Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Adv Sci (Weinh) ; : e2404365, 2024 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-39159143

RESUMEN

Ferroptosis is a novel form of cell death caused by the accumulation of lipid peroxides in an iron-dependent manner. However, the precise mechanism underlying the exploitation of ferroptosis by influenza A viruses (IAV) remains unclear. The results demonstrate that IAV promotes its own replication through ferritinophagy by sensitizing cells to ferroptosis, with hemagglutinin identified as a key trigger in this process. Hemagglutinin interacts with autophagic receptors nuclear receptor coactivator 4 (NCOA4) and tax1-binding protein 1 (TAX1BP1), facilitating the formation of ferritin-NCOA4 condensates and inducing ferritinophagy. Further investigation shows that hemagglutinin-induced ferritinophagy causes cellular lipid peroxidation, inhibits aggregation of mitochondrial antiviral signaling protein (MAVS), and suppresses the type I interferon response, thereby contributing to viral replication. Collectively, a novel mechanism by which IAV hemagglutinin induces ferritinophagy resulting in cellular lipid peroxidation, consequently impairing MAVS-mediated antiviral immunity, is revealed.

2.
Vet Microbiol ; 257: 109096, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33940459

RESUMEN

Receptors for activated C kinase 1 (RACK1) could competitively combine with mitochondrial antiviral signaling protein (MAVS) to inhibit the type I interferon (IFN) signaling pathway during viral infection in vitro. However, whether RACK1 can degrade MAVS to enhance viral replication is still unknown. In this study, we found that bovine epidemic fever virus (BEFV) infection triggered the expression of RACK1. Overexpression of RACK1 promoted BEFV replication, while knockdown of RACK1 inhibited the replication of BEFV. Further research showed that RACK1 inhibited the type I IFN signaling pathway during BEFV infection by degrading MAVS, and RACK1 degraded MAVS via the ubiquitin-proteasome system. Mechanistically, RACK1 up-regulated the expression of E3 ubiquitin ligase STIP1 homology and U-box containing protein 1 (STUB1), thereby promoting the ubiquitination and degradation of MAVS. In addition, RACK1 degraded MAVS by enhancing the interaction between STUB1 and MAVS but not via its interaction with STUB1. Overall, our study reveals a novel mechanism by which RACK1 inhibits the type I IFN signaling pathway to BEFV infection through degradation of MAVS, thereby promoting viral infection. These findings provide a new perspective for the MAVS degradation regulated by RACK1.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Virus de la Fiebre Efímera Bovina/fisiología , Inmunidad Innata , Receptores de Cinasa C Activada/genética , Ubiquitina-Proteína Ligasas/genética , Regulación hacia Arriba , Replicación Viral/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Bovinos , Línea Celular , Cricetinae , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Células HEK293 , Humanos , Interferón Tipo I/antagonistas & inhibidores , Interferón Tipo I/inmunología , Transducción de Señal/inmunología
3.
Viruses ; 13(3)2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33807534

RESUMEN

The mitochondrial antiviral-signaling protein (MAVS, also known as VISA, IPS-1, or CARDIF) plays an essential role in the type I interferon (IFN) response and in retinoic acid-inducible gene I (RIG-I) mediated antiviral innate immunity in mammals. In this study, the caprine MAVS gene (caMAVS, 1566 bp) was identified and cloned. The caMAVS shares the highest amino acid similarity (98.1%) with the predicted sheep MAVS. Confocal microscopy analysis of partial deletion mutants of caMAVS revealed that the transmembrane and the so-called Non-Characterized domains are indispensable for intracellular localization to mitochondria. Overexpression of caMAVS in caprine endometrial epithelial cells up-regulated the mRNA levels of caprine interferon-stimulated genes. We concluded that caprine MAVS mediates the activation of the type I IFN pathway. We further demonstrated that both the CARD-like domain and the transmembrane domain of caMAVS were essential for the activation of the IFN-ß promotor. The interaction between caMAVS and caprine RIG-I and the vital role of the CARD and NC domain in this interaction was demonstrated by co-immunoprecipitation. Upon infection with the Peste des Petits Ruminants Virus (PPRV, genus Morbillivirus), the level of MAVS was greatly reduced. This reduction was prevented by the addition of the proteasome inhibitor MG132. Moreover, we found that viral protein V could interact and colocalize with MAVS. Together, we identified caMAVS as a RIG-I interactive protein involved in the activation of type I IFN pathways in caprine cells and as a target for PPRV immune evasion.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Inductores de Interferón/inmunología , Peste de los Pequeños Rumiantes/inmunología , Virus de la Peste de los Pequeños Rumiantes/inmunología , Animales , Chlorocebus aethiops , Células Epiteliales , Cabras , Células HEK293 , Humanos , Interferón Tipo I/inmunología , Células Vero
4.
J Biol Chem ; 295(40): 13862-13874, 2020 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-32747444

RESUMEN

Inhibitors against the NS3-4A protease of hepatitis C virus (HCV) have proven to be useful drugs in the treatment of HCV infection. Although variants have been identified with mutations that confer resistance to these inhibitors, the mutations do not restore replicative fitness and no secondary mutations that rescue fitness have been found. To gain insight into the molecular mechanisms underlying the lack of fitness compensation, we screened known resistance mutations in infectious HCV cell culture with different genomic backgrounds. We observed that the Q41R mutation of NS3-4A efficiently rescues the replicative fitness in cell culture for virus variants containing mutations at NS3-Asp168 To understand how the Q41R mutation rescues activity, we performed protease activity assays complemented by molecular dynamics simulations, which showed that protease-peptide interactions far outside the targeted peptide cleavage sites mediate substrate recognition by NS3-4A and support protease cleavage kinetics. These interactions shed new light on the mechanisms by which NS3-4A cleaves its substrates, viral polyproteins and a prime cellular antiviral adaptor protein, the mitochondrial antiviral signaling protein MAVS. Peptide binding is mediated by an extended hydrogen-bond network in NS3-4A that was effectively optimized for protease-MAVS binding in Asp168 variants with rescued replicative fitness from NS3-Q41R. In the protease harboring NS3-Q41R, the N-terminal cleavage products of MAVS retained high affinity to the active site, rendering the protease susceptible for potential product inhibition. Our findings reveal delicately balanced protease-peptide interactions in viral replication and immune escape that likely restrict the protease adaptive capability and narrow the virus evolutionary space.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Hepacivirus/fisiología , Simulación de Dinámica Molecular , Inhibidores de Proteasas/farmacología , Replicación Viral/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Sustitución de Aminoácidos , Línea Celular Tumoral , Humanos , Mutación Missense , Serina Proteasas/química , Serina Proteasas/genética , Serina Proteasas/metabolismo , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/genética
5.
Int J Mol Sci ; 20(15)2019 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-31382586

RESUMEN

Peroxisomes are ubiquitous organelles with well-defined functions in lipid and reactive oxygen species metabolism, having a significant impact on a large number of important diseases. Growing evidence points to them, in concert with mitochondria, as important players within the antiviral response. In this review we summarize and discuss the recent findings concerning the relevance of peroxisomes within innate immunity. We not only emphasize their importance as platforms for cellular antiviral signaling but also review the current information concerning their role in the control of bacterial infections. We furthermore review the recent data that pinpoints peroxisomes as regulators of inflammatory processes.


Asunto(s)
Infecciones Bacterianas/inmunología , Inmunidad Innata , Peroxisomas/inmunología , Proteínas Adaptadoras Transductoras de Señales/inmunología , Antivirales/uso terapéutico , Infecciones Bacterianas/microbiología , Infecciones Bacterianas/virología , Humanos , Peroxisomas/microbiología , Peroxisomas/virología , Especies Reactivas de Oxígeno/química , Especies Reactivas de Oxígeno/inmunología
6.
BMC Microbiol ; 18(1): 224, 2018 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-30587113

RESUMEN

BACKGROUND: Bovine ephemeral fever virus (BEFV), the causative agent of bovine ephemeral fever, is an economically important pathogen of cattle and water buffalo. MicroRNAs (miRNAs) are endogenous 21-23 nt small non-coding RNA molecules that binding to a multiple of target mRNAs and functioning in the regulation of viral replication including the miRNA-mediated antiviral defense. However, the reciprocal interaction between bovine ephemeral fever virus replication and host miRNAs still remain poorly understood. The aim of our study herein was to investigate the exact function of miR-3470b and its molecular mechanisms during BEFV infection. RESULTS: In this study, we found a set of microRNAs induced by BEFV infection using small RNA deep sequencing, and further identified BEFV infection could significantly up-regulate the miR-3470b expression in Baby Hamster Syrian Kidney cells (BHK-21) after 24 h and 48 h post-infection (pi) compared to normal BHK-21 cells without BEFV infection. Additionally, the target association between miR-3470b and mitochondrial antiviral signaling protein (MAVS) was predicted by target gene prediction tools and further validated using a dual-luciferase reporter assay, and the expression of MAVS mRNA and protein levels was negatively associated with miR-3470b levels. Furthermore, the miR-3470b mimic transfection significantly contributed to increase the BEFV N mRNA, G protein level and viral titer, respectively, whereas the miR-3470b inhibitor had the opposite effect on BEFV replication. Moreover, the overexpression of MAVS or silencing of miR-3470b by its inhibitors suppressed BEFV replication, and knockdown of MAVS by small interfering RNA also promoted the replication of BEFV. CONCLUSIONS: Our findings is the first to reveal that miR-3470b as a novel host factor regulates BEFV replication via directly targeting the MAVS gene in BHK-21 cells and may provide a potential strategy for developing effective antiviral therapy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Virus de la Fiebre Efímera Bovina/fisiología , Fiebre Efímera/inmunología , Fiebre Efímera/virología , Riñón/inmunología , MicroARNs/genética , Replicación Viral , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Bovinos , Cricetinae , Fiebre Efímera/genética , Virus de la Fiebre Efímera Bovina/genética , Interacciones Huésped-Patógeno , Riñón/virología , Mesocricetus , MicroARNs/inmunología , Conejos
7.
Methods Mol Biol ; 1714: 237-247, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29177867

RESUMEN

During infection, the cytosolic detection of viral double-stranded RNA (dsRNA) leads to the oligomerization and activation of mitochondrial antiviral signaling protein (MAVS) and the subsequent production of type I interferon (IFN). Here, we describe a novel method of visualizing and quantifying the aggregation of MAVS in response to dsRNA stimulation or viral infection in vitro using confocal microscopy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Interferón Tipo I/metabolismo , Lentivirus/genética , Microscopía Confocal/métodos , ARN Bicatenario/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/ultraestructura , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Células HEK293 , Humanos , Ratones , Multimerización de Proteína
8.
Biochem Biophys Res Commun ; 446(2): 523-8, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24613846

RESUMEN

Mitochondrial antiviral-signaling protein (MAVS), as a critical adaptor of RIG-I signaling, bridges viral RNA recognition and downstream signal activation. However, the regulating mechanisms of MAVS are not well understood. In this study, we demonstrated that eukaryotic elongation factor 1B gamma (eEF1Bγ) activates NF-кB signaling pathway through targeting MAVS. GST-pull down and mass spectrometric analysis suggested that eEF1Bγ binds to the CARD domain of MAVS. The interaction and mitochondrial colocalization of eEF1Bγ and MAVS were further verified by co-immunoprecipitation (co-IP) and immunofluorescence microscopy assays. The dual-luciferase assays showed that ectopic expression of eEF1Bγ significantly promotes the activities of transcription factor NF-кB and promoters of downstream proinflammatory cytokines Interleukin-8 (IL-8) and Interleukin-6 (IL-6). eEF1Bγ increases the abundance of MAVS by promoting its K63-linked polyubiquitination and attenuating its K48-linked polyubiquitination. Besides, proline-rich (Pro) region and CARD domain of MAVS are indispensable for the process of eEF1Bγ mediated ubiquitination. Collectively, these results demonstrated that eEF1Bγ functions as a positive regulator of NF-кB signal by targeting MAVS for activation, which provides a new regulating mechanism of antiviral responses.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , FN-kappa B/metabolismo , Factor 1 de Elongación Peptídica/metabolismo , Transducción de Señal/fisiología , Células HEK293 , Células HeLa , Humanos , Regulación hacia Arriba/fisiología
9.
Mol Immunol ; 57(2): 100-10, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24096085

RESUMEN

Measles virus (MV) infects CD150Tg/Ifnar (IFN alpha receptor)(-/-) mice but not CD150 (a human MV receptor)-transgenic (Tg) mice. We have shown that bone marrow-derived dendritic cells (BMDCs) from CD150Tg/Ifnar(-/-) mice are permissive to MV in contrast to those from simple CD150Tg mice, which reveals a crucial role of type I interferon (IFN) in natural tropism against MV. Yet, the mechanism whereby BMDCs produce initial type I IFN has not been elucidated in MV infection. RNA virus infection usually allows cells to generate double-stranded RNA and induce activation of IFN regulatory factor (IRF) 3/7 transcription factors, leading to the production of type I IFN through the retinoic acid-inducible gene I (RIG-I)/melanoma differentiation-associated gene 5 (MDA5)-mitochondrial antiviral signaling protein (MAVS) pathway. In mouse experimental BMDCs models, we found CD150Tg/Mavs(-/-)BMDCs, but not CD150Tg/Irf3(-/-)/Irf7(-/-)BMDCs, permissive to MV. IFN-α/ß were not induced in MV-infected CD150Tg/Mavs(-/-)BMDCs, while IFN-ß was subtly induced in CD150Tg/Irf3(-/-)/Irf7(-/-)BMDCs. In vivo systemic infection was therefore established by transfer of MV-infected CD150Tg/Mavs(-/-) BMDCs to CD150Tg/Ifnar(-/-) mice. These data indicate that MAVS-dependent, IRF3/7-independent IFN-ß induction triggers the activation of the IFNAR pathway so as to restrict the spread of MV by infected BMDCs. Hence, MAVS participates in the initial induction of type I IFN in BMDCs and IFNAR protects against MV spreading. We also showed the importance of IL-10-producing CD4(+) T cells induced by MV-infected BMDCs in vitro, which may account for immune modulation due to the functional aberration of DCs.


Asunto(s)
Células Dendríticas/inmunología , Factor 3 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/genética , Interferón beta/metabolismo , Sarampión/inmunología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/deficiencia , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Antígenos CD/metabolismo , Células de la Médula Ósea/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Línea Celular , Chlorocebus aethiops , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/metabolismo , Células Dendríticas/citología , Factor 3 Regulador del Interferón/deficiencia , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/deficiencia , Factor 7 Regulador del Interferón/metabolismo , Helicasa Inducida por Interferón IFIH1 , Interferón beta/biosíntesis , Interleucina-10/metabolismo , Virus del Sarampión/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Interferón alfa y beta/genética , Receptores de Superficie Celular/metabolismo , Transducción de Señal/inmunología , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA