Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 286
Filtrar
1.
Nat Commun ; 15(1): 4153, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755212

RESUMEN

Viral myocarditis, an inflammatory disease of the heart, causes significant morbidity and mortality. Type I interferon (IFN)-mediated antiviral responses protect against myocarditis, but the mechanisms are poorly understood. We previously identified A Disintegrin And Metalloproteinase domain 9 (ADAM9) as an important factor in viral pathogenesis. ADAM9 is implicated in a range of human diseases, including inflammatory diseases; however, its role in viral infection is unknown. Here, we demonstrate that mice lacking ADAM9 are more susceptible to encephalomyocarditis virus (EMCV)-induced death and fail to mount a characteristic type I IFN response. This defect in type I IFN induction is specific to positive-sense, single-stranded RNA (+ ssRNA) viruses and involves melanoma differentiation-associated protein 5 (MDA5)-a key receptor for +ssRNA viruses. Mechanistically, ADAM9 binds to MDA5 and promotes its oligomerization and thereby downstream mitochondrial antiviral-signaling protein (MAVS) activation in response to EMCV RNA stimulation. Our findings identify a role for ADAM9 in the innate antiviral response, specifically MDA5-mediated IFN production, which protects against virus-induced cardiac damage, and provide a potential therapeutic target for treatment of viral myocarditis.


Asunto(s)
Proteínas ADAM , Infecciones por Cardiovirus , Virus de la Encefalomiocarditis , Inmunidad Innata , Interferón Tipo I , Helicasa Inducida por Interferón IFIH1 , Proteínas de la Membrana , Ratones Noqueados , Miocarditis , Animales , Virus de la Encefalomiocarditis/inmunología , Helicasa Inducida por Interferón IFIH1/metabolismo , Helicasa Inducida por Interferón IFIH1/genética , Helicasa Inducida por Interferón IFIH1/inmunología , Interferón Tipo I/metabolismo , Interferón Tipo I/inmunología , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/virología , Proteínas ADAM/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/inmunología , Ratones , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Miocarditis/inmunología , Miocarditis/virología , Humanos , Ratones Endogámicos C57BL , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Transducción de Señal/inmunología , Masculino , Células HEK293
2.
mBio ; 12(3)2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33975932

RESUMEN

Intestinal microbiomes are of vital importance in antagonizing systemic viral infection. However, very little literature has shown whether commensal bacteria play a crucial role in protecting against enteric virus systemic infection from the aspect of modulating host innate immunity. In the present study, we utilized an enteric virus, encephalomyocarditis virus (EMCV), to inoculate mice treated with phosphate-buffered saline (PBS) or given an antibiotic cocktail (Abx) orally or intraperitoneally to examine the impact of microbiota depletion on virulence and viral replication in vivo Microbiota depletion exacerbated the mortality, neuropathogenesis, viremia, and viral burden in brains following EMCV infection. Furthermore, Abx-treated mice exhibited severely diminished mononuclear phagocyte activation and impaired type I interferon (IFN) production and expression of IFN-stimulated genes (ISG) in peripheral blood mononuclear cells (PBMC), spleens, and brains. With the help of fecal bacterial 16S rRNA sequencing of PBS- and Abx-treated mice, we identified a single commensal bacterium, Blautia coccoides, that can restore mononuclear phagocyte- and IFNAR (IFN-α/ß receptor)-dependent type I IFN responses to restrict systemic enteric virus infection. These findings may provide insight into the development of novel therapeutics for preventing enteric virus infection or possibly alleviating clinical diseases by activating host systemic innate immune responses via respective probiotic treatment using B. coccoidesIMPORTANCE While cumulative data indicate that indigenous commensal bacteria can facilitate enteric virus infection, little is known regarding whether intestinal microbes have a protective role in antagonizing enteric systemic infection by modulating host innate immunity. Although accumulating literature has pointed out that the microbiota has a fundamental impact on host systemic antiviral innate immune responses mediated by type I interferon (IFN), only a few specific commensal bacteria species have been revealed to be capable of regulating IFN-I and ISG expression, not to mention the underlying mechanisms. Thus, it is important to understand the cross talk between microbiota and host anti-enteric virus innate immune responses and characterize the specific bacterial species that possess protective functions. Our study demonstrates how fundamental innate immune mediators such as mononuclear phagocytes and type I IFN are regulated by commensal bacteria to antagonize enteric virus systemic infection. In particular, we have identified a novel commensal bacterium, Blautia coccoides, that can restrict enteric virus replication and neuropathogenesis by activating IFN-I and ISG responses in mononuclear phagocytes via an IFNAR- and STAT1-mediated signaling pathway.


Asunto(s)
Infecciones por Cardiovirus/prevención & control , Virus de la Encefalomiocarditis/inmunología , Microbioma Gastrointestinal/inmunología , Inmunidad Innata , Interferón Tipo I/inmunología , Viremia/inmunología , Viremia/prevención & control , Animales , Antibacterianos/administración & dosificación , Infecciones por Cardiovirus/inmunología , Clostridiales/inmunología , Virus de la Encefalomiocarditis/patogenicidad , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Simbiosis/inmunología , Replicación Viral/inmunología
3.
Nat Commun ; 12(1): 2970, 2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-34016972

RESUMEN

Activation of MAVS, an adaptor molecule in Rig-I-like receptor (RLR) signaling, is indispensable for antiviral immunity, yet the molecular mechanisms modulating MAVS activation are not completely understood. Ubiquitination has a central function in regulating the activity of MAVS. Here, we demonstrate that a mitochondria-localized deubiquitinase USP18 specifically interacts with MAVS, promotes K63-linked polyubiquitination and subsequent aggregation of MAVS. USP18 upregulates the expression and production of type I interferon following infection with Sendai virus (SeV) or Encephalomyocarditis virus (EMCV). Mice with a deficiency of USP18 are more susceptible to RNA virus infection. USP18 functions as a scaffold protein to facilitate the re-localization of TRIM31 and enhances the interaction between TRIM31 and MAVS in mitochondria. Our results indicate that USP18 functions as a post-translational modulator of MAVS-mediated antiviral signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Infecciones por Cardiovirus/inmunología , Infecciones por Respirovirus/inmunología , Ubiquitina Tiolesterasa/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/aislamiento & purificación , Animales , Infecciones por Cardiovirus/virología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Virus de la Encefalomiocarditis/inmunología , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Inmunidad Innata , Interferón Tipo I/metabolismo , Lisina/metabolismo , Masculino , Ratones , Ratones Noqueados , Procesamiento Proteico-Postraduccional/inmunología , Células RAW 264.7 , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Infecciones por Respirovirus/virología , Virus Sendai/inmunología , Transducción de Señal/inmunología , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/aislamiento & purificación , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/inmunología
4.
J Gen Virol ; 102(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33533710

RESUMEN

Encephalomyocarditis virus (EMCV) infects a wide range of hosts and can cause encephalitis, myocarditis, reproductive disorders and diabetes mellitus in selected mammalian species. As for humans, EMCV infection seems to occur by the contact with animals and can cause febrile illnesses in some infected patients. Here we isolated EMCV strain ZM12/14 from a natal multimammate mouse (Mastomys natalensis: M. natalensis) in Zambia. Pairwise sequence similarity of the ZM12/14 P1 region consisting of antigenic capsid proteins showed the highest similarity of nucleotide (80.7 %) and amino acid (96.2%) sequence with EMCV serotype 1 (EMCV-1). Phylogenetic analysis revealed that ZM12/14 clustered into EMCV-1 at the P1 and P3 regions but segregated from known EMCV strains at the P2 region, suggesting a unique evolutionary history. Reverse transcription PCR (RT-PCR) screening and neutralizing antibody assays for EMCV were performed using collected tissues and serum from various rodents (n=179) captured in different areas in Zambia. We detected the EMCV genome in 19 M. natalensis (19/179=10.6 %) and neutralizing antibody for EMCV in 33 M. natalensis (33/179=18.4 %). However, we did not detect either the genome or neutralizing antibody in other rodent species. High neutralizing antibody litres (≧320) were observed in both RT-PCR-negative and -positive animals. Inoculation of ZM12/14 caused asymptomatic persistent infection in BALB/c mice with high antibody titres and high viral loads in some organs, consistent with the above epidemiological results. This study is the first report of the isolation of EMCV in Zambia, suggesting that M. natalensis may play a role as a natural reservoir of infection.


Asunto(s)
Infecciones por Cardiovirus/veterinaria , Reservorios de Enfermedades/virología , Virus de la Encefalomiocarditis/aislamiento & purificación , Murinae/virología , Enfermedades de los Roedores/virología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Infecciones por Cardiovirus/epidemiología , Infecciones por Cardiovirus/virología , Virus de la Encefalomiocarditis/genética , Virus de la Encefalomiocarditis/inmunología , Virus de la Encefalomiocarditis/patogenicidad , Evolución Molecular , Genoma Viral , Ratones Endogámicos BALB C , Filogenia , Prevalencia , Enfermedades de los Roedores/epidemiología , Musarañas/virología , Zambia/epidemiología
5.
Nat Commun ; 11(1): 5536, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-33139700

RESUMEN

MAVS and MITA are essential adaptor proteins mediating innate antiviral immune responses against RNA and DNA viruses, respectively. Here we show that RNF115 plays dual roles in response to RNA or DNA virus infections by catalyzing distinct types of ubiquitination of MAVS and MITA at different phases of viral infection. RNF115 constitutively interacts with and induces K48-linked ubiquitination and proteasomal degradation of homeostatic MAVS in uninfected cells, whereas associates with and catalyzes K63-linked ubiquitination of MITA after HSV-1 infection. Consistently, the protein levels of MAVS are substantially increased in Rnf115-/- organs or cells without viral infection, and HSV-1-induced aggregation of MITA is impaired in Rnf115-/- cells compared to the wild-type counterparts. Consequently, the Rnf115-/- mice exhibit hypo- and hyper-sensitivity to EMCV and HSV-1 infection, respectively. These findings highlight dual regulation of cellular antiviral responses by RNF115-mediated ubiquitination of MAVS and MITA and contribute to our understanding of innate immune signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Infecciones por Cardiovirus/inmunología , Herpes Simple/inmunología , Inmunidad Innata , Proteínas de la Membrana/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Infecciones por Cardiovirus/patología , Infecciones por Cardiovirus/virología , Modelos Animales de Enfermedad , Virus de la Encefalomiocarditis/inmunología , Femenino , Células HEK293 , Herpes Simple/patología , Herpes Simple/virología , Herpesvirus Humano 1/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Lisina/metabolismo , Macrófagos/inmunología , Macrófagos/virología , Masculino , Ratones , Ratones Noqueados , Cultivo Primario de Células , Agregado de Proteínas/inmunología , ARN Interferente Pequeño/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/fisiología , Ubiquitinación/inmunología
6.
J Immunol ; 205(1): 168-180, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32393512

RESUMEN

The RNA helicase DDX39A plays an important role in the RNA splicing/export process. In our study, human DDX39A facilitated RNA virus escape from innate immunity to promote virus proliferation by trapping TRAF3, TRAF6, and MAVS mRNAs in the HEK293T cell nucleus. DDX39A was a target for SUMOylation. SUMO1, 2, and 3 modifications were found on immunoprecipitated DDX39A. However, only the SUMO1 modification decreased in vesicular stomatitis virus-infected HEK293T cells. Further studies have found that viral infection reduced SUMO1 modification of DDX39A and enhanced its ability to bind innate immunity-associated mRNAs by regulating the abundance of RanBP2 with SUMO1 E3 ligase activity. RanBP2 acted as an E3 SUMO ligase of DDX39A, which enhanced SUMO1 modification of DDX39A and attenuated its ability to bind RNA. This work described that specific mRNAs encoding antiviral signaling components were bound and sequestered in the nucleus by DDX39A to limit their expression, which proposed a new protein SUMOylation model to regulate innate immunity in viral infection.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética , Infecciones por Virus ARN/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Núcleo Celular/metabolismo , Chlorocebus aethiops , ARN Helicasas DEAD-box/genética , Regulación hacia Abajo , Virus de la Encefalomiocarditis/genética , Virus de la Encefalomiocarditis/inmunología , Técnicas de Inactivación de Genes , Células HEK293 , Células HeLa , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/inmunología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Chaperonas Moleculares/genética , Proteínas de Complejo Poro Nuclear/genética , Infecciones por Virus ARN/virología , ARN Mensajero/metabolismo , ARN Viral/inmunología , ARN Viral/metabolismo , Proteína SUMO-1/metabolismo , Virus Sendai/genética , Virus Sendai/inmunología , Sumoilación/inmunología , Factor 3 Asociado a Receptor de TNF/genética , Transcripción Genética/inmunología , Células Vero , Vesiculovirus/genética , Vesiculovirus/inmunología , Replicación Viral/inmunología
7.
Biochem Biophys Res Commun ; 527(1): 1-7, 2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32446351

RESUMEN

Bats are potential natural hosts of Encephalomyocarditis virus (EMCV) and Japanese encephalitis virus (JEV). Bats appear to have some unique features in their innate immune system that inhibit viral replication causing limited clinical symptoms, and thus, contributing to the virus spill over to humans. Here, kidney epithelial cell lines derived from four bat species (Pteropus dasymallus, Rousettus leschenaultii, Rhinolophus ferrumequinum, and Miniopterus fuliginosus) and two non-bat species (Homo sapiens and Mesocricetus auratus) were infected with EMCV and JEV. The replication of EMCV and JEV was lower in the bat cell lines derived from R. leschenaultii, R. ferrumequinum, and M. fuliginosus with a higher expression level of pattern recognition receptors (PRRs) (TLR3, RIG-I, and MDA5) and interferon-beta (IFN-ß) than that in the non-bat cell lines and a bat cell line derived from P. dasymallus. The knockdown of TLR3, RIG-I, and MDA5 in Rhinolophus bat cell line using antisense RNA oligonucleotide led to decrease IFN-ß expression and increased viral replication. These results suggest that TLR3, RIG-I, and MDA5 are important for antiviral response against EMCV and JEV in Rhinolophus bats.


Asunto(s)
Infecciones por Cardiovirus/veterinaria , Quirópteros/virología , Virus de la Encefalitis Japonesa (Especie)/inmunología , Encefalitis Japonesa/veterinaria , Virus de la Encefalomiocarditis/inmunología , Interferón beta/inmunología , Receptores de Reconocimiento de Patrones/inmunología , Animales , Enfermedades de las Aves/inmunología , Enfermedades de las Aves/virología , Infecciones por Cardiovirus/inmunología , Línea Celular , Quirópteros/inmunología , Encefalitis Japonesa/inmunología , Humanos , Inmunidad Innata
8.
Nat Commun ; 10(1): 4624, 2019 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-31604929

RESUMEN

Cytosolic mitochondrial DNA (mtDNA) activates cGAS-mediated antiviral immune responses, but the mechanism by which RNA viruses stimulate mtDNA release remains unknown. Here we show that viroporin activity of influenza virus M2 or encephalomyocarditis virus (EMCV) 2B protein triggers translocation of mtDNA into the cytosol in a MAVS-dependent manner. Although influenza virus-induced cytosolic mtDNA stimulates cGAS- and DDX41-dependent innate immune responses, the nonstructural protein 1 (NS1) of influenza virus associates with mtDNA to evade the STING-dependent antiviral immunity. The STING-dependent antiviral signaling is amplified in neighboring cells through gap junctions. In addition, we find that STING-dependent recognition of influenza virus is essential for limiting virus replication in vivo. Our results show a mechanism by which influenza virus stimulates mtDNA release and highlight the importance of DNA sensing pathway in limiting influenza virus replication.


Asunto(s)
ADN Mitocondrial/inmunología , Virus de la Influenza A/inmunología , Proteínas de la Matriz Viral/inmunología , Proteínas Virales/inmunología , Transporte Activo de Núcleo Celular , Núcleo Celular/metabolismo , Citosol/metabolismo , ADN Mitocondrial/metabolismo , Virus de la Encefalomiocarditis/inmunología , Células HEK293 , Interacciones Microbiota-Huesped , Humanos , Inmunidad Innata , Transducción de Señal , Proteínas Virales/metabolismo
9.
Antiviral Res ; 161: 70-84, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30312637

RESUMEN

Encephalomyocarditis virus (EMCV) is one of the most important picornavirus. It infects many mammalian species and causes encephalitis, myocarditis, neurologic diseases, diabetes and reproductive disorders in pigs. And it evolves mechanisms for escaping innate immune responses. But the viral pathogenesis has not been understood completely. In this study, we firstly found that EMCV protein 2C is a strong IFN-ß antagonist that interacts with MDA5 to inhibit induction of the IFN-ß signal pathway. The mutations in amino acid residue V26 of 2C decrease the inhibition of IFN-ß promoter activity and lost the ability to interact with MDA5, compared with wild type 2C protein. The rescued viruses with mutations in 2C (rV26A and rK25-3A) induced significantly higher IFN-ß mRNA and protein levels in PK-15, HEK-293A and N2a cells, compared to wild type EMCV and the repaired viruses rV26A(R) and rK25-3A(R). These data indicate that the amino acid residue V26 of EMCV 2C plays important roles in inhibiting type I IFN production by interacting with MDA5.


Asunto(s)
Proteínas Portadoras/genética , Virus de la Encefalomiocarditis/genética , Helicasa Inducida por Interferón IFIH1/inmunología , Interferón beta/antagonistas & inhibidores , Transducción de Señal , Proteínas no Estructurales Virales/genética , Virus de la Encefalomiocarditis/inmunología , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Mutación , Regiones Promotoras Genéticas , ARN Viral/genética
10.
Virology ; 528: 7-18, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30550976

RESUMEN

The contributions of RIG-I and MDA5 receptors to sensing viruses of the Picornaviridae family were investigated. The picornaviruses encephalomyocarditis virus (EMCV) and Coxsackievirus B3 (CVB3) are detected by both MDA5 and RIG-I in bone marrow derived macrophages. In macrophages from wild type mice, type I IFN is produced early after infection; IFNß synthesis is reduced in the absence of each sensor, while IFNα production is reduced in the absence of MDA5. EMCV and CVB3 do not replicate in murine macrophages, and their detection is different in murine embryonic fibroblasts (MEFs), in which the viruses replicate to high titers. In MEFs RIG-I was essential for the expression of type I IFNs but contributes to increased yields of CVB3, while MDA5 inhibited CVB3 replication but in an IFN independent manner. These observations demonstrate functional redundancy within the innate immune response to picornaviruses.


Asunto(s)
Proteína 58 DEAD Box/inmunología , Virus de la Encefalomiocarditis/inmunología , Enterovirus Humano B/inmunología , Inmunidad Innata , Helicasa Inducida por Interferón IFIH1/inmunología , Animales , Fibroblastos/inmunología , Fibroblastos/virología , Interacciones Huésped-Patógeno/inmunología , Interferón Tipo I/inmunología , Macrófagos/inmunología , Macrófagos/virología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos ICR , Ratones Noqueados , Transducción de Señal , Replicación Viral
11.
Int Immunopharmacol ; 62: 1-6, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29960044

RESUMEN

Because PEGylated molecules exhibit different physicochemical properties from those of the parent molecules, PEGylated interferonß-1a (pegIFNß-1a) may be able to be used with retained bioactivity in Multiple Sclerosis (MS) patients who have previously developed neutralizing antibodies (NABs) to recombinant interferonß (rIFNß). Hence, the objective of the present study was to test whether pegIFNß-1a is less antigenic for NABs in vitro than rIFNß. Two in vitro assays were used to quantitate NABs in 115 sera obtained from MS patients included in the INSIGHT study: the cytopathic effect (CPE) assay, and the MxA protein induction assay. NABs cross-reactivity was assessed by comparing dilutions of serum with fixed doses of rIFNß-1a Avonex® and pegIFNß-1a Plegridy®. NABs were shown to cross-react in both assays. The y-intercept (c), the slope of the line of agreement (b), the Pearson coefficients as well as the Bland-Altman analysis, indicated that there is good level of agreement between NAB titers against the two IFNß-1a formulations, with both the CPE (c = 0.1044 ±â€¯0.1305; b = 0.8438 ±â€¯0.06654; r2 = 0.587; bias index ±â€¯SD = -0.01702 ±â€¯0.6334), and the MxA protein induction (c = 0.08246 ±â€¯0.1229; b = 0.8878 ±â€¯0.06613; r2 = 0.615; bias index ±â€¯SD = -0.09965 ±â€¯0.6467) assays. Until further in vivo evidence is established, clinicians should consider the current in vitro data demonstrating NAB cross-reactivity between pegIFNß-1a and rIFNß when discussing new treatment options with MS patients.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Interferón beta/inmunología , Esclerosis Múltiple/sangre , Proteínas Recombinantes/inmunología , Células A549 , Bioensayo , Reacciones Cruzadas , Efecto Citopatogénico Viral , Virus de la Encefalomiocarditis/inmunología , Humanos , Esclerosis Múltiple/inmunología , Proteínas de Resistencia a Mixovirus/biosíntesis , Pruebas de Neutralización , Polietilenglicoles
12.
PLoS Pathog ; 14(2): e1006886, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29462185

RESUMEN

Melanoma differentiation-associated gene-5 (MDA5) recognizes distinct subsets of viruses including Encephalomyocarditis virus (EMCV) of picornavirus family, but the molecular mechanisms underlying the specificity of the viral recognition of MDA5 in immune cells remain obscure. DHX29 is an RNA helicase required for the translation of 5' structured mRNA of host and many picornaviruses (such as EMCV). We identify that DXH29 as a key RNA co-sensor, plays a significant role for specific recognition and triggering anti-EMCV immunity. We have observed that DHX29 regulates MDA5-, but not RIG-I-, mediated type I interferon signaling by preferentially interacting with structured RNAs and specifically with MDA5 for enhancing MDA5-dsRNA binding affinity. Overall, our results identify a critical role for DHX29 in innate immune response and provide molecular insights into the mechanisms by which DHX29 recognizes 5' structured EMCV RNA and interacts with MDA5 for potent type I interferon signaling and antiviral immunity.


Asunto(s)
Infecciones por Cardiovirus/inmunología , Virus de la Encefalomiocarditis/inmunología , Inmunidad Innata/genética , Helicasa Inducida por Interferón IFIH1/fisiología , ARN Helicasas/fisiología , ARN Viral/inmunología , Animales , Infecciones por Cardiovirus/genética , Células Cultivadas , Chlorocebus aethiops , Virus de la Encefalomiocarditis/genética , Células HEK293 , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , ARN Helicasas/genética , ARN Viral/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Células Vero
13.
J Med Virol ; 90(7): 1184-1191, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29476627

RESUMEN

Encephalomyocarditis virus (EMCV) is as a potential zoonotic agent with a wide host range. Here, applying gene splicing by overlap extension PCR (SOE-PCR), we describe a simple method for producing single-chain variable fragment (scFv) antibody against EMCV that configurates in the orientation of VH-(GGGGS)4 -VL. DNA template was resverse transcribed by total RNA that derived from hyperimmunized antibody positive mice spleen after inoculation inactivated EMCV-PV21 as antigen. Using the degenerate primers designed for the variable regions of IgG of murine antibody, the 417 bp of gene encoding VH-linker (VHL) and 360 bp of gene encoding linker-VL (LVL) of the anti-EMCV was individually amplified from DNA template by PCR, repectively. The 762 bp gene encoding anti-EMCV scFv was constructed by SOE-PCR when the mixed VHL and LVL genes were used as the template. The amplified gene subcloned into pGEX-6P1 to yield pGEX-6P1/EMCV-scFv. Recombinant vector transformed into the Escherichia coli BL21 (DE3) and a 53 KDa GST-scFv fusion protein was obtained by SDS-PAGE electrophoresis. Animal experiment results showed that the pretective rate of mice in group A which challenged 500 µL 104 TCID50 EMCV per mouse for 7 d post-inoculation scFv 3 d (0.5 mg purified recombinant scFv per mouse) was 91.67% (11/12). The serum anti-EMCV antibody titer in group A mice was most significantly higher than that in positive control mouse (P < 0.01), coversely the serum relative mRNA copies were significantly lower than that in positive control mouse (P < 0.05). These findings indicated that recombinant anti-EMCV scFv has remarkable anti-EMCV effect in mice.


Asunto(s)
Anticuerpos Antivirales/inmunología , Virus de la Encefalomiocarditis/inmunología , Anticuerpos de Cadena Única/inmunología , Animales , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/genética , Anticuerpos Antivirales/aislamiento & purificación , Infecciones por Cardiovirus/prevención & control , Clonación Molecular , Modelos Animales de Enfermedad , Escherichia coli/genética , Escherichia coli/metabolismo , Femenino , Expresión Génica , Ratones Endogámicos BALB C , Reacción en Cadena de la Polimerasa , Anticuerpos de Cadena Única/biosíntesis , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/aislamiento & purificación , Resultado del Tratamiento
14.
Biochem Biophys Res Commun ; 494(1-2): 227-233, 2017 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-29032202

RESUMEN

Laboratory of genetics and physiology 2 (LGP2) and melanoma differentiation-associated gene 5 (MDA5) cooperatively detect viral RNA in the cytoplasm of Cardiovirus-infected cells and activate innate immune responses. Here, we evaluated whether the double-stranded RNA-binding protein PACT plays a role in this anti-viral response to further elucidate the mechanism. Immunoprecipitation experiments demonstrated that PACT interacts with LGP2 and that this interaction is enhanced by encephalomyocarditis virus (EMCV) infection. In vitro interaction analyses using purified recombinant proteins confirmed that the single-stranded Theiler's murine encephalitis virus genome enhanced the interaction between LGP2 and PACT. Small interfering RNA knockdown experiments further indicated that PACT is required for Cardiovirus-triggered interferon responses. To support this functional interaction with LGP2, overexpressed PACT was shown to enhance EMCV-triggered interferon promoter activity only when LGP2 and MDA5 were co-expressed but not when MDA5 is expressed alone. Together, our findings indicate a possible role of PACT in regulating the Cardiovirus-triggered immune responses mediated by MDA5 and LGP2, which opens the door to novel therapeutic strategies in interferon-related autoimmune diseases and cancer.


Asunto(s)
Infecciones por Cardiovirus/inmunología , Virus de la Encefalomiocarditis , Helicasa Inducida por Interferón IFIH1/inmunología , ARN Helicasas/inmunología , Proteínas de Unión al ARN/inmunología , Animales , Infecciones por Cardiovirus/genética , Infecciones por Cardiovirus/virología , Línea Celular , Chlorocebus aethiops , ARN Helicasas DEAD-box/antagonistas & inhibidores , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/inmunología , Virus de la Encefalomiocarditis/genética , Virus de la Encefalomiocarditis/inmunología , Técnicas de Silenciamiento del Gen , Células HEK293 , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/genética , Helicasa Inducida por Interferón IFIH1/genética , Interferón beta/genética , Ratones , Regiones Promotoras Genéticas , ARN Helicasas/genética , ARN Interferente Pequeño/genética , ARN Viral/genética , ARN Viral/inmunología , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Ribonucleasa III/antagonistas & inhibidores , Ribonucleasa III/genética , Ribonucleasa III/inmunología , Células Vero
15.
Immunity ; 47(3): 498-509.e6, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28916264

RESUMEN

Double-stranded RNA (dsRNA) is a common by-product of viral infections and acts as a potent trigger of antiviral immunity. In the nematode C. elegans, sid-1 encodes a dsRNA transporter that is highly conserved throughout animal evolution, but the physiological role of SID-1 and its orthologs remains unclear. Here, we show that the mammalian SID-1 ortholog, SIDT2, is required to transport internalized extracellular dsRNA from endocytic compartments into the cytoplasm for immune activation. Sidt2-deficient mice exposed to extracellular dsRNA, encephalomyocarditis virus (EMCV), and herpes simplex virus 1 (HSV-1) show impaired production of antiviral cytokines and-in the case of EMCV and HSV-1-reduced survival. Thus, SIDT2 has retained the dsRNA transport activity of its C. elegans ortholog, and this transport is important for antiviral immunity.


Asunto(s)
Inmunidad Innata , Proteínas de la Membrana/metabolismo , Transporte de ARN , ARN Bicatenario/inmunología , ARN Bicatenario/metabolismo , Animales , Infecciones por Cardiovirus/genética , Infecciones por Cardiovirus/inmunología , Línea Celular , Citoplasma , Proteína 58 DEAD Box/metabolismo , Modelos Animales de Enfermedad , Virus de la Encefalomiocarditis/genética , Virus de la Encefalomiocarditis/inmunología , Endosomas/metabolismo , Femenino , Expresión Génica , Técnicas de Inactivación de Genes , Herpes Simple/genética , Herpes Simple/inmunología , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Lisosomas/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteínas de Transporte de Nucleótidos , Unión Proteica , Transporte de Proteínas , ARN Viral/genética , ARN Viral/metabolismo , Transducción de Señal , Receptor Toll-Like 3/metabolismo
16.
Nat Immunol ; 18(7): 744-752, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28553952

RESUMEN

The single-nucleotide polymorphism rs1990760 in the gene encoding the cytosolic viral sensor IFIH1 results in an amino-acid change (A946T; IFIH1T946) that is associated with multiple autoimmune diseases. The effect of this polymorphism on both viral sensing and autoimmune pathogenesis remains poorly understood. Here we found that human peripheral blood mononuclear cells (PBMCs) and cell lines expressing the risk variant IFIH1T946 exhibited heightened basal and ligand-triggered production of type I interferons. Consistent with those findings, mice with a knock-in mutation encoding IFIH1T946 displayed enhanced basal expression of type I interferons, survived a lethal viral challenge and exhibited increased penetrance in autoimmune models, including a combinatorial effect with other risk variants. Furthermore, IFIH1T946 mice manifested an embryonic survival defect consistent with enhanced responsiveness to RNA self ligands. Together our data support a model wherein the production of type I interferons driven by an autoimmune risk variant and triggered by ligand functions to protect against viral challenge, which probably accounts for its selection within human populations but provides this advantage at the cost of modestly promoting the risk of autoimmunity.


Asunto(s)
Autoinmunidad/genética , Infecciones por Cardiovirus/genética , Interferón Tipo I/inmunología , Helicasa Inducida por Interferón IFIH1/genética , Adolescente , Adulto , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Autoinmunidad/inmunología , Southern Blotting , Infecciones por Cardiovirus/inmunología , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Virus de la Encefalomiocarditis/inmunología , Femenino , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Immunoblotting , Helicasa Inducida por Interferón IFIH1/inmunología , Masculino , Ratones , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virosis/genética , Virosis/inmunología , Adulto Joven
17.
Virol J ; 14(1): 45, 2017 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-28259172

RESUMEN

BACKGROUD: Encephalomyocarditis virus (EMCV) has been discovered on pig farms worldwide and can cause myocarditis in piglets and reproductive failure in sows. However, little is known about the host transcriptional responses to infection and host-pathogen interactions. METHODS: In this study, transcription profiling was performed by Illumina RNA-Sequencing (RNA-seq) to identify EMCV induced differentially expressed genes in BHK-21 cells at serial time points (12, 24, and 30 h post infection (hpi)), using mock infected cells as control. RESULTS: We identified 237, 241, and 207 differentially expressed genes (DEGs) respectively, majority of which were up-regulated. A large number of DEGs clustered into host defense, cellular signaling and metabolism categories. Moreover, short time series expression analysis revealed that 12 hpi was an important time point for expression change, indicating host virus resistance. CONCLUSIONS: This RNA-seq analysis provides the first data for understanding the network of virus host interactions under EMCV infection in vitro, and for identifying host components which involved in the virus infection course.


Asunto(s)
Virus de la Encefalomiocarditis/inmunología , Virus de la Encefalomiocarditis/patogenicidad , Células Epiteliales/virología , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno , Animales , Línea Celular , Cricetinae , Análisis de Secuencia de ARN , Factores de Tiempo
18.
Immunobiology ; 222(2): 350-357, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27665995

RESUMEN

Viral infections can give rise to secondary bacterial infections. In the present study, we examined the role of invariant natural killer T (iNKT) cells in lipopolysaccharide (LPS)-induced lethal shock during encephalomyocarditis virus (EMCV) infection. Wild-type (WT) mice and Jα18 gene knockout (Jα18 KO) mice were inoculated with EMCV, 5days prior to challenging with LPS. The survival rate of Jα18 KO mice subjected to EMCV and LPS was significantly higher than that of WT mice. TNF-α and nitric oxide (NO) production were increased in WT mice, than that in Jα18 KO mice, after the administration of EMCV and LPS. EMCV infection increased the number of iNKT cells and IFN-γ production by iNKT cells in WT mice. Moreover, EMCV infection enhanced the expression of Toll-like receptor 4 (TLR4) in the lung and spleen. IFN-γ also increased the expression of TLR4 in splenocytes. These findings indicated that EMCV infection activated iNKT cells, and IFN-γ secreted from the iNKT cells up-regulated the expression of TLR4 in various tissues. As a result, EMCV-infected mice were susceptible to LPS and easily developed the lethal shock. In conclusion, iNKT cells were involved in the development of LPS-induced lethal shock during EMCV infection.


Asunto(s)
Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/metabolismo , Virus de la Encefalomiocarditis/inmunología , Lipopolisacáridos/efectos adversos , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Choque Séptico/etiología , Choque Séptico/metabolismo , Animales , Biomarcadores , Infecciones por Cardiovirus/mortalidad , Infecciones por Cardiovirus/virología , Coinfección , Citocinas/metabolismo , Modelos Animales de Enfermedad , Lipopolisacáridos/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Óxido Nítrico/metabolismo , Choque Séptico/mortalidad , Factor de Necrosis Tumoral alfa/metabolismo
19.
J Virol ; 90(24): 11106-11121, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27707918

RESUMEN

The role of retinoic acid-inducible gene I (RIG-I) in foot-and-mouth disease virus (FMDV)-infected cells remains unknown. Here, we showed that RIG-I inhibits FMDV replication in host cells. FMDV infection increased the transcription of RIG-I, while it decreased RIG-I protein expression. A detailed analysis revealed that FMDV leader proteinase (Lpro), as well as 3C proteinase (3Cpro) and 2B protein, decreased RIG-I protein expression. Lpro and 3Cpro are viral proteinases that can cleave various host proteins and are responsible for several of the viral polyprotein cleavages. However, for the first time, we observed 2B-induced reduction of host protein. Further studies showed that 2B-mediated reduction of RIG-I is specific to FMDV, but not other picornaviruses, including encephalomyocarditis virus, enterovirus 71, and coxsackievirus A16. Moreover, we found the decreased protein level of RIG-I is independent of the cleavage of eukaryotic translation initiation factor 4 gamma, the induction of cellular apoptosis, or the association of proteasome, lysosome, and caspase pathways. A direct interaction was observed between RIG-I and 2B. The carboxyl-terminal amino acids 105 to 114 and amino acids 135 to 144 of 2B were essential for the reduction of RIG-I, while residues 105 to 114 were required for the interaction. These data suggest the antiviral role of RIG-I against FMDV and a novel antagonistic mechanism of FMDV that is mediated by 2B protein. IMPORTANCE: This study demonstrated that RIG-I could suppress FMDV replication during virus infection. FMDV infection increased the transcriptional expression of RIG-I, while it decreased RIG-I protein expression. FMDV 2B protein interacted with RIG-I and induced reduction of RIG-I. 2B-induced reduction of RIG-I was independent of the induction of the cleavage of eukaryotic translation initiation factor 4 gamma or cellular apoptosis. In addition, proteasome, lysosome, and caspase pathways were not involved in this process. This study provides new insight into the immune evasion mediated by FMDV and identifies 2B as an antagonistic factor for FMDV to evade the antiviral response.


Asunto(s)
Cisteína Endopeptidasas/genética , Proteína 58 DEAD Box/genética , Endopeptidasas/genética , Factor 4G Eucariótico de Iniciación/genética , Virus de la Fiebre Aftosa/genética , Interacciones Huésped-Patógeno , Proteínas Virales/genética , Proteínas Reguladoras y Accesorias Virales/genética , Proteasas Virales 3C , Secuencia de Aminoácidos , Animales , Sistemas CRISPR-Cas , Línea Celular , Cricetulus , Cisteína Endopeptidasas/inmunología , Proteína 58 DEAD Box/inmunología , Virus de la Encefalomiocarditis/genética , Virus de la Encefalomiocarditis/inmunología , Endopeptidasas/inmunología , Enterovirus/genética , Enterovirus/inmunología , Enterovirus Porcinos/genética , Enterovirus Porcinos/inmunología , Células Epiteliales , Factor 4G Eucariótico de Iniciación/inmunología , Virus de la Fiebre Aftosa/inmunología , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Unión Proteica , Transducción de Señal , Especificidad de la Especie , Porcinos , Proteínas Virales/inmunología , Proteínas Reguladoras y Accesorias Virales/inmunología
20.
J Immunol ; 196(9): 3877-86, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27016603

RESUMEN

Moloney leukemia virus 10, homolog (MOV10) is an IFN-inducible RNA helicase, associated with small RNA-induced silencing. In this article, we report that MOV10 exhibits antiviral activity, independent of its helicase function, against a number of positive- and negative-strand RNA viruses by enhancing type I IFN induction. Using a number of genome-edited knockout human cells, we show that IFN regulatory factor 3-mediated IFN induction and downstream IFN signaling through IFN receptor was necessary to inhibit virus replication by MOV10. MOV10 enhanced IFN regulatory factor 3-mediated transcription of IFN. However, this IFN induction by MOV10 was unique and independent of the known retinoic acid-inducible gene I/mitochondrial antiviral-signaling protein-mediated RNA-sensing pathway. Upon virus infection, MOV10 specifically required inhibitor of κB kinase ε, not TANK-binding kinase 1, for its antiviral activity. The important role of MOV10 in mediating antiviral signaling was further supported by the finding that viral proteases from picornavirus family specifically targeted MOV10 as a possible innate immune evasion mechanism. These results establish MOV10, an evolutionary conserved protein involved in RNA silencing, as an antiviral gene against RNA viruses that uses an retinoic acid-inducible gene I-like receptor-independent pathway to enhance IFN response.


Asunto(s)
Infecciones por Cardiovirus/inmunología , Virus de la Encefalomiocarditis/inmunología , ARN Helicasas/metabolismo , Infecciones por Rhabdoviridae/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Evasión Inmune , Inmunidad Innata , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , ARN Helicasas/genética , Interferencia de ARN , ARN Viral/genética , Receptores de Ácido Retinoico/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...