Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Biomolecules ; 11(8)2021 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-34439917

RESUMEN

Melanoma differentiation-associated protein 5 (MDA5) is a crucial RIG-I-like receptor RNA helicase enzyme encoded by IFIH1 in humans. Single nucleotide polymorphisms in the IFIH1 results in fatal genetic disorders such as Aicardi-Goutières syndrome and Singleton-Merten syndrome, and in increased risk of type I diabetes in humans. In this study, we chose four different amino acid substitutions of the MDA5 protein responsible for genetic disorders: MDA5L372F, MDA5A452T, MDA5R779H, and MDA5R822Q and analyzed their structural and functional relationships using molecular dynamic simulations. Our results suggest that the mutated complexes are relatively more stable than the wild-type MDA5. The radius of gyration, interaction energies, and intra-hydrogen bond analysis indicated the stability of mutated complexes over the wild type, especially MDA5L372F and MDA5R822Q. The dominant motions exhibited by the wild-type and mutant complexes varied significantly. Moreover, the betweenness centrality of the wild-type and mutant complexes showed shared residues for intra-signal propagation. The observed results indicate that the mutations lead to a gain of function, as reported in previous studies, due to increased interaction energies and stability between RNA and MDA5 in mutated complexes. These findings are expected to deepen our understanding of MDA5 variants and may assist in the development of relevant therapeutics against the disorders.


Asunto(s)
Enfermedades de la Aorta/genética , Enfermedades Autoinmunes del Sistema Nervioso/genética , Hipoplasia del Esmalte Dental/genética , Helicasa Inducida por Interferón IFIH1/genética , Metacarpo/anomalías , Enfermedades Musculares/genética , Mutación , Malformaciones del Sistema Nervioso/genética , Odontodisplasia/genética , Osteoporosis/genética , Calcificación Vascular/genética , Biología Computacional , Humanos , Enlace de Hidrógeno , Helicasa Inducida por Interferón IFIH1/fisiología , Conformación Molecular , Simulación de Dinámica Molecular , Proteínas Mutantes/genética , Mutación Missense , Fenotipo , Análisis de Componente Principal , ARN/metabolismo , Termodinámica
3.
Cell Death Dis ; 11(8): 718, 2020 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-32879301

RESUMEN

The RIG-I-like receptor (RLR) melanoma differentiation-associated gene 5 (MDA5) plays a key role in triggering innate antiviral response during infection by RNA viruses. MDA5 activation leads to transcription induction of type-I interferon (IFN) and proinflammatory cytokines. MDA5 has also been associated with autoimmune and autoinflammatory diseases by dysfunctional activation of innate immune response in the absence of infection. Here, we show how foot-and-mouth disease virus (FMDV) counteracts the specific antiviral effect exerted by MDA5 targeting the protein for cleavage by the viral Leader protease (Lpro). MDA5 overexpression had an inhibitory effect on FMDV infection in IFN-competent cells. Remarkably, immunostimulatory viral RNA co-immunoprecipitated with MDA5 in infected cells. Moreover, specific cleavage of MDA5 by Lpro was detected in co-transfected cells, as well as during the course of FMDV infection. A significant reduction in IFN induction associated with MDA5 cleavage was detected by comparison with a non-cleavable MDA5 mutant protein with preserved antiviral activity. The Lpro cleavage site in MDA5 was identified as the RGRAR sequence in the conserved helicase motif VI, coinciding with that recently reported for Lpro in LGP2, another member of the RLRs family involved in antiviral defenses. Interestingly, specific mutations within the MDA5 Lpro target sequence have been associated with immune disease in mice and humans. Our results reveal a pleiotropic strategy for immune evasion based on a viral protease targeting phylogenetically conserved domains of immune sensors. Identification of viral strategies aimed to disrupt MDA5 functionality may also contribute to develop new treatment tools for MDA5-related disorders.


Asunto(s)
Endopeptidasas/metabolismo , Virus de la Fiebre Aftosa/metabolismo , Helicasa Inducida por Interferón IFIH1/metabolismo , Animales , Línea Celular , Proteína 58 DEAD Box/metabolismo , Endopeptidasas/genética , Virus de la Fiebre Aftosa/genética , Pleiotropía Genética/genética , Células HEK293 , Humanos , Inmunidad Innata , Interferón Tipo I/metabolismo , Helicasa Inducida por Interferón IFIH1/fisiología , Proteolisis , ARN Viral/inmunología , Receptores Inmunológicos/metabolismo , Transducción de Señal , Porcinos
4.
Front Immunol ; 11: 678, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32425931

RESUMEN

The innate immune system, which senses invading pathogens, plays a critical role as the first line of host defense. After recognition of foreign RNA ligands (e.g., RNA viruses), host cells generate an innate immune or antiviral response via the interferon-mediated signaling pathway. Retinoic acid-inducible gene I (RIG-1) acts as a major sensor that recognizes a broad range of RNA ligands in mammals; however, chickens lack a RIG-1 homolog, meaning that RNA ligands should be recognized by other cellular sensors such as melanoma differentiation-associated protein 5 (MDA5) and toll-like receptors (TLRs). However, it is unclear which of these cellular sensors compensates for the loss of RIG-1 to act as the major sensor for RNA ligands. Here, we show that chicken MDA5 (cMDA5), rather than chicken TLRs (cTLRs), plays a pivotal role in the recognition of RNA ligands, including poly I:C and influenza virus. First, we used a knockdown approach to show that both cMDA5 and cTLR3 play roles in inducing interferon-mediated innate immune responses against RNA ligands in chicken DF-1 cells. Furthermore, targeted knockout of cMDA5 or cTLR3 in chicken DF-1 cells revealed that loss of cMDA5 impaired the innate immune responses against RNA ligands; however, the responses against RNA ligands were retained after loss of cTLR3. In addition, double knockout of cMDA5 and cTLR3 in chicken DF-1 cells abolished the innate immune responses against RNA ligands, suggesting that cMDA5 is the major sensor whereas cTLR3 is a secondary sensor. Taken together, these findings provide an understanding of the functional role of cMDA5 in the recognition of RNA ligands in chicken DF-1 cells and may facilitate the development of an innate immune-deficient cell line or chicken model.


Asunto(s)
Inmunidad Innata , Helicasa Inducida por Interferón IFIH1/fisiología , ARN Bicatenario/metabolismo , Receptor Toll-Like 3/fisiología , Animales , Línea Celular , Pollos , Proteína 58 DEAD Box/fisiología , Fibroblastos/inmunología , Interferón beta/genética , Ligandos , Orthomyxoviridae/fisiología , Poli I-C/farmacología , Regiones Promotoras Genéticas , Replicación Viral
5.
J Immunol ; 203(9): 2508-2519, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31548332

RESUMEN

IFN responses to viral infection are necessary to establish intrinsic antiviral state, but if unchecked can lead to heightened inflammation. Recently, we showed that TLR2 activation contributes to limitation of rhinovirus (RV)-induced IFN response in the airway epithelial cells. We also demonstrated that compared with normal airway epithelial cells, those from patients with chronic obstructive pulmonary disease (COPD) show higher IFN responses to RV, but the underlying mechanisms are not known. Initially, RV-induced IFN responses depend on dsRNA receptor activation and then are amplified via IFN-stimulated activation of JAK/STAT signaling. In this study, we show that in normal cells, TLR2 limits RV-induced IFN responses by attenuating STAT1 and STAT2 phosphorylation and this was associated with TLR2-dependent SIRT-1 expression. Further, inhibition of SIRT-1 enhanced RV-induced IFN responses, and this was accompanied by increased STAT1/STAT2 phosphorylation, indicating that TLR2 may limit RV-induced IFN responses via SIRT-1. COPD airway epithelial cells showed attenuated IL-8 responses to TLR2 agonist despite expressing TLR2 similar to normal, indicating dysregulation in TLR2 signaling pathway. Unlike normal, COPD cells failed to show RV-induced TLR2-dependent SIRT-1 expression. Pretreatment with quercetin, which increases SIRT-1 expression, normalized RV-induced IFN levels in COPD airway epithelial cells. Inhibition of SIRT-1 in quercetin-pretreated COPD cells abolished the normalizing effects of quercetin on RV-induced IFN expression in these cells, confirming that quercetin exerts its effect via SIRT-1. In summary, we show that TLR2 is required for limiting RV-induced IFNs, and this pathway is dysregulated in COPD airway epithelial cells, leading to exaggerated IFN production.


Asunto(s)
Bronquios/inmunología , Interferones/biosíntesis , Enfermedad Pulmonar Obstructiva Crónica/etiología , Rhinovirus/patogenicidad , Sirtuina 1/fisiología , Receptor Toll-Like 2/fisiología , Células Cultivadas , Células Epiteliales , Humanos , Helicasa Inducida por Interferón IFIH1/fisiología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , ARN Bicatenario/fisiología , Factores de Transcripción STAT/fisiología , Transducción de Señal/fisiología , Sirtuina 1/genética , Proteína 1 Supresora de la Señalización de Citocinas/fisiología
6.
PLoS One ; 14(5): e0216056, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31067281

RESUMEN

The airway epithelial barrier is critical for preventing pathogen invasion and translocation of inhaled particles into the lung. Epithelial cells also serve an important sentinel role after infection and release various pro-inflammatory mediators that recruit and activate immune cells. Airway epithelial barrier disruption has been implicated in a growing number of respiratory diseases including viral infections. It is thought that when a pathogen breaks the barrier and gains access to the host tissue, pro-inflammatory mediators increase, which further disrupts the barrier and initiates a vicious cycle of leak. However, it is difficult to study airway barrier integrity in vivo, and little is known about relationship between epithelial barrier function and airway inflammation. Current assays of pulmonary barrier integrity quantify the leak of macromolecules from the vasculature into the airspaces (or "inside/out" leak). However, it is also important to measure the ease with which inhaled particles, allergens, or pathogens can enter the subepithelial tissues (or "outside/in" leak). We challenged mice with inhaled double stranded RNA (dsRNA) and explored the relationship between inside/out and outside/in barrier function and airway inflammation. Using wild-type and gene-targeted mice, we studied the roles of the dsRNA sensors Toll Like Receptor 3 (TLR3) and Melanoma Differentiation-Associated protein 5 (MDA5). Here we report that after acute challenge with inhaled dsRNA, airway barrier dysfunction occurs in a TLR3-dependent manner, whereas leukocyte accumulation is largely MDA5-dependent. We conclude that airway barrier dysfunction and inflammation are regulated by different mechanisms at early time points after exposure to inhaled dsRNA.


Asunto(s)
Inflamación/inducido químicamente , Helicasa Inducida por Interferón IFIH1/fisiología , ARN Bicatenario/farmacología , Mucosa Respiratoria/efectos de los fármacos , Receptor Toll-Like 3/fisiología , Administración por Inhalación , Animales , Líquido del Lavado Bronquioalveolar/química , Quimiocina CCL3/análisis , Femenino , Inflamación/metabolismo , Inflamación/fisiopatología , Interferón gamma/análisis , Interleucina-6/análisis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Bicatenario/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/fisiología
7.
J Virol ; 92(17)2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29899107

RESUMEN

Emerging evidence indicates that long noncoding RNAs (lncRNAs) regulate various biological processes, especially innate and adaptive immunity. However, the relationship between lncRNAs and the interferon (IFN) pathway remains largely unknown. Here, we report that lncRNA ITPRIP-1 (lncITPRIP-1) is involved in viral infection and plays a crucial role in the virus-triggered IFN signaling pathway through the targeting of melanoma differentiation-associated gene 5 (MDA5). LncITPRIP-1 can be induced by viral infection, which is not entirely dependent on the IFN signal. Besides, there is no coding potential found in the lncITPRIP-1 transcript. LncITPRIP-1 binds to the C terminus of MDA5, and it possesses the ability to boost the oligomerization of both the full length and the 2 caspase activation and recruitment domains of MDA5 in a K63-linked polyubiquitination-independent manner. Amazingly, we also found that MDA5 can suppress hepatitis C virus (HCV) replication independently of IFN signaling through its C-terminal-deficient domain bound to viral RNA, in which lncITPRIP-1 plays a role as an assistant. In addition, the expression of lncITPRIP-1 is highly consistent with MDA5 expression, indicating that lncITPRIP-1 may function as a cofactor of MDA5. All the data suggest that lncITPRIP-1 enhances the innate immune response to viral infection through the promotion of oligomerization and activation of MDA5. Our study discovers the first lncRNA ITPRIP-1 involved in MDA5 activation.IMPORTANCE Hepatitis C virus infection is a global health issue, and there is still no available vaccine, which makes it urgent to reveal the underlying mechanisms of HCV and host factors. Although RIG-I has been recognized as the leading cytoplasmic sensor against HCV for a long time, recent findings that MDA5 regulates the IFN response to HCV have emerged. Our work validates the significant role of MDA5 in IFN signaling and HCV infection and proposes the first lncRNA inhibiting HCV replication by promoting the activation of MDA5 and mediating the association between MDA5 and HCV RNA, the study of which may shed light on the MDA5 function and treatment for hepatitis C patients. Our suggested model of how lncITPRIP-1 orchestrates signal transduction for IFN production illustrates the essential role of lncRNAs in virus elimination.


Asunto(s)
Inmunidad Innata/fisiología , Helicasa Inducida por Interferón IFIH1/genética , Interferones/inmunología , Proteínas de la Membrana/fisiología , ARN Largo no Codificante/fisiología , Transducción de Señal/inmunología , Línea Celular , Regulación de la Expresión Génica/inmunología , Hepacivirus/genética , Hepacivirus/inmunología , Hepatitis C/inmunología , Hepatitis C/virología , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/inmunología , Helicasa Inducida por Interferón IFIH1/fisiología , Interferones/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , ARN Viral/genética , Transducción de Señal/genética
8.
Cancer Immunol Immunother ; 67(7): 1091-1103, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29696308

RESUMEN

Cytotoxic T lymphocytes (CTLs) are effective components of the immune system capable of destroying tumor cells. Generation of CTLs using peptide vaccines is a practical approach to treat cancer. We have previously described a peptide vaccination strategy that generates vast numbers of endogenous tumor-reactive CTLs after two sequential immunizations (prime-boost) using poly-ICLC adjuvant, which stimulates endosomal toll-like receptor 3 (TLR3) and cytoplasmic melanoma differentiation antigen 5 (MDA5). Dendritic cells (DCs) play an important role not only in antigen presentation but are critical in generating costimulatory cytokines that promote CTL expansion. Poly-ICLC was shown to be more effective than poly-IC in generating type-I interferon (IFN-I) in various DC subsets, through its enhanced ability to escape the endosomal compartment and stimulate MDA5. In our system, IFN-I did not directly function as a T cell costimulatory cytokine, but enhanced CTL expansion through the induction of IL15. With palmitoylated peptide vaccines, CD8α+ DCs were essential for peptide crosspresentation. For vaccine boosts, non-professional antigen-presenting cells were able to present minimal epitope peptides, but DCs were still required for CTL expansions through the production of IFN-I mediated by poly-ICLC. Overall, these results clarify the roles of DCs, TLR3, MDA5, IFN-I and IL15 in the generation of vast and effective antitumor CTL responses using peptide and poly-IC vaccines.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Células Dendríticas/inmunología , Interferón Tipo I/metabolismo , Helicasa Inducida por Interferón IFIH1/fisiología , Melanoma Experimental/terapia , Linfocitos T Citotóxicos/inmunología , Vacunas de Subunidad/administración & dosificación , Animales , Carboximetilcelulosa de Sodio/administración & dosificación , Carboximetilcelulosa de Sodio/análogos & derivados , Células Dendríticas/efectos de los fármacos , Inductores de Interferón/administración & dosificación , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Poli I-C/administración & dosificación , Polilisina/administración & dosificación , Polilisina/análogos & derivados , Linfocitos T Citotóxicos/efectos de los fármacos , Receptor Toll-Like 3/fisiología , Células Tumorales Cultivadas , Vacunación
9.
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
10.
J Leukoc Biol ; 103(2): 185-192, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28974542

RESUMEN

IFNs protect us against infection from viral pathogens, but can also induce damaging inflammation and are associated with the development of autoimmune conditions. By dissecting the response that is mediated by different IFN-regulated genes, we hoped to identify targets that will enable us to preserve the defense against pathogens while minimizing immune disease. Toward this, several reports have identified that variability in the gene that encodes the melanoma differentiation-associated protein (MDA)-5 and other molecules in this pathway correlated with the risk of autoimmune diseases. The evidence for MDA5 activity as a cause of autoimmune disease is discussed.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Helicasa Inducida por Interferón IFIH1/fisiología , Animales , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata/inmunología , Interferones/metabolismo , Virus/inmunología
11.
J Exp Med ; 214(2): 459-473, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28031478

RESUMEN

MDA5 plays a critical role in antiviral innate immunity by functioning as a cytoplasmic double-stranded RNA sensor that can activate type I interferon signaling pathways, but the mechanism for the activation of MDA5 is poorly understood. Here, we show that TRIM65 specifically interacts with MDA5 and promotes K63-linked ubiquitination of MDA5 at lysine 743, which is critical for MDA5 oligomerization and activation. Trim65 deficiency abolishes MDA5 agonist or encephalomyocarditis virus (EMCV)-induced interferon regulatory factor 3 (IRF3) activation and type I interferon production but has no effect on retinoic acid-inducible I (RIG-I), Toll-like receptor 3 (TLR3), or cyclic GMP-AMP synthase signaling pathways. Importantly, Trim65-/- mice are more susceptible to EMCV infection than controls and cannot produce type I interferon in vivo. Collectively, our results identify TRIM65 as an essential component for the MDA5 signaling pathway and provide physiological evidence showing that ubiquitination is important for MDA5 oligomerization and activation.


Asunto(s)
Infecciones por Cardiovirus/inmunología , Virus de la Encefalomiocarditis , Inmunidad Innata , Helicasa Inducida por Interferón IFIH1/fisiología , Proteínas de Motivos Tripartitos/fisiología , Ubiquitina-Proteína Ligasas/fisiología , Ubiquitinación , Animales , Humanos , Helicasa Inducida por Interferón IFIH1/química , Ratones , Ratones Endogámicos C57BL , Multimerización de Proteína , Transducción de Señal
12.
Vet Res ; 47(1): 74, 2016 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-27449021

RESUMEN

Avian Tembusu virus (ATMUV) is a newly emerged flavivirus that belongs to the Ntaya virus group. ATMUV is a highly pathogenic virus causing significant economic loss to the Chinese poultry industry. However, little is known about the role of host innate immune mechanism in defending against ATMUV infection. In this study, we found that ATMUV infection significantly up-regulated the expression of type I and type III interferons (IFN) and some critical IFN-stimulated genes (ISG) in vivo and in vitro. This innate immune response was induced by genomic RNA of ATMUV. Furthermore, we observed that ATMUV infection triggered IFN response mainly through MDA5 and TLR3-dependent signaling pathways. Strikingly, shRNA-based disruption of IPS-1, IRF3 or IRF7 expression significantly reduced the production of IFN in the 293T cell model. Moreover, NF-κB was shown to be activated in both chicken and human cells during the ATMUV infection. Inhibition of NF-κB signaling also resulted in a clear decrease in expression of IFN. Importantly, experiments revealed that treatment with IFN significantly impaired ATMUV replication in the chicken cell. Consistently, type I IFN also exhibited promising antiviral activity against ATMUV replication in the human cell. Together, these data indicate that ATMUV infection triggers host innate immune response through MDA5 and TLR3-dependent signaling that controls IFN production, and thereby induces an effective antiviral immunity.


Asunto(s)
Infecciones por Flavivirus/veterinaria , Flavivirus/inmunología , Helicasa Inducida por Interferón IFIH1/fisiología , Enfermedades de las Aves de Corral/virología , Transducción de Señal/inmunología , Receptor Toll-Like 3/fisiología , Animales , Embrión de Pollo/virología , Pollos/inmunología , Pollos/virología , Infecciones por Flavivirus/inmunología , Infecciones por Flavivirus/virología , Inmunidad Innata/inmunología , Interferones/fisiología , Enfermedades de las Aves de Corral/inmunología , ARN Viral/genética , Reacción en Cadena en Tiempo Real de la Polimerasa/veterinaria
13.
Cancer Res ; 76(8): 2166-76, 2016 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-26893477

RESUMEN

Melanoma differentiation-associated gene 5 (MDA-5, IFIH1), a cytosolic innate pattern recognition receptor, functions as a first line of defense against viral infection by sensing double-stranded RNA. Ectopic expression of MDA-5 has been shown to induce cancer cell death, but the mechanism of action by which MDA-5 exerts these cytotoxic effects is unclear. Here, we demonstrate that ectopic expression of MDA-5 via replication-incompetent adenovirus (Ad.Mda-5) initiates multiple signaling cascades, culminating in cytotoxicity and type I IFN production in mouse and human prostate cancer cells. This intrinsic dual activity of MDA-5 required the adaptor protein IFNß promoter stimulator 1 (IPS-1, MAVS) and could be functionally uncoupled. MDA-5 lacking N-terminal caspase recruitment domains (CARD) engaged an intracellular death program in cancer cells but was unable to efficiently stimulate the expression of IFNß. In contrast to cancer cells susceptible to MDA-5-mediated cytotoxicity, normal cells were highly resistant and instead developed a robust type I IFN response. Strikingly, intratumoral delivery of Ad.Mda-5 led to regression of preestablished prostate cancers and development of long-lasting antitumor immune memory, which was primarily attributed to the activation of tumor-reactive cytotoxic T lymphocytes and/or natural killer cells. Using the CARD-truncated MDA-5 mutant, silencing of IPS-1, and antibody blockade of the IFNα/ß receptor, we further demonstrate that type I IFN signaling was crucial for in situ MDA-5-induced protective antitumor immunity. Therefore, deliberately targeting the evolutionarily conserved MDA-5-IPS-1 antiviral pathway in tumors can provoke parallel tumoricidal and immunostimulatory effects that bridge innate and adaptive immune responses for the therapeutic treatment of cancer. Cancer Res; 76(8); 2166-76. ©2016 AACR.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Adenoviridae/fisiología , Apoptosis/fisiología , Virus Defectuosos/fisiología , Interferón Tipo I/fisiología , Helicasa Inducida por Interferón IFIH1/fisiología , Neoplasias de la Próstata/patología , Animales , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias de la Próstata/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA