Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Nat Immunol ; 17(12): 1373-1380, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27695001

RESUMEN

The respiratory tract is heavily populated with innate immune cells, but the mechanisms that control such cells are poorly defined. Here we found that the E3 ubiquitin ligase TRIM29 was a selective regulator of the activation of alveolar macrophages, the expression of type I interferons and the production of proinflammatory cytokines in the lungs. We found that deletion of TRIM29 enhanced macrophage production of type I interferons and protected mice from infection with influenza virus, while challenge of Trim29-/- mice with Haemophilus influenzae resulted in lethal lung inflammation due to massive production of proinflammatory cytokines by macrophages. Mechanistically, we demonstrated that TRIM29 inhibited interferon-regulatory factors and signaling via the transcription factor NF-κB by degrading the adaptor NEMO and that TRIM29 directly bound NEMO and subsequently induced its ubiquitination and proteolytic degradation. These data identify TRIM29 as a key negative regulator of alveolar macrophages and might have important clinical implications for local immunity and immunopathology.


Asunto(s)
Infecciones por Haemophilus/inmunología , Haemophilus influenzae/inmunología , Virus de la Influenza A/inmunología , Macrófagos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Sistema Respiratorio/inmunología , Factores de Transcripción/metabolismo , Animales , Células Cultivadas , Inmunidad Innata , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Macrófagos/microbiología , Macrófagos/virología , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Proteolisis , Transducción de Señal , Factores de Transcripción/genética , Ubiquitinación
2.
PLoS Pathog ; 19(9): e1011641, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37708231

RESUMEN

RNA viruses cause numerous infectious diseases in humans and animals. The crosstalk between RNA viruses and the innate DNA sensing pathways attracts increasing attention. Recent studies showed that the cGAS-STING pathway plays an important role in restricting RNA viruses via mitochondria DNA (mtDNA) mediated activation. However, the mechanisms of cGAS mediated innate immune evasion by RNA viruses remain unknown. Here, we report that seneca valley virus (SVV) protease 3C disrupts mtDNA mediated innate immune sensing by cleaving porcine cGAS (pcGAS) in a species-specific manner. Mechanistically, a W/Q motif within the N-terminal domain of pcGAS is a unique cleavage site recognized by SVV 3C. Three conserved catalytic residues of SVV 3C cooperatively contribute to the cleavage of pcGAS, but not human cGAS (hcGAS) or mouse cGAS (mcGAS). Additionally, upon SVV infection and poly(dA:dT) transfection, pcGAS and SVV 3C colocalizes in the cells. Furthermore, SVV 3C disrupts pcGAS-mediated DNA binding, cGAMP synthesis and interferon induction by specifically cleaving pcGAS. This work uncovers a novel mechanism by which the viral protease cleaves the DNA sensor cGAS to evade innate immune response, suggesting a new antiviral approach against picornaviruses.


Asunto(s)
Nucleotidiltransferasas , Péptido Hidrolasas , Picornaviridae , Animales , Humanos , Ratones , ADN Mitocondrial , Endopeptidasas , Mitocondrias , Picornaviridae/fisiología , Porcinos , Nucleotidiltransferasas/metabolismo
4.
Cell Mol Life Sci ; 79(6): 313, 2022 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-35604464

RESUMEN

Gastroenteritis is inflammation of the lining of stomach and intestines and causes significant morbidity and mortality worldwide. Many viruses, especially RNA viruses are the most common cause of enteritis. Innate immunity is the first line of host defense against enteric RNA viruses and virus-induced intestinal inflammation. The first layer of defense against enteric RNA viruses in the intestinal tract is intestinal epithelial cells (IECs), dendritic cells and macrophages under the intestinal epithelium. These innate immune cells express pathogen-recognition receptors (PRRs) for recognizing enteric RNA viruses through sensing viral pathogen-associated molecular patterns (PAMPs). As a result of this recognition type I interferon (IFN), type III IFN and inflammasome activation occurs, which function cooperatively to clear infection and reduce viral-induced intestinal inflammation. In this review, we summarize recent findings about mechanisms involved in enteric RNA virus-induced intestinal inflammation. We will provide an overview of the enteric RNA viruses, their RNA sensing mechanisms by host PRRs, and signaling pathways triggered by host PRRs, which shape the intestinal immune response to maintain intestinal homeostasis.


Asunto(s)
Virus ARN , Humanos , Inmunidad Innata , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Intestinos , Moléculas de Patrón Molecular Asociado a Patógenos/metabolismo
5.
J Biomed Sci ; 29(1): 55, 2022 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-35909127

RESUMEN

BACKGROUND: Infections by viruses including severe acute respiratory syndrome coronavirus 2 could cause organ inflammations such as myocarditis, pneumonia and encephalitis. Innate immunity to viral nucleic acids mediates antiviral immunity as well as inflammatory organ injury. However, the innate immune mechanisms that control viral induced organ inflammations are unclear. METHODS: To understand the role of the E3 ligase TRIM18 in controlling viral myocarditis and organ inflammation, wild-type and Trim18 knockout mice were infected with coxsackievirus B3 for inducing viral myocarditis, influenza A virus PR8 strain and human adenovirus for inducing viral pneumonia, and herpes simplex virus type I for inducing herpes simplex encephalitis. Mice survivals were monitored, and heart, lung and brain were harvested for histology and immunohistochemistry analysis. Real-time PCR, co-immunoprecipitation, immunoblot, enzyme-linked immunosorbent assay, luciferase assay, flow cytometry, over-expression and knockdown techniques were used to understand the molecular mechanisms of TRIM18 in regulating type I interferon (IFN) production after virus infection in this study. RESULTS: We find that knockdown or deletion of TRIM18 in human or mouse macrophages enhances production of type I IFN in response to double strand (ds) RNA and dsDNA or RNA and DNA virus infection. Importantly, deletion of TRIM18 protects mice from viral myocarditis, viral pneumonia, and herpes simplex encephalitis due to enhanced type I IFN production in vivo. Mechanistically, we show that TRIM18 recruits protein phosphatase 1A (PPM1A) to dephosphorylate TANK binding kinase 1 (TBK1), which inactivates TBK1 to block TBK1 from interacting with its upstream adaptors, mitochondrial antiviral signaling (MAVS) and stimulator of interferon genes (STING), thereby dampening antiviral signaling during viral infections. Moreover, TRIM18 stabilizes PPM1A by inducing K63-linked ubiquitination of PPM1A. CONCLUSIONS: Our results indicate that TRIM18 serves as a negative regulator of viral myocarditis, lung inflammation and brain damage by downregulating innate immune activation induced by both RNA and DNA viruses. Our data reveal that TRIM18 is a critical regulator of innate immunity in viral induced diseases, thereby identifying a potential therapeutic target for treatment.


Asunto(s)
Encefalitis por Herpes Simple , Miocarditis , Ubiquitina-Proteína Ligasas , Virosis , Animales , Antivirales , Humanos , Inmunidad Innata , Inflamación/genética , Ratones , Miocarditis/genética , Miocarditis/virología , Proteína Fosfatasa 2C , ARN , Ubiquitina-Proteína Ligasas/genética
6.
EMBO Rep ; 21(7): e49666, 2020 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-32352641

RESUMEN

Inflammasomes are intracellular complexes that form in the cytosol of inflammatory cells. NLRP3 is one of the sensor proteins in the complex that can recognize a wide variety of stimuli ranging from microbial components to environmental particulates. Here, we report that in mouse airway epithelial cells (AECs), inflammasome activation is inhibited by EphA2, a member of the transmembrane tyrosine kinase receptor family, via tyrosine phosphorylation of NLRP3 in a model of reovirus infection. We find that EphA2 depletion markedly enhances interleukin-1ß (IL-1ß) and interleukin-18 (IL-18) production in response to the virus. EphA2-/- mice show stronger inflammatory infiltration and enhanced inflammasome activation upon viral infection, and aggravated asthma symptoms upon ovalbumin (ova) induction. Mechanistically, EphA2 binds to NLRP3 and induces its phosphorylation at Tyr132, thereby interfering with ASC speck formation and blocking the activation of the NLRP3-inflammasome. These data demonstrate that reovirus employs EphA2 to suppress inflammasome activation in AECs and that EphA2 deficiency causes a pathological exacerbation of asthma in an ova-induced asthma model.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Animales , Proteínas Portadoras , Células Epiteliales/metabolismo , Inflamasomas/genética , Inflamasomas/metabolismo , Interleucina-18 , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/genética
7.
J Immunol ; 203(4): 873-880, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31270148

RESUMEN

NK cells play an important role in immune surveillance and protective immunity, mainly through rapid cytokine release and cytolytic activities. But how such responses are negatively regulated remains poorly defined. In this study, we demonstrated that the E3 ubiquitin ligase TRIM29 is a crucial regulator of NK cell functions. We found that TRIM29 was not expressed in resting NK cells, but was readily upregulated following activation, especially after IL-12 plus IL-18 stimulation. The levels of TRIM29 expression were inversely correlated with IFN-γ production by NK cells, suggesting that TRIM29 inhibits NK cell functions. Indeed, deficiency of TRIM29, specifically in NK cells, resulted in an enhanced IFN-γ production and consequently protected mice from murine CMV infection. Mechanistically, we showed that once induced in NK cells, TRIM29 ubiquitinates and degrades the TGF-ß-activated kinase 1 binding protein 2 (TAB2), a key adaptor protein in IFN-γ production by NK cells. These results identify TRIM29 as a negative regulator of NK cell functions and may have important clinical implications.


Asunto(s)
Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/inmunología , Factores de Transcripción/inmunología , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Ratones , Ubiquitinación
8.
J Immunol ; 201(1): 183-192, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29769269

RESUMEN

The innate immunity is critically important in protection against virus infections, and in the case of RNA viral infections, the signaling mechanisms that initiate robust protective innate immunity without triggering autoimmune inflammation remain incompletely defined. In this study, we found the E3 ligase TRIM29 was specifically expressed in poly I:C-stimulated human myeloid dendritic cells. The induced TRIM29 played a negative role in type I IFN production in response to poly I:C or dsRNA virus reovirus infection. Importantly, the challenge of wild-type mice with reovirus led to lethal infection. In contrast, deletion of TRIM29 protected the mice from this developing lethality. Additionally, TRIM29-/- mice have lower titers of reovirus in the heart, intestine, spleen, liver, and brain because of elevated production of type I IFN. Mechanistically, TRIM29 was shown to interact with MAVS and subsequently induce its K11-linked ubiquitination and degradation. Taken together, TRIM29 regulates negatively the host innate immune response to RNA virus, which could be employed by RNA viruses for viral pathogenesis.


Asunto(s)
Inmunidad Innata/inmunología , Interferón Tipo I/biosíntesis , Infecciones por Reoviridae/inmunología , Reoviridae/inmunología , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Células Cultivadas , Células Dendríticas/inmunología , Humanos , Interferón Tipo I/inmunología , Ratones , Ratones Noqueados , Poli I-C , Factores de Transcripción/genética , Ubiquitinación
9.
J Virol ; 89(5): 2944-55, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25552708

RESUMEN

UNLABELLED: Arenavirus pathogens cause a wide spectrum of diseases in humans ranging from central nervous system disease to lethal hemorrhagic fevers with few treatment options. The reason why some arenaviruses can cause severe human diseases while others cannot is unknown. We find that the Z proteins of all known pathogenic arenaviruses, lymphocytic choriomeningitis virus (LCMV) and Lassa, Junin, Machupo, Sabia, Guanarito, Chapare, Dandenong, and Lujo viruses, can inhibit retinoic acid-inducible gene 1 (RIG-i) and Melanoma Differentiation-Associated protein 5 (MDA5), in sharp contrast to those of 14 other nonpathogenic arenaviruses. Inhibition of the RIG-i-like receptors (RLRs) by pathogenic Z proteins is mediated by the protein-protein interactions of Z and RLRs, which lead to the disruption of the interactions between RLRs and mitochondrial antiviral signaling (MAVS). The Z-RLR interactive interfaces are located within the N-terminal domain (NTD) of the Z protein and the N-terminal CARD domains of RLRs. Swapping of the LCMV Z NTD into the nonpathogenic Pichinde virus (PICV) genome does not affect virus growth in Vero cells but significantly inhibits the type I interferon (IFN) responses and increases viral replication in human primary macrophages. In summary, our results show for the first time an innate immune-system-suppressive mechanism shared by the diverse pathogenic arenaviruses and thus shed important light on the pathogenic mechanism of human arenavirus pathogens. IMPORTANCE: We show that all known human-pathogenic arenaviruses share an innate immune suppression mechanism that is based on viral Z protein-mediated RLR inhibition. Our report offers important insights into the potential mechanism of arenavirus pathogenesis, provides a convenient way to evaluate the pathogenic potential of known and/or emerging arenaviruses, and reveals a novel target for the development of broad-spectrum therapies to treat this group of diverse pathogens. More broadly, our report provides a better understanding of the mechanisms of viral immune suppression and host-pathogen interactions.


Asunto(s)
ARN Helicasas DEAD-box/antagonistas & inhibidores , Interacciones Huésped-Patógeno , Interferones/antagonistas & inhibidores , Virus de la Coriomeningitis Linfocítica/fisiología , Virus Pichinde/fisiología , Proteínas Virales/metabolismo , Replicación Viral , Animales , Células Cultivadas , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/metabolismo , Humanos , Tolerancia Inmunológica , Interferones/biosíntesis , Virus de la Coriomeningitis Linfocítica/genética , Virus Pichinde/genética , Unión Proteica , Mapeo de Interacción de Proteínas , Receptores Inmunológicos , Recombinación Genética , Proteínas Virales/genética
10.
J Virol ; 89(24): 12513-7, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26423945

RESUMEN

Several arenavirus pathogens, such as Lassa and Junin viruses, inhibit macrophage activation, the molecular mechanism of which is unclear. We show that lymphocytic choriomeningitis virus (LCMV) can also inhibit macrophage activation, in contrast to Pichinde and Tacaribe viruses, which are not known to naturally cause human diseases. Using a recombinant Pichinde virus system, we show that the LCMV Z N-terminal domain (NTD) mediates the inhibition of macrophage activation and immune functions.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Proteínas Portadoras/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Activación de Macrófagos , Macrófagos/inmunología , Virus Pichinde/inmunología , Animales , Humanos , Péptidos y Proteínas de Señalización Intracelular , Macrófagos/virología , Ratones , Estructura Terciaria de Proteína
11.
J Virol ; 89(13): 6595-607, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25878103

RESUMEN

UNLABELLED: Arenaviruses cause severe hemorrhagic fever diseases in humans, and there are limited preventative and therapeutic measures against these diseases. Previous structural and functional analyses of arenavirus nucleoproteins (NPs) revealed a conserved DEDDH exoribonuclease (RNase) domain that is important for type I interferon (IFN) suppression, but the biological roles of the NP RNase in viral replication and host immune suppression have not been well characterized. Infection of guinea pigs with Pichinde virus (PICV), a prototype arenavirus, can serve as a surrogate small animal model for arenavirus hemorrhagic fevers. In this report, we show that mutation of each of the five RNase catalytic residues of PICV NP diminishes the IFN suppression activity and slightly reduces the viral RNA replication activity. Recombinant PICVs with RNase catalytic mutations can induce high levels of IFNs and barely grow in IFN-competent A549 cells, in sharp contrast to the wild-type (WT) virus, while in IFN-deficient Vero cells, both WT and mutant viruses can replicate at relatively high levels. Upon infection of guinea pigs, the RNase mutant viruses stimulate strong IFN responses, fail to replicate productively, and can become WT revertants. Serial passages of the RNase mutants in vitro can also generate WT revertants. Thus, the NP RNase function is essential for the innate immune suppression that allows the establishment of a productive early viral infection, and it may be partly involved in the process of viral RNA replication. IMPORTANCE: Arenaviruses, such as Lassa, Lujo, and Machupo viruses, can cause severe and deadly hemorrhagic fever diseases in humans, and there are limited preventative and treatment options against these diseases. Development of broad-spectrum antiviral drugs depends on a better mechanistic understanding of the conserved arenavirus proteins in viral infection. The nucleoprotein (NPs) of all arenaviruses carry a unique exoribonuclease (RNase) domain that has been shown to be critical for the suppression of type I interferons. However, the functional roles of the NP RNase in arenavirus replication and host immune suppression have not been characterized systematically. Using a prototype arenavirus, Pichinde virus (PICV), we characterized the viral growth and innate immune suppression of recombinant RNase-defective mutants in both cell culture and guinea pig models. Our study suggests that the NP RNase plays an essential role in the suppression of host innate immunity, and possibly in viral RNA replication, and that it can serve as a novel target for developing antiviral drugs against arenavirus pathogens.


Asunto(s)
Exorribonucleasas/metabolismo , Interacciones Huésped-Patógeno , Evasión Inmune , Nucleoproteínas/metabolismo , Virus Pichinde/enzimología , Virus Pichinde/fisiología , Replicación Viral , Secuencias de Aminoácidos , Sustitución de Aminoácidos , Animales , Infecciones por Arenaviridae/inmunología , Infecciones por Arenaviridae/virología , Línea Celular , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Exorribonucleasas/genética , Cobayas , Humanos , Masculino , Nucleoproteínas/genética , Virus Pichinde/genética , Virus Pichinde/inmunología
12.
J Virol ; 87(17): 9788-801, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23824799

RESUMEN

Host cells activate innate immune signaling pathways to defend against invading pathogens. To survive within an infected host, viruses have evolved intricate strategies to counteract host immune responses. Herpesviruses, including herpes simplex virus type 1 (HSV-1), have large genomes and therefore have the capacity to encode numerous proteins that modulate host innate immune responses. Here we define the contribution of HSV-1 tegument protein VP16 in the inhibition of beta interferon (IFN-ß) production. VP16 was demonstrated to significantly inhibit Sendai virus (SeV)-induced IFN-ß production, and its transcriptional activation domain was not responsible for this inhibition activity. Additionally, VP16 blocked the activation of the NF-κB promoter induced by SeV or tumor necrosis factor alpha treatment and expression of NF-κB-dependent genes through interaction with p65. Coexpression analysis revealed that VP16 selectively blocked IFN regulatory factor 3 (IRF-3)-mediated but not IRF-7-mediated transactivation. VP16 was able to bind to IRF-3 but not IRF-7 in vivo, based on coimmunoprecipitation analysis, but it did not affect IRF-3 dimerization, nuclear translocation, or DNA binding activity. Rather, VP16 interacted with the CREB binding protein (CBP) coactivator and efficiently inhibited the formation of the transcriptional complexes IRF-3-CBP in the context of HSV-1 infection. These results illustrate that VP16 is able to block the production of IFN-ß by inhibiting NF-κB activation and interfering with IRF-3 to recruit its coactivator CBP, which may be important to the early events leading to HSV-1 infection.


Asunto(s)
Proteína de Unión a CREB/metabolismo , Proteína Vmw65 de Virus del Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/biosíntesis , Factor de Transcripción ReIA/metabolismo , Animales , Chlorocebus aethiops , Células HEK293 , Células HeLa , Proteína Vmw65 de Virus del Herpes Simple/química , Proteína Vmw65 de Virus del Herpes Simple/genética , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata , Factor 3 Regulador del Interferón/antagonistas & inhibidores , Factor 7 Regulador del Interferón/metabolismo , Interferón beta/genética , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , Virus Sendai/inmunología , Virus Sendai/patogenicidad , Factor de Transcripción ReIA/antagonistas & inhibidores , Factor de Transcripción ReIA/genética , Activación Transcripcional , Células Vero
13.
Nat Commun ; 15(1): 3481, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38664417

RESUMEN

Viral myocarditis, an inflammatory disease of the myocardium, is a significant cause of sudden death in children and young adults. The current coronavirus disease 19 pandemic emphasizes the need to understand the pathogenesis mechanisms and potential treatment strategies for viral myocarditis. Here, we found that TRIM29 was highly induced by cardiotropic viruses and promoted protein kinase RNA-like endoplasmic reticulum kinase (PERK)-mediated endoplasmic reticulum (ER) stress, apoptosis, and reactive oxygen species (ROS) responses that promote viral replication in cardiomyocytes in vitro. TRIM29 deficiency protected mice from viral myocarditis by promoting cardiac antiviral functions and reducing PERK-mediated inflammation and immunosuppressive monocytic myeloid-derived suppressor cells (mMDSC) in vivo. Mechanistically, TRIM29 interacted with PERK to promote SUMOylation of PERK to maintain its stability, thereby promoting PERK-mediated signaling pathways. Finally, we demonstrated that the PERK inhibitor GSK2656157 mitigated viral myocarditis by disrupting the TRIM29-PERK connection, thereby bolstering cardiac function, enhancing cardiac antiviral responses, and curbing inflammation and immunosuppressive mMDSC in vivo. Our findings offer insight into how cardiotropic viruses exploit TRIM29-regulated PERK signaling pathways to instigate viral myocarditis, suggesting that targeting the TRIM29-PERK axis could mitigate disease severity.


Asunto(s)
Adenina , Estrés del Retículo Endoplásmico , Indoles , Miocarditis , Miocitos Cardíacos , eIF-2 Quinasa , Animales , Humanos , Masculino , Ratones , Adenina/análogos & derivados , Apoptosis , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Miocarditis/virología , Miocarditis/metabolismo , Miocarditis/patología , Miocardio/patología , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/virología , Miocitos Cardíacos/patología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Replicación Viral
14.
J Virol ; 86(7): 3528-40, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22301138

RESUMEN

The interferon (IFN)-mediated antiviral response is a major defense of the host immune system. In order to complete their life cycle, viruses must modulate host IFN-mediated immune responses. Herpes simplex virus 1 (HSV-1) is a large DNA virus containing more than 80 genes, many of which encode proteins that are involved in virus-host interactions and show immune modulatory capabilities. In this study, we demonstrate that the US11 protein, an RNA binding tegument protein of HSV-1, is a novel antagonist of the beta IFN (IFN-ß) pathway. US11 significantly inhibited Sendai virus (SeV)-induced IFN-ß production, and its double-stranded RNA (dsRNA) binding domain was indispensable for this inhibition activity. Additionally, wild-type HSV-1 coinfection showed stronger inhibition than US11 mutant HSV-1 in SeV-induced IFN-ß production. Coimmunoprecipitation analysis demonstrated that the US11 protein in HSV-1-infected cells interacts with endogenous RIG-I and MDA-5 through its C-terminal RNA-binding domain, which was RNA independent. Expression of US11 in both transfected and HSV-1-infected cells interferes with the interaction between MAVS and RIG-I or MDA-5. Finally, US11 dampens SeV-mediated IRF3 activation. Taken together, the combined data indicate that HSV-1 US11 binds to RIG-I and MDA-5 and inhibits their downstream signaling pathway, preventing the production of IFN-ß, which may contribute to the pathogenesis of HSV-1 infection.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Regulación hacia Abajo , Herpes Simple/metabolismo , Herpesvirus Humano 1/metabolismo , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Proteínas Virales/metabolismo , Línea Celular , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/química , ARN Helicasas DEAD-box/genética , Herpes Simple/genética , Herpes Simple/virología , Herpesvirus Humano 1/química , Herpesvirus Humano 1/genética , Helicasa Inducida por Interferón IFIH1 , Interferón beta/genética , Interferón beta/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Receptores Inmunológicos , Proteínas Virales/química , Proteínas Virales/genética
15.
Comput Struct Biotechnol J ; 21: 2801-2808, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37168870

RESUMEN

Protein ubiquitination is a post-translation modification mediated by E3 ubiquitin ligases. The RING domain E3 ligases are the largest family of E3 ubiquitin ligases, they act as a scaffold, bringing the E2-ubiquitin complex and its substrate together to facilitate direct ubiquitin transfer. However, the quaternary structures of RING E3 ligases that perform ubiquitin transfer remain poorly understood. In this study, we solved the crystal structure of TRIM56, a member of the RING E3 ligase. The structure of the coiled-coil domain indicated that the two anti-parallel dimers bound together to form a tetramer at a small crossing angle. This tetramer structure allows two RING domains to exist on each side to form an active homodimer in supporting ubiquitin transfer from E2 to its nearby substrate recruited by the C-terminal domains on the same side. These findings suggest that the coiled-coil domain-mediated tetramer is a feasible scaffold for facilitating the recruitment and transfer of ubiquitin to accomplish E3 ligase activity.

16.
J Gen Virol ; 93(Pt 9): 1869-1875, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22622329

RESUMEN

Herpes simplex virus type 1 (HSV-1) triplex is a complex of three protein subunits, consisting of two copies of VP23 and one copy of VP19C. Here, we identified a non-classical NLS of VP19C between aa 50 and 61, and the nuclear import of VP19C was mediated by RanGTP and importin ß1-, but not importin α5-, dependent pathway. Additionally, recombinant virus harbouring this NLS mutation (NLSm) replicates less efficiently as wild-type. These data strongly suggested that the nuclear import of VP19C is required for efficient HSV-1 production.


Asunto(s)
Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Núcleo Celular/metabolismo , Herpes Simple/virología , Herpesvirus Humano 1/fisiología , Señales de Localización Nuclear , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Proteínas de la Cápside/genética , Línea Celular , Núcleo Celular/genética , Herpes Simple/metabolismo , Herpesvirus Humano 1/química , Herpesvirus Humano 1/genética , Humanos , Datos de Secuencia Molecular , Ensamble de Virus
17.
J Virol ; 85(21): 11079-89, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21835786

RESUMEN

Varicella-zoster virus (VZV) infection of differentiated cells within the host and establishment of latency likely requires evasion of innate immunity and limits secretion of antiviral cytokines. Here we report that its immediate-early protein ORF61 antagonizes the beta interferon (IFN-ß) pathway. VZV infection down-modulated the Sendai virus (SeV)-activated IFN-ß pathway, including mRNA of IFN-ß and its downstream interferon-stimulated genes (ISGs), ISG54 and ISG56. Through a primary screening of VZV genes, we found that ORF61 inhibited SeV-mediated activation of IFN-ß and ISRE (IFN-stimulated response element) promoter activities but only slightly affected NF-κB promoter activity, implying that the IFN-ß pathway may be blocked in the IRF3 branch. An indirect immunofluorescence assay demonstrated that ectopic expression of ORF61 abrogated the detection of IRF3 in SeV-infected cells; however, it did not affect endogenous dormant IRF3 in noninfected cells. Additionally, ORF61 was shown to be partially colocalized with activated IRF3 in the nucleus upon treatment with MG132, an inhibitor of proteasomes, and the direct interaction between ORF61 and activated IRF3 was confirmed by a coimmunoprecipitation assay. Furthermore, Western blot analysis demonstrated that activated IRF3 was ubiquitinated in the presence of ORF61, suggesting that ORF61 degraded phosphorylated IRF3 via a ubiquitin-proteasome pathway. Semiquantitative reverse transcription-PCR (RT-PCR) analysis demonstrated that the level of ISG54 and ISG56 mRNAs was also downregulated by ORF61. Taken together, our results convincingly demonstrate that ORF61 down-modulates the IRF3-mediated IFN-ß pathway by degradation of activated IRF3 via direct interaction, which may contribute to the pathogenesis of VZV infection.


Asunto(s)
Herpesvirus Humano 3/inmunología , Herpesvirus Humano 3/patogenicidad , Evasión Inmune , Inmunidad Innata , Factor 3 Regulador del Interferón/antagonistas & inhibidores , Proteínas Virales/metabolismo , Western Blotting , Línea Celular , Humanos , Inmunoprecipitación , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/antagonistas & inhibidores , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virus Sendai/inmunología , Ubiquitina/metabolismo
18.
J Virol ; 85(4): 1881-6, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21147926

RESUMEN

It has been reported that herpes simplex virus type 1 UL3, UL4, and UL20.5 proteins are localized to small, dense nuclear bodies together with ICP22 in infected cells. In the present study, we comprehensively characterized these interactions by subcellular colocalization, coimmunoprecipitation, and bimolecular fluorescence complementation assays. For the first time, it was demonstrated that both UL3 and UL20.5 are targeted to small, dense nuclear bodies by a direct interaction with ICP22, whereas UL4 colocalizes with ICP22 through its interaction with UL3 but not UL20.5 or ICP22. There was no detectable interaction between UL3 and UL20.5.


Asunto(s)
Herpesvirus Humano 1/patogenicidad , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Virales/metabolismo , Núcleo Celular/metabolismo , Células HEK293 , Herpesvirus Humano 1/metabolismo , Humanos , Inmunoprecipitación , Fracciones Subcelulares/metabolismo , Proteínas Virales/genética
19.
Med Microbiol Immunol ; 201(3): 381-7, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22628116

RESUMEN

Human cytomegalovirus UL79 protein is recently reported to be required for transcription or efficient accumulation of late viral mRNAs during viral infection. An absolute nuclear distribution of UL79 proteins has been observed with immunofluorescence assay, both during the infection of Flag-tagged UL79 recombinant virus and in the HFFs expressing HA-tagged UL79, with or without virus infection. However, little is known about the nuclear import mechanism of UL79 protein. Here, by utilizing living cells fluorescent microscopy, a predominant nuclear localization of UL79 protein in living cells was detected. Furthermore, the nuclear import of UL79 protein was demonstrated to be dependent on the transportin-1-mediated pathway. Finally, a hydrophobic PY-nuclear localization signal (PY-NLS) was delineated between the amino acids 66-92 of UL79 protein. Collectively, we provide evidence that a PY-NLS, firstly described in viral proteins, is responsible for the nuclear accumulation of UL79 protein.


Asunto(s)
Núcleo Celular/metabolismo , Citomegalovirus/metabolismo , Señales de Localización Nuclear/genética , Proteínas Virales/metabolismo , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Células COS , Chlorocebus aethiops , Citomegalovirus/genética , Células HEK293 , Humanos , Microscopía Fluorescente , Datos de Secuencia Molecular , Proteínas Virales/química , Proteínas Virales/genética , beta Carioferinas/metabolismo
20.
Arch Virol ; 157(7): 1383-6, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22527865

RESUMEN

The herpes simplex virus type I protein VP22 has been reported to have the property of intercellular trafficking. However, there is little direct evidence to demonstrate that VP22 can shuttle freely between living cells. Here, we employ a novel and simple assay using live cell fluorescence microscopy to investigate the intercellular transport property. Our results demonstrated that VP22, bovine herpesvirus-1 VP22, HSV-1 US11 and HIV Tat could not shuttle into neighboring cells via direct contact.


Asunto(s)
Comunicación Celular/fisiología , Regulación Viral de la Expresión Génica/fisiología , Microscopía Fluorescente/métodos , Proteínas Virales/metabolismo , Animales , Transporte Biológico , Células COS , Chlorocebus aethiops , Células HEK293 , Humanos , Transporte de Proteínas/fisiología , Proteínas Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA