results establish the relevance of intracellular C3 to airway epithelial biology and suggest novel pathways for complement-mediated host protection in the airway.


Asunto(s)
Bronquios/citología , Complemento C3/metabolismo , Células Epiteliales/fisiología , Muerte Celular , Línea Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Complemento C3/genética , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Caliciformes/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/patología , Estrés Fisiológico
6.
Nature ; 494(7436): 201-6, 2013 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-23364696

RESUMEN

The lysosomal degradation pathway of autophagy has a crucial role in defence against infection, neurodegenerative disorders, cancer and ageing. Accordingly, agents that induce autophagy may have broad therapeutic applications. One approach to developing such agents is to exploit autophagy manipulation strategies used by microbial virulence factors. Here we show that a peptide, Tat-beclin 1-derived from a region of the autophagy protein, beclin 1, which binds human immunodeficiency virus (HIV)-1 Nef-is a potent inducer of autophagy, and interacts with a newly identified negative regulator of autophagy, GAPR-1 (also called GLIPR2). Tat-beclin 1 decreases the accumulation of polyglutamine expansion protein aggregates and the replication of several pathogens (including HIV-1) in vitro, and reduces mortality in mice infected with chikungunya or West Nile virus. Thus, through the characterization of a domain of beclin 1 that interacts with HIV-1 Nef, we have developed an autophagy-inducing peptide that has potential efficacy in the treatment of human diseases.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/uso terapéutico , Autofagia/efectos de los fármacos , Proteínas de la Membrana/química , Proteínas de la Membrana/uso terapéutico , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Secuencia de Aminoácidos , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Reguladoras de la Apoptosis/farmacología , Beclina-1 , Permeabilidad de la Membrana Celular , Células Cultivadas , Virus Chikungunya/efectos de los fármacos , VIH-1/efectos de los fármacos , VIH-1/metabolismo , VIH-1/fisiología , Células HeLa , Humanos , Macrófagos/citología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/farmacología , Ratones , Datos de Secuencia Molecular , Fragmentos de Péptidos/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Replicación Viral/efectos de los fármacos , Virus del Nilo Occidental/efectos de los fármacos , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo
7.
Proc Natl Acad Sci U S A ; 112(5): 1577-82, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25605921

RESUMEN

Protein modification by the ubiquitin-like protein ISG15 is an interferon (IFN) effector system, which plays a major role in antiviral defense. ISG15 modification is counteracted by the isopeptidase USP18, a major negative regulator of IFN signaling, which was also shown to exert its regulatory function in an isopeptidase-independent manner. To dissect enzymatic and nonenzymatic functions of USP18 in vivo, we generated knock-in mice (USP18(C61A/C61A)) expressing enzymatically inactive USP18. USP18(C61A/C61A) mice displayed increased levels of ISG15 conjugates, validating that USP18 is a major ISG15 isopeptidase in vivo. Unlike USP18(-/-) mice, USP18(C61A/C61A) animals did not exhibit morphological abnormalities, fatal IFN hypersensitivity, or increased lethality, clearly showing that major USP18 functions are unrelated to its protease activity. Strikingly, elevated ISGylation in USP18(C61A/C61A) mice was accompanied by increased viral resistance against vaccinia virus and influenza B virus infections. Enhanced resistance upon influenza B infection in USP18(C61A/C61A) mice was completely reversed in USP18(C61A/C61A) mice, which additionally lack ISG15, providing evidence that the observed reduction in viral titers is ISG15 dependent. These results suggest that increasing ISGylation by specific inhibition of USP18 protease activity could constitute a promising antiviral strategy with only a minimal risk of severe adverse effects.


Asunto(s)
Citocinas/metabolismo , Farmacorresistencia Viral , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Animales , Antivirales/farmacología , Células Cultivadas , Virus de la Influenza B/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ubiquitinas/metabolismo
8.
J Virol ; 89(1): 337-49, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25320315

RESUMEN

UNLABELLED: ISG15 is a diubiquitin-like modifier and one of the most rapidly induced genes upon type I interferon stimulation. Hundreds of host proteins and a number of viral proteins have been shown to be ISGylated, and understanding how these modifications affect the interferon response and virus replication has been of considerable interest. ISG15(-/-) mice exhibit increased susceptibility to viral infection, and in the case of influenza B virus and vaccinia virus, ISG15 conjugation has been shown to restrict virus replication in vivo. A number of studies have also found that ISG15 is capable of antagonizing replication of some viruses in tissue culture. However, recent findings have demonstrated that ISG15 can protect mice from Chikungunya virus infection without affecting the virus burden. In order to better understand the function of ISG15 in vivo, we characterized the pathogenesis of influenza A virus and Sendai virus in ISG15(-/-) mice. We found that ISG15 protects mice from virus induced lethality by a conjugation-dependent mechanism in both of these models. However, surprisingly, we found that ISG15 had minimal effect on virus replication and did not have an obvious role in the modulation of the acute immune response to infection. Instead, we observed an increase in the number of diseased small airways in mice lacking ISG15. This ability of ISG15 to protect mice in a conjugation-dependent, but nonantiviral, manner from respiratory virus infection represents a previously undescribed role for ISG15 and demonstrates the importance of further characterization of ISG15 in vivo. IMPORTANCE: It has previously been demonstrated that ISG15(-/-) mice are more susceptible to a number of viral infections. Since ISG15 is one of the most strongly induced genes after type I interferon stimulation, analysis of ISG15 function has largely focused on its role as an antiviral molecule during acute infection. Although a number of studies have shown that ISG15 does have a small effect on virus replication in tissue culture, few studies have confirmed this mechanism of protection in vivo. In these studies we have found that while ISG15(-/-) mice are more susceptible to influenza A virus and Sendai virus infections, ISGylation does not appear to mediate this protection through the direct inhibition of virus replication or the modulation of the acute immune response. Thus, in addition to showing a novel mode of ISG15 mediated protection from virus infection, this study demonstrates the importance of studying the role of ISG15 in vivo.


Asunto(s)
Citocinas/metabolismo , Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Respirovirus/inmunología , Virus Sendai/inmunología , Animales , Citocinas/deficiencia , Modelos Animales de Enfermedad , Femenino , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/virología , Infecciones por Respirovirus/virología , Análisis de Supervivencia , Ubiquitinas/deficiencia , Ubiquitinas/metabolismo
9.
PLoS Pathog ; 10(8): e1004350, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25166009

RESUMEN

In this study, we have identified a unique mechanism in which human cytomegalovirus (HCMV) protein pUL79 acts as an elongation factor to direct cellular RNA polymerase II for viral transcription during late times of infection. We and others previously reported that pUL79 and its homologues are required for viral transcript accumulation after viral DNA synthesis. We hypothesized that pUL79 represented a unique mechanism to regulate viral transcription at late times during HCMV infection. To test this hypothesis, we analyzed the proteome associated with pUL79 during virus infection by mass spectrometry. We identified both cellular transcriptional factors, including multiple RNA polymerase II (RNAP II) subunits, and novel viral transactivators, including pUL87 and pUL95, as protein binding partners of pUL79. Co-immunoprecipitation (co-IP) followed by immunoblot analysis confirmed the pUL79-RNAP II interaction, and this interaction was independent of any other viral proteins. Using a recombinant HCMV virus where pUL79 protein is conditionally regulated by a protein destabilization domain ddFKBP, we showed that this interaction did not alter the total levels of RNAP II or its recruitment to viral late promoters. Furthermore, pUL79 did not alter the phosphorylation profiles of the RNAP II C-terminal domain, which was critical for transcriptional regulation. Rather, a nuclear run-on assay indicated that, in the absence of pUL79, RNAP II failed to elongate and stalled on the viral DNA. pUL79-dependent RNAP II elongation was required for transcription from all three kinetic classes of viral genes (i.e. immediate-early, early, and late) at late times during virus infection. In contrast, host gene transcription during HCMV infection was independent of pUL79. In summary, we have identified a novel viral mechanism by which pUL79, and potentially other viral factors, regulates the rate of RNAP II transcription machinery on viral transcription during late stages of HCMV infection.


Asunto(s)
Infecciones por Citomegalovirus/genética , Regulación Viral de la Expresión Génica/genética , Factores de Elongación de Péptidos/genética , ARN Polimerasa II/genética , Proteínas Virales/genética , Inmunoprecipitación de Cromatina , Citomegalovirus/genética , Fibroblastos/virología , Genes Virales , Células HEK293 , Humanos , Immunoblotting , Inmunoprecipitación , Espectrometría de Masas , Transcripción Genética , Transfección
10.
J Virol ; 88(16): 9277-86, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24899198

RESUMEN

UNLABELLED: Human noroviruses (HuNoV) are the leading cause of nonbacterial gastroenteritis worldwide. Similar to HuNoV, murine noroviruses (MNV) are enteric pathogens spread via the fecal-oral route and have been isolated from numerous mouse facilities worldwide. Type I and type II interferons (IFN) restrict MNV-1 replication; however, the antiviral effectors impacting MNV-1 downstream of IFN signaling are largely unknown. Studies using dendritic cells, macrophages, and mice deficient in free and conjugated forms of interferon-stimulated gene 15 (ISG15) revealed that ISG15 conjugation contributes to protection against MNV-1 both in vitro and in vivo. ISG15 inhibited a step early in the viral life cycle upstream of viral genome transcription. Directly transfecting MNV-1 RNA into IFN-stimulated mouse embryonic fibroblasts (MEFs) and bone marrow-derived dendritic cells (BMDC) lacking ISG15 conjugates bypassed the antiviral activity of ISG15, further suggesting that ISG15 conjugates restrict the MNV-1 life cycle at the viral entry/uncoating step. These results identify ISG15 as the first type I IFN effector regulating MNV-1 infection both in vitro and in vivo and for the first time implicate the ISG15 pathway in the regulation of early stages of MNV-1 replication. IMPORTANCE: Type I IFNs are important in controlling murine norovirus 1 (MNV-1) infections; however, the proteins induced by IFNs that restrict viral growth are largely unknown. This report reveals that interferon-stimulated gene 15 (ISG15) mitigates MNV-1 replication both in vitro and in vivo. In addition, it shows that ISG15 inhibits MNV-1 replication by targeting an early step in the viral life cycle, MNV-1 entry and/or uncoating. These results identify ISG15 as the first type I IFN effector regulating MNV-1 infection both in vitro and in vivo and for the first time implicate the ISG15 pathway in the regulation of viral entry/uncoating.


Asunto(s)
Antivirales/metabolismo , Citocinas/genética , Citocinas/metabolismo , Interferón Tipo I/metabolismo , Estadios del Ciclo de Vida/genética , Norovirus/genética , Animales , Línea Celular , Células Dendríticas/metabolismo , Células Dendríticas/virología , Fibroblastos/metabolismo , Fibroblastos/virología , Genoma Viral/genética , Interferón Tipo I/genética , Macrófagos/metabolismo , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL , Transcripción Genética/genética , Ubiquitinas/genética , Ubiquitinas/metabolismo , Replicación Viral/genética
11.
J Virol ; 88(5): 2414-25, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24335291

RESUMEN

UNLABELLED: Chikungunya virus (CHIKV) is an alphavirus transmitted by mosquitoes that is known to cause severe arthritis and myositis in affected patients. The ongoing epidemic began in eastern Africa in 2004 and then spread to islands of the Indian Ocean, India, and Southeast Asia, ultimately afflicting millions. During this outbreak, more severe disease manifestations, including fatalities, have been documented. The reasons for this change in pathogenesis are multifactorial but likely include mutations that have arisen in the viral genome which could alter disease pathogenesis. To test this hypothesis, we used a murine model of CHIKV to compare the disease pathogeneses of two recombinant strains of CHIKV, the first derived from the La Reunion outbreak in 2006 (LR2006 OPY1) and the second isolated from Senegal in 1983 (37997). While the two strains exhibited similar growth in mammalian cells in vitro, we observed more severe clinical disease and pathology in mice infected with the LR2006 OPY1 strain of CHIKV, which included prolonged viremia and elevated viral titers and persistence in the muscle, resulting in devastating myonecrosis. Both CHIKV strains infected connective tissue fibroblasts of the muscle, but only the LR2006 OPY1 strain replicated within myofibers in vivo, despite similar growth of the two strains in these cell types in vitro. However, when the 37997 strain was administered directly into muscle, myofiber infection was comparable to that in LR2006 OPY1-infected mice. These results indicate that differences in the ability of the strain of CHIKV to establish infection in myofibers may contribute to the increased disease severity. IMPORTANCE: CHIKV is an emerging pathogen that causes significant morbidity. Little is known about the pathogenesis of the disease, and this study suggests that the ability of a recent epidemic strain to infect myofibers results in increased disease severity. Better understanding of how CHIKV causes disease contributes to the ultimate goal of creating therapeutics to alleviate the impact of this debilitating virus.


Asunto(s)
Infecciones por Alphavirus/virología , Virus Chikungunya/fisiología , Fibras Musculares Esqueléticas/virología , Infecciones por Alphavirus/metabolismo , Animales , Fiebre Chikungunya , Virus Chikungunya/efectos de los fármacos , Virus Chikungunya/patogenicidad , Citocinas/metabolismo , Modelos Animales de Enfermedad , Fibroblastos/virología , Genoma Viral , Mediadores de Inflamación/metabolismo , Interferón beta/biosíntesis , Interferón beta/farmacología , Ratones , Ratones Noqueados , Fibras Musculares Esqueléticas/patología , Recombinación Genética
12.
J Virol ; 88(1): 538-46, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24173234

RESUMEN

Viruses have long been studied not only for their pathology and associated disease but also as model systems for understanding cellular and immunological processes. Rodent herpesvirus Peru (RHVP) is a recently characterized rhadinovirus related to murine gammaherpesvirus 68 (MHV68) and Kaposi's sarcoma-associated herpesvirus (KSHV) that establishes acute and latent infection in laboratory mice. RHVP encodes numerous unique proteins that we hypothesize might facilitate host immune evasion during infection. We report here that open reading frame (ORF) R17 encodes a high-affinity chemokine binding protein that broadly recognizes human and murine CC and C chemokines. The interaction of R17 with chemokines is generally characterized by rapid association kinetics, and in the case of CCL3, CCL4, CCL5, CCL24, and XCL1, extremely stable complexes are formed. Functionally, R17 potently inhibited CCL2-driven chemotaxis of the human monocytic cell line THP-1, CCL3-driven chemotaxis of peripheral blood mononuclear cells, and CCL2-mediated calcium flux. Our studies also reveal that R17 binds to glycosaminoglycans (GAGs) in a process dependent upon two BBXB motifs and that chemokine and GAG binding can occur simultaneously at distinct sites. Collectively, these studies suggest that R17 may play a role in RHVP immune evasion through the targeted sabotage of chemokine-mediated immune surveillance.


Asunto(s)
Receptores de Quimiocina/genética , Rhadinovirus/genética , Animales , Calcio/metabolismo , Quimiocinas/metabolismo , Quimiotaxis de Leucocito , Células HEK293 , Humanos , Ratones , Sistemas de Lectura Abierta , Receptores de Quimiocina/metabolismo , Resonancia por Plasmón de Superficie
13.
J Virol ; 88(20): 11825-33, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25100850

RESUMEN

To combat emerging coronaviruses, developing safe and efficient platforms to evaluate viral protease activities and the efficacy of protease inhibitors is a high priority. Here, we exploit a biosafety level 2 (BSL-2) chimeric Sindbis virus system to evaluate protease activities and the efficacy of inhibitors directed against the papain-like protease (PLpro) of severe acute respiratory syndrome coronavirus (SARS-CoV), a biosafety level 3 (BSL-3) pathogen. We engineered Sindbis virus to coexpress PLpro and a substrate, murine interferon-stimulated gene 15 (ISG15), and found that PLpro mediates removal of ISG15 (deISGylation) from cellular proteins. Mutation of the catalytic cysteine residue of PLpro or addition of a PLpro inhibitor blocked deISGylation in virus-infected cells. Thus, deISGylation is a marker of PLpro activity. Infection of alpha/beta interferon receptor knockout (IFNAR(-/-)) mice with these chimeric viruses revealed that PLpro deISGylation activity removed ISG15-mediated protection during viral infection. Importantly, administration of a PLpro inhibitor protected these mice from lethal infection, demonstrating the efficacy of a coronavirus protease inhibitor in a mouse model. However, this PLpro inhibitor was not sufficient to protect the mice from lethal infection with SARS-CoV MA15, suggesting that further optimization of the delivery and stability of PLpro inhibitors is needed. We extended the chimeric-virus platform to evaluate the papain-like protease/deISGylating activity of Middle East respiratory syndrome coronavirus (MERS-CoV) to provide a small-animal model to evaluate PLpro inhibitors of this recently emerged pathogen. This platform has the potential to be universally adaptable to other viral and cellular enzymes that have deISGylating activities. Importance: Evaluating viral protease inhibitors in a small-animal model is a critical step in the path toward antiviral drug development. We modified a biosafety level 2 chimeric virus system to facilitate evaluation of inhibitors directed against highly pathogenic coronaviruses. We used this system to demonstrate the in vivo efficacy of an inhibitor of the papain-like protease of severe acute respiratory syndrome coronavirus. Furthermore, we demonstrate that the chimeric-virus system can be adapted to study the proteases of emerging human pathogens, such as Middle East respiratory syndrome coronavirus. This system provides an important tool to rapidly assess the efficacy of protease inhibitors targeting existing and emerging human pathogens, as well as other enzymes capable of removing ISG15 from cellular proteins.


Asunto(s)
Coronavirus/fisiología , Modelos Animales de Enfermedad , Papaína/metabolismo , Péptido Hidrolasas/metabolismo , Animales , Chlorocebus aethiops , Coronavirus/enzimología , Cricetinae , Ratones , Células Vero
14.
J Immunol ; 188(6): 2488-92, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22327075

RESUMEN

Bone marrow stromal Ag 2 (BST2) is a transmembrane protein that prevents virus release from infected cells. It was also reported that BST2 inhibits type I IFN production by plasmacytoid dendritic cells. To determine BST2 impact on antiviral responses in vivo, we generated BST2(-/-) mice. Following infection with a murine retrovirus, BST2(-/-) mice had slightly elevated viral loads; however, infection with other enveloped viruses revealed unexpected roles of BST2. BST2(-/-) mice showed reduced type I IFN production by plasmacytoid dendritic cells. Moreover, BST2(-/-) mice had lower viral titers in lungs following intranasal infection with vesicular stomatitis virus expressing OVA and influenza B and increased numbers of virus-specific CD8 T cells in the lungs, suggesting that BST2 may facilitate entry and/or replication of enveloped viruses and modulate priming of CD8 T cells. These findings suggest complex roles of BST2 beyond retroviral control in vivo, possibly reflecting the involvement of BST2 in endocytosis and intracellular trafficking of viruses, viral nucleic acids, and Ags.


Asunto(s)
Antígenos CD/inmunología , Interferón Tipo I/inmunología , Glicoproteínas de Membrana/inmunología , Virosis/inmunología , Animales , Antígenos CD/metabolismo , Separación Celular , Citometría de Flujo , Interferón Tipo I/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Virosis/metabolismo
15.
PLoS Pathog ; 7(10): e1002322, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22028657

RESUMEN

Chikungunya virus (CHIKV) is a re-emerging alphavirus that has caused significant disease in the Indian Ocean region since 2005. During this outbreak, in addition to fever, rash and arthritis, severe cases of CHIKV infection have been observed in infants. Challenging the notion that the innate immune response in infants is immature or defective, we demonstrate that both human infants and neonatal mice generate a robust type I interferon (IFN) response during CHIKV infection that contributes to, but is insufficient for, the complete control of infection. To characterize the mechanism by which type I IFNs control CHIKV infection, we evaluated the role of ISG15 and defined it as a central player in the host response, as neonatal mice lacking ISG15 were profoundly susceptible to CHIKV infection. Surprisingly, UbE1L⁻/⁻ mice, which lack the ISG15 E1 enzyme and therefore are unable to form ISG15 conjugates, displayed no increase in lethality following CHIKV infection, thus pointing to a non-classical role for ISG15. No differences in viral loads were observed between wild-type (WT) and ISG15⁻/⁻ mice, however, a dramatic increase in proinflammatory cytokines and chemokines was observed in ISG15⁻/⁻ mice, suggesting that the innate immune response to CHIKV contributes to their lethality. This study provides new insight into the control of CHIKV infection, and establishes a new model for how ISG15 functions as an immunomodulatory molecule in the blunting of potentially pathologic levels of innate effector molecules during the host response to viral infection.


Asunto(s)
Infecciones por Alphavirus/inmunología , Virus Chikungunya/fisiología , Citocinas/inmunología , Interferón Tipo I/inmunología , Enzimas Activadoras de Ubiquitina/inmunología , Ubiquitinas/inmunología , Infecciones por Alphavirus/diagnóstico , Infecciones por Alphavirus/metabolismo , Animales , Animales Recién Nacidos , Fiebre Chikungunya , Virus Chikungunya/patogenicidad , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Interferón Tipo I/metabolismo , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Recombinación Genética , Estudios Retrospectivos , Enzimas Activadoras de Ubiquitina/metabolismo , Ubiquitinas/metabolismo
16.
Cell Host Microbe ; 30(1): 8-9, 2022 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-35026136

RESUMEN

Effector-triggered immunity involves "guarded" host processes that, when perturbed by pathogen factors, prompt a secondary response. A recent study published in Nature by Gaidt et al. demonstrates that MORC3 serves as both the guard and the guarded antiviral host factor-creating a "heads, I win; tails, you lose!" scenario.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Adenosina Trifosfatasas/genética , Antivirales , Proteínas de Unión al ADN/genética , Interacciones Microbiota-Huesped/inmunología , Humanos , Ubiquitina-Proteína Ligasas , Factores de Virulencia/inmunología
17.
J Virol ; 84(10): 5423-30, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20219937

RESUMEN

Interferon-stimulated expression and conjugation of the ubiquitin-like modifier ISG15 restricts replication of several viruses. Here, we established complete E1-activating, E2-conjugating, and E3 ligase-dependent expression systems for assaying both human and mouse ISGylation. We confirm that human HerC5, but not human HerC6, has ISG15 E3 ligase activity and identify mouse HerC6 as a bona fide ISG15 E3 ligase. Furthermore, we demonstrate that influenza B virus NS1 protein potently antagonizes human but not mouse ISGylation, a property dependent on B/NS1 binding the N-terminal domain of human but not mouse ISG15. Using chimeric human/mouse ISG15 constructs, we show that the B/NS1:ISG15 interaction is both necessary and sufficient to inhibit ISGylation regardless of the ligation machinery used. Inability to block ISGylation in certain species may contribute to limiting influenza B virus host range.


Asunto(s)
Citocinas/antagonistas & inhibidores , Virus de la Influenza B/inmunología , Virus de la Influenza B/patogenicidad , Ubiquitinas/antagonistas & inhibidores , Proteínas no Estructurales Virales/metabolismo , Factores de Virulencia/metabolismo , Secuencia de Aminoácidos , Animales , Humanos , Péptidos y Proteínas de Señalización Intracelular/inmunología , Ratones , Datos de Secuencia Molecular , Unión Proteica , Alineación de Secuencia , Ubiquitina-Proteína Ligasas/inmunología
18.
iScience ; 24(10): 103213, 2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34632326

RESUMEN

The emergence of SARS-CoV-2 has led to a global health crisis that, in addition to vaccines and immunomodulatory therapies, calls for the identification of antiviral therapeutics. The papain-like protease (PLpro) activity of nsp3 is an attractive drug target as it is essential for viral polyprotein cleavage and for deconjugation of ISG15, an antiviral ubiquitin-like protein. We show here that 6-Thioguanine (6-TG), an orally available and widely available generic drug, inhibits SARS-CoV-2 replication in Vero-E6 cells with an EC50 of approximately 2 µM. 6-TG also inhibited PLpro-catalyzed polyprotein cleavage and de-ISGylation in cells and inhibited proteolytic activity of the purified PLpro domain in vitro. We therefore propose that 6-TG is a direct-acting antiviral that could potentially be repurposed and incorporated into the set of treatment and prevention options for COVID-19.

19.
J Virol ; 83(2): 1147-51, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19004958

RESUMEN

ISG15 functions as a critical antiviral molecule against influenza virus, with infection inducing both the conjugation of ISG15 to target proteins and production of free ISG15. Here, we report that mice lacking the ISG15 E1 enzyme UbE1L fail to form ISG15 conjugates. Both UbE1L(-/-) and ISG15(-/-) mice display increased susceptibility to influenza B virus infection, including non-mouse-adapted strains. Finally, we demonstrate that ISG15 controls influenza B virus infection through its action within radioresistant stromal cells and not bone marrow-derived cells. Thus, the conjugation of ISG15 to target proteins within stromal cells is critical to its activity against influenza virus.


Asunto(s)
Citocinas/deficiencia , Predisposición Genética a la Enfermedad , Virus de la Influenza B/inmunología , Infecciones por Orthomyxoviridae/inmunología , Enzimas Activadoras de Ubiquitina/deficiencia , Animales , Pulmón/virología , Ratones , Análisis de Supervivencia , Ubiquitinas/deficiencia
20.
J Virol ; 83(4): 1602-10, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19073728

RESUMEN

Interferon (IFN)-stimulated gene 15 (ISG15) is a ubiquitin-like molecule that conjugates to target proteins via a C-terminal LRLRGG motif and has antiviral function in vivo. We used structural modeling to predict human ISG15 (hISG15) residues important for interacting with its E1 enzyme, UbE1L. Kinetic analysis revealed that mutation of arginine 153 to alanine (R153A) ablated hISG15-hUbE1L binding and transthiolation of UbcH8. Mutation of other predicted UbE1L-interacting residues had minimal effects on the transfer of ISG15 from UbE1L to UbcH8. The capacity of hISG15 R153A to form protein conjugates in 293T cells was markedly diminished. Mutation of the homologous residue in mouse ISG15 (mISG15), arginine 151, to alanine (R151A) also attenuated protein ISGylation following transfection into 293T cells. We assessed the role of ISG15-UbE1L interactions in control of virus infection by constructing double subgenomic Sindbis viruses that expressed the mISG15 R151A mutant. While expression of mISG15 protected alpha/beta-IFN-receptor-deficient (IFN-alphabetaR(-/-)) mice from lethality following Sindbis virus infection, expression of mISG15 R151A conferred no survival benefit. The R151A mutation also attenuated ISG15's ability to decrease Sindbis virus replication in IFN-alphabetaR(-/-) mice or prolong survival of ISG15(-/-) mice. The importance of UbE1L was confirmed by demonstrating that mice lacking this ISG15 E1 enzyme were highly susceptible to Sindbis virus infection. Together, these data support a role for protein conjugation in the antiviral effects of ISG15.


Asunto(s)
Infecciones por Alphavirus/inmunología , Citocinas/metabolismo , Mapeo de Interacción de Proteínas , Virus Sindbis/inmunología , Enzimas Activadoras de Ubiquitina/metabolismo , Ubiquitinas/metabolismo , Sustitución de Aminoácidos/genética , Animales , Línea Celular , Citocinas/genética , Humanos , Masculino , Ratones , Mutagénesis Sitio-Dirigida , Mutación Missense , Unión Proteica , Análisis de Supervivencia , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA