Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Nat Microbiol ; 6(4): 467-478, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33727702

RESUMEN

Activation of the RIG-I-like receptors, retinoic-acid inducible gene I (RIG-I) and melanoma differentiation-associated protein 5 (MDA5), establishes an antiviral state by upregulating interferon (IFN)-stimulated genes (ISGs). Among these is ISG15, the mechanistic roles of which in innate immunity still remain enigmatic. In the present study, we report that ISG15 conjugation is essential for antiviral IFN responses mediated by the viral RNA sensor MDA5. ISGylation of the caspase activation and recruitment domains of MDA5 promotes its oligomerization and thereby triggers activation of innate immunity against a range of viruses, including coronaviruses, flaviviruses and picornaviruses. The ISG15-dependent activation of MDA5 is antagonized through direct de-ISGylation mediated by the papain-like protease of SARS-CoV-2, a recently emerged coronavirus that has caused the COVID-19 pandemic. Our work demonstrates a crucial role for ISG15 in the MDA5-mediated antiviral response, and also identifies a key immune evasion mechanism of SARS-CoV-2, which may be targeted for the development of new antivirals and vaccines to combat COVID-19.


Asunto(s)
Proteasas Similares a la Papaína de Coronavirus/metabolismo , Citocinas/metabolismo , Inmunidad Innata , Helicasa Inducida por Interferón IFIH1/antagonistas & inhibidores , SARS-CoV-2/enzimología , SARS-CoV-2/inmunología , Ubiquitinas/metabolismo , Aedes , Animales , Chlorocebus aethiops , Cricetinae , Células HEK293 , Humanos , Helicasa Inducida por Interferón IFIH1/metabolismo , Leucocitos Mononucleares , Ratones , Células Vero
2.
Front Immunol ; 12: 820163, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35116041

RESUMEN

Objective: The effectiveness of rituximab in anti-melanoma differentiation-associated gene 5 (MDA5) dermatomyositis (DM) with interstitial lung disease (ILD) has been explored only in isolated case reports and small series. This paper aims to review the current evidence regarding rituximab (RTX) use in the treatment of ILD related to anti-MDA5 DM (anti-MDA5 DM-ILD). Methods: We conducted a review by searching PubMed, Web of Science, Embase, and Cochrane for articles with information on patients with anti-MDA5 DM and RTX treatment, published until August 2021, in English language. The selected studies listed variation in chest high-resolution computed tomography (HRCT) and/or pulmonary function test (PFT) as a primary outcome, in patients with anti-MDA5 DM-related ILD after using RTX. Results: Of the 145 potentially eligible articles, 17 were selected. The information gathered from a total of 35 patients with anti-MDA5 DM-ILD was reviewed, including 13 men and 22 women. Patient age at onset was 47.60 ± 13.72 years old. A total of 11.43% (4/35) of the patients were found to have chronic ILD (C-ILD) and 88.57% (31/30) exhibited rapidly progressive ILD (RP-ILD). Most patients (29/30) had typical DM rashes. Prior to RTX administration, the majority of patients (27/35) were treated with medium- or high-dose glucocorticoids and at least one additional immunotherapeutic agent. With regard to RTX efficacy for ILD in anti-MDA5 DM, 71.43% (25/35) of the patients responded to treatment. Skin rash also improved in more than half of the patients after RTX treatment. The most common side effects were infections, reported by 37.14% (13/35) of the patients after using RTX. Conclusion: As a CD20 targeting drug, RTX is a promising therapeutic tool for anti-MDA5 DM-ILD, although the risk of infections should be considered before treatment. Further prospective controlled studies are required to evaluate the optimal RTX treatment regimen. Systematic Review Registration: https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42021289714, identifier CRD42021289714.


Asunto(s)
Antirreumáticos/uso terapéutico , Dermatomiositis/tratamiento farmacológico , Dermatomiositis/etiología , Helicasa Inducida por Interferón IFIH1/genética , Enfermedades Pulmonares Intersticiales/complicaciones , Rituximab/uso terapéutico , Adulto , Anciano , Antirreumáticos/farmacología , Biomarcadores , Dermatomiositis/diagnóstico , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Femenino , Humanos , Helicasa Inducida por Interferón IFIH1/antagonistas & inhibidores , Helicasa Inducida por Interferón IFIH1/metabolismo , Enfermedades Pulmonares Intersticiales/diagnóstico , Enfermedades Pulmonares Intersticiales/etiología , Masculino , Persona de Mediana Edad , Terapia Molecular Dirigida , Rituximab/farmacología , Resultado del Tratamiento
5.
J Microbiol Biotechnol ; 29(10): 1665-1674, 2019 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-31581385

RESUMEN

Zika virus (ZIKV) is a mosquito-transmitted, emerging Flavivirus that causes Guillain-Barré syndrome and microcephaly in adults and fetuses, respectively. Since ZIKV was first isolated in 1947, severe outbreaks have occurred at various places worldwide, including Yap Island in 2007, French Polynesia in 2013, and Brazil in 2015. Although incidences of ZIKV infection and dissemination have drastically increased, the mechanisms underlying the pathogenesis of ZIKV have not been sufficiently studied. In addition, despite extensive research, the exact roles of individual ZIKV genes in the viral evasion of the host innate immune responses remain elusive. Besides, it is still possible that more than one ZIKV-encoded protein may negatively affect type I interferon (IFN) induction. Hence, in this study, we aimed to determine the modulations of the IFN promoter activity, induced by the MDA5/RIG-I signaling pathway, by over-expressing individual ZIKV genes. Our results show that two nonstructural proteins, NS2A and NS4A, significantly down-regulated the promoter activity of IFN-ß by inhibiting multiple signaling molecules involved in the activation of IFN-ß. Interestingly, while NS2A suppressed both full-length and constitutively active RIG-I, NS4A had inhibitory activity only on full-length RIG-I. In addition, while NS2A inhibited all forms of IRF3 (full-length, regulatory domain-deficient, and constitutively active), NS4A could not inhibit constitutively active IRF3-5D. Taken together, our results showed that NS2A and NS4A play major roles as antagonists of MDA5/RIG-I-mediated IFN-ß induction and more importantly, these two viral proteins seem to inhibit induction of the type I IFN responses in differential mechanisms. We believe this study expands our understanding regarding the mechanisms via which ZIKV controls the innate immune responses in cells and may pave the way to development of ZIKV-specific therapeutics.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , Helicasa Inducida por Interferón IFIH1/metabolismo , Interferón beta/genética , Proteínas no Estructurales Virales/metabolismo , Virus Zika/inmunología , Proteína 58 DEAD Box/antagonistas & inhibidores , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Evasión Inmune , Factor 3 Regulador del Interferón/antagonistas & inhibidores , Factor 3 Regulador del Interferón/metabolismo , Helicasa Inducida por Interferón IFIH1/antagonistas & inhibidores , Regiones Promotoras Genéticas , Receptores Inmunológicos , Transducción de Señal , Proteínas no Estructurales Virales/genética
6.
Cell Host Microbe ; 26(4): 493-503.e6, 2019 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-31600501

RESUMEN

14-3-3 protein family members facilitate the translocation of RIG-I-like receptors (RLRs) to organelles that mediate downstream RLR signaling, leading to interferon production. 14-3-3ϵ promotes the cytosolic-to-mitochondrial translocation of RIG-I, while 14-3-3η facilitates MDA5 translocation to mitochondria. We show that the NS3 protein of Zika virus (ZIKV) antagonizes antiviral gene induction by RIG-I and MDA5 by binding to and sequestering the scaffold proteins 14-3-3ϵ and 14-3-3η. 14-3-3-binding is mediated by a negatively charged RLDP motif in NS3 that is conserved in ZIKV strains of African and Asian lineages and is similar to the one found in dengue and West Nile viruses. ZIKV NS3 is sufficient to inhibit the RLR-14-3-3ϵ/η interaction and to suppress antiviral signaling. Mutational perturbation of 14-3-3ϵ/η binding in a recombinant ZIKV leads to enhanced innate immune responses and impaired growth kinetics. Our study provides molecular understanding of immune evasion functions of ZIKV, which may guide vaccine and anti-flaviviral therapy development.


Asunto(s)
Proteínas 14-3-3/metabolismo , Evasión Inmune/inmunología , Péptido Hidrolasas/metabolismo , Proteínas Virales/metabolismo , Infección por el Virus Zika/inmunología , Virus Zika/inmunología , Células A549 , Animales , Línea Celular , Chlorocebus aethiops , Proteína 58 DEAD Box/antagonistas & inhibidores , Células HEK293 , Células HeLa , Humanos , Inmunidad Innata/inmunología , Helicasa Inducida por Interferón IFIH1/antagonistas & inhibidores , Interferón beta/inmunología , Mitocondrias/metabolismo , Péptido Hidrolasas/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores Inmunológicos , Serina Endopeptidasas , Células Vero , Proteínas Virales/genética , Virus Zika/genética
7.
Nat Commun ; 9(1): 4841, 2018 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-30451863

RESUMEN

The RIG-I like receptors (RLRs) RIG-I and MDA5 are cytosolic RNA helicases best characterized as restriction factors for RNA viruses. However, evidence suggests RLRs participate in innate immune recognition of other pathogens, including DNA viruses. Kaposi's sarcoma-associated herpesvirus (KSHV) is a human gammaherpesvirus and the etiological agent of Kaposi's sarcoma and primary effusion lymphoma (PEL). Here, we demonstrate that RLRs restrict KSHV lytic reactivation and we demonstrate that restriction is facilitated by the recognition of host-derived RNAs. Misprocessed noncoding RNAs represent an abundant class of RIG-I substrates, and biochemical characterizations reveal that an infection-dependent reduction in the cellular triphosphatase DUSP11 results in an accumulation of select triphosphorylated noncoding RNAs, enabling their recognition by RIG-I. These findings reveal an intricate relationship between RNA processing and innate immunity, and demonstrate that an antiviral innate immune response can be elicited by the sensing of misprocessed cellular RNAs.


Asunto(s)
Proteína 58 DEAD Box/genética , Herpesvirus Humano 8/inmunología , Interacciones Huésped-Patógeno , Helicasa Inducida por Interferón IFIH1/genética , Procesamiento Postranscripcional del ARN , ARN no Traducido/genética , Secuencia de Bases , Línea Celular Tumoral , Proteína 58 DEAD Box/antagonistas & inhibidores , Proteína 58 DEAD Box/inmunología , Fosfatasas de Especificidad Dual/genética , Fosfatasas de Especificidad Dual/inmunología , Perfilación de la Expresión Génica , Células HEK293 , Herpesvirus Humano 8/genética , Humanos , Inmunidad Innata , Helicasa Inducida por Interferón IFIH1/antagonistas & inhibidores , Helicasa Inducida por Interferón IFIH1/inmunología , Linfocitos/inmunología , Linfocitos/virología , Conformación de Ácido Nucleico , Fosforilación , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , ARN no Traducido/inmunología , Receptores Inmunológicos , Transducción de Señal , Activación Viral
8.
PLoS Pathog ; 14(10): e1007298, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30286211

RESUMEN

Natural killer (NK) cells provide the first line of defense against malaria parasite infection. However, the molecular mechanisms through which NK cells are activated by parasites are largely unknown, so is the molecular basis underlying the variation in NK cell responses to malaria infection in the human population. Here, we compared transcriptional profiles of responding and non-responding NK cells following exposure to Plasmodium-infected red blood cells (iRBCs) and identified MDA5, a RIG-I-like receptor involved in sensing cytosolic RNAs, to be differentially expressed. Knockout of MDA5 in responding human NK cells by CRISPR/cas9 abolished NK cell activation, IFN-γ secretion, lysis of iRBCs. Similarly, inhibition of TBK1/IKKε, an effector molecule downstream of MDA5, also inhibited activation of responding NK cells. Conversely, activation of MDA5 by liposome-packaged poly I:C restored non-responding NK cells to lyse iRBCs. We further show that microvesicles containing large parasite RNAs from iRBCs activated NK cells by fusing with NK cells. These findings suggest that NK cells are activated through the MDA5 pathway by parasite RNAs that are delivered to the cytoplasm of NK cells by microvesicles from iRBCs. The difference in MDA5 expression between responding and non-responding NK cells following exposure to iRBCs likely contributes to the variation in NK cell responses to malaria infection in the human population.


Asunto(s)
Micropartículas Derivadas de Células/inmunología , Eritrocitos/inmunología , Helicasa Inducida por Interferón IFIH1/metabolismo , Células Asesinas Naturales/inmunología , Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Sistemas CRISPR-Cas , Células Cultivadas , Citoplasma/metabolismo , Eritrocitos/metabolismo , Eritrocitos/parasitología , Humanos , Helicasa Inducida por Interferón IFIH1/antagonistas & inhibidores , Helicasa Inducida por Interferón IFIH1/genética , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/parasitología , Activación de Linfocitos , Malaria Falciparum/metabolismo , Malaria Falciparum/parasitología , Plasmodium falciparum/aislamiento & purificación
9.
J Interferon Cytokine Res ; 38(8): 319-332, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30130154

RESUMEN

Excessive interferon (IFN) production and signaling can lead to immunological and developmental defects giving rise to autoimmune diseases referred to collectively as "type I interferonopathies." A subset of these diseases is caused by monogenic mutations affecting proteins involved in nucleic acid sensing, homeostasis, and metabolism. Interferonopathic mutations in the cytosolic antiviral sensor MDA5 render it constitutively hyperactive, resulting in chronic IFN production and IFN-stimulated gene expression. Few therapeutic options are available for patients with interferonopathic diseases, but a large number of IFN evasion and antagonism strategies have evolved in viral pathogens that can counteract IFN production and signaling to enhance virus replication. To test the hypothesis that these natural IFN suppressors could be used to subdue the activity of interferonopathic signaling proteins, hyperactive MDA5 variants were assessed for susceptibility to a family of viral MDA5 inhibitors. In this study, Paramyxovirus V proteins were tested for their ability to counteract constitutively active MDA5 proteins. Results indicate that the V proteins are able to bind to and disrupt the signaling activity of these MDA5 proteins, irrespective of their specific mutations, reducing IFN production and IFN-stimulated gene expression to effectively suppress the hyperactive antiviral response.


Asunto(s)
Helicasa Inducida por Interferón IFIH1/antagonistas & inhibidores , Helicasa Inducida por Interferón IFIH1/metabolismo , Proteínas Virales/metabolismo , Células HEK293 , Humanos , Transducción de Señal , Vesiculovirus/química , Vesiculovirus/metabolismo
10.
Medicine (Baltimore) ; 97(26): e11024, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29952940

RESUMEN

RATIONALE: Among all dermatomyositis (DM) patients, antimelanoma differentiation-associated gene 5 antibody (anti-MDA5 Ab) positive patients have significantly poor short-term mortality, whereas they experience less relapses over the long term after the remission. We report the case of a patient with anti-MDA5 Ab-positive clinically amyopathic dermatomyositis (CADM) with the recurrence of interstitial lung disease (ILD) after 7 years of remission. There has been no case report of an anti-MDA5 Ab-positive DM patient with the recurrence of ILD after 7 years of long-term remission. PATIENT CONCERNS: A 70-year-old Japanese woman was diagnosed with anti-MDA5 Ab-positive CADM and ILD. After achieving 7 years long-term remission, she was admitted to our department with erythema on the fingers and interstitial pneumonia. Her anti-MDA5 Ab titer was elevated. DIAGNOSES: We diagnosed recurrent CADM complicated with ILD. INTERVENTIONS: We successfully treated her with 1,000 mg of methyl-prednisolone pulse and intravenous cyclophosphamide therapy followed by prednisolone 50 mg/day and an increase of cyclosporine. OUTCOMES: After that treatment, the patient's skin symptoms and interstitial pneumonia were relieved. All laboratory investigations such as ferritin, the serum markers of interstitial pneumonia (i.e., SP-A, SP-D), and the titer of anti-MDA5 Ab showed signs of improvement. LESSONS: Her case suggests that careful physical examinations and monitoring the serum markers are important even after long-term remission is achieved.


Asunto(s)
Autoanticuerpos/sangre , Dermatomiositis/inmunología , Helicasa Inducida por Interferón IFIH1/antagonistas & inhibidores , Enfermedades Pulmonares Intersticiales/complicaciones , Administración Intravenosa , Anciano , Antiinflamatorios/uso terapéutico , Pueblo Asiatico/etnología , Biomarcadores/sangre , Ciclofosfamida/administración & dosificación , Ciclofosfamida/uso terapéutico , Ciclosporina/administración & dosificación , Ciclosporina/uso terapéutico , Dermatomiositis/complicaciones , Dermatomiositis/diagnóstico , Dermatomiositis/tratamiento farmacológico , Femenino , Ferritinas/sangre , Ferritinas/efectos de los fármacos , Humanos , Inmunosupresores/uso terapéutico , Helicasa Inducida por Interferón IFIH1/inmunología , Enfermedades Pulmonares Intersticiales/diagnóstico por imagen , Enfermedades Pulmonares Intersticiales/tratamiento farmacológico , Enfermedades Pulmonares Intersticiales/inmunología , Metilprednisolona/administración & dosificación , Metilprednisolona/uso terapéutico , Recurrencia , Tomografía Computarizada por Rayos X , Resultado del Tratamiento
11.
Clin Respir J ; 12(8): 2378-2389, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29873202

RESUMEN

BACKGROUND: Acute exacerbation of idiopathic pulmonary fibrosis (AE-IPF) has an extremely poor prognosis. The role of ferritin in the pathogenesis of AE-IPF is not well known while serum ferritin is a key prognostic indicator for patients with clinically amyopathic dermatomyositis with rapidly progressive interstitial pneumonia. OBJECTIVE: To elucidate the clinical importance of serum ferritin in patients with AE-IPF. METHODS: Thirty-seven patients (48 episodes), who were diagnosed with AE-IPF and treated at our hospital between 1997 and 2015, were retrospectively studied. RESULTS: Patients with AE-IPF had significantly higher levels of serum ferritin than baseline levels at the first diagnosis of IPF (P = 0.0017). Receiver operating characteristic analysis showed the cut-off value 174 ng/mL for the separation of AE (area under the curve, 0.700). No patients with AE-IPF were positive for anti- melanoma differentiation-associated gene 5 antibody. Patients with AE-IPF and higher ferritin (≥174 ng/mL) had lower %FVC and %DLCO before AE, and those with much higher ferritin (≥500 ng/mL) had significantly worse prognosis than those with lower ferritin (log-rank, P = 0.024). Immunohistochemical staining in autopsy specimens showed alveolar macrophages that were producing ferritin. Finally, in multivariate Cox proportional hazards analyses, serum ferritin level of ≥500 ng/mL was a significant worse prognostic factor (hazard ratio 5.280, P = 0.046). CONCLUSION: Higher serum ferritin may be related to a worse prognosis in patients with AE-IPF.


Asunto(s)
Ferritinas/sangre , Fibrosis Pulmonar Idiopática/mortalidad , Helicasa Inducida por Interferón IFIH1/antagonistas & inhibidores , Anciano , Anciano de 80 o más Años , Progresión de la Enfermedad , Femenino , Ferritinas/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Fibrosis Pulmonar Idiopática/fisiopatología , Helicasa Inducida por Interferón IFIH1/metabolismo , Macrófagos/patología , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos
12.
Front Immunol ; 9: 711, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29686682

RESUMEN

The innate immune system provides the first line of defense against pathogen infection though also influences pathways involved in cancer immunosurveillance. The innate immune system relies on a limited set of germ line-encoded sensors termed pattern recognition receptors (PRRs), signaling proteins and immune response factors. Cytosolic receptors mediate recognition of danger damage-associated molecular patterns (DAMPs) signals. Once activated, these sensors trigger multiple signaling cascades, converging on the production of type I interferons and proinflammatory cytokines. Recent studies revealed that PRRs respond to nucleic acids (NA) released by dying, damaged, cancer cells, as danger DAMPs signals, and presence of signaling proteins across cancer types suggests that these signaling mechanisms may be involved in cancer biology. DAMPs play important roles in shaping adaptive immune responses through the activation of innate immune cells and immunological response to danger DAMPs signals is crucial for the host response to cancer and tumor rejection. Furthermore, PRRs mediate the response to NA in several vaccination strategies, including DNA immunization. As route of double-strand DNA intracellular entry, DNA immunization leads to expression of key components of cytosolic NA-sensing pathways. The involvement of NA-sensing mechanisms in the antitumor response makes these pathways attractive drug targets. Natural and synthetic agonists of NA-sensing pathways can trigger cell death in malignant cells, recruit immune cells, such as DCs, CD8+ T cells, and NK cells, into the tumor microenvironment and are being explored as promising adjuvants in cancer immunotherapies. In this minireview, we discuss how cGAS-STING and RIG-I-MAVS pathways have been targeted for cancer treatment in preclinical translational researches. In addition, we present a targeted selection of recent clinical trials employing agonists of cytosolic NA-sensing pathways showing how these pathways are currently being targeted for clinical application in oncology.


Asunto(s)
Inmunoterapia , Neoplasias/inmunología , Neoplasias/metabolismo , Ácidos Nucleicos/inmunología , Receptores Inmunológicos/metabolismo , Transducción de Señal , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Citosol/inmunología , Proteína 58 DEAD Box/antagonistas & inhibidores , Evaluación Preclínica de Medicamentos , Humanos , Inmunidad Innata , Helicasa Inducida por Interferón IFIH1/antagonistas & inhibidores , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/metabolismo , Neoplasias/terapia , Ácidos Nucleicos/metabolismo , Transducción de Señal/efectos de los fármacos , Vacunas de ADN/genética , Vacunas de ADN/inmunología
13.
Biol Trace Elem Res ; 181(1): 122-132, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28432527

RESUMEN

This experimental study investigated the effect of Agaricus blazei Murill polysaccharide (ABP) on cadmium (Cd) poisoning on the melanoma differentiation-associated gene 5 (MDA5) signaling pathway and antioxidant function of peripheral blood lymphocytes (PBLs) in chickens. The experiments were divided into four groups: 7-day-old chickens with normal saline (0.2 mL single/day), Cd (140 mg/kg), ABP (30 mg/mL, 0.2 mL single/day), and Cd + ABP(140 mg/kg/day + 0.2 mL ABP). Peripheral blood was collected on the 20th, 40th, and 60th days for each group, and PBLs were separated. We attempted to detect the expression of MDA5, downstream signaling molecules, and convergence protein (interferon promoter-stimulating factor 1); transcription factors (IRF3 and NF-κB); the content of cytokines (IL-1ß, IL-6, TNF-α, and IFN-ß) in PBLs; and the antioxidant index of superoxide dismutase (SOD), malondialdhyde (MDA), and glutathione peroxidase (GSH-Px). The results showed that ABP can reduce the accumulation of Cd in the peripheral blood of chickens; reduce the expression of MDA5 and downstream signaling molecules; and reduce the content of IL-1ß, IL-6, TNF-α, and IFN-ß in PBLs of chickens. The activity of antioxidant enzymes (SOD and GSH-Px) significantly increased, and the content of MDA decreased. These results showed that they have a certain protective effect of ABP on Cd poisoning in chicken PBLs caused by injury.


Asunto(s)
Antioxidantes/farmacología , Intoxicación por Cadmio/tratamiento farmacológico , Helicasa Inducida por Interferón IFIH1/antagonistas & inhibidores , Linfocitos/efectos de los fármacos , Polisacáridos/farmacología , Transducción de Señal/efectos de los fármacos , Agaricus , Animales , Antioxidantes/química , Cadmio/análisis , Pollos , Glutatión Peroxidasa/metabolismo , Helicasa Inducida por Interferón IFIH1/genética , Linfocitos/metabolismo , Polisacáridos/química , Superóxido Dismutasa/metabolismo
14.
J Cell Mol Med ; 21(11): 3087-3099, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28560754

RESUMEN

Interleukin (IL)-6-type cytokines have no direct antiviral activity; nevertheless, they display immune-modulatory functions. Oncostatin M (OSM), a member of the IL-6 family, has recently been shown to induce a distinct number of classical interferon stimulated genes (ISG). Most of them are involved in antigen processing and presentation. However, induction of retinoic acid-inducible gene (RIG)-I-like receptors (RLR) has not been investigated. Here we report that OSM has the capability to induce the expression of the DExD/H-Box RNA helicases RIG-I and melanoma differentiation antigen 5 (MDA5) as well as of the transcription factors interferon regulatory factor (IRF)1, IRF7 and IRF9 in primary fibroblasts. Induction of the helicases depends on tyrosine as well as serine phosphorylation of STAT1. Moreover, we could show that the OSM-induced STAT1 phosphorylation is predominantly counter-regulated by a strong STAT3-dependent SOCS3 induction, as Stat3 as well as Socs3 knock-down results in an enhanced and prolonged helicase and IRF expression. Other factors involved in regulation of STAT1 or IRF1 activity, like protein tyrosine phosphatase, non-receptor type 2 (PTPN2), promyelocytic leukaemia protein (PML) or small ubiquitin-related modifier 1 (SUMO1), play a minor role in OSM-mediated induction of RLR. Remarkably, OSM and interferon-γ (IFN-γ) synergize to mediate transcription of RLR and pre-treatment of fibroblasts with OSM fosters the type I interferon production in response to a subsequent encounter with double-stranded RNA. Together, these findings suggest that the OSM-induced JAK/STAT1 signalling is implicated in virus protection of non-professional immune cells and may cooperate with interferons to enhance RLR expression in these cells.


Asunto(s)
Proteína 58 DEAD Box/genética , Fibroblastos/efectos de los fármacos , Inmunidad Innata , Helicasa Inducida por Interferón IFIH1/genética , Oncostatina M/farmacología , Factor de Transcripción STAT1/genética , Línea Celular Tumoral , Proteína 58 DEAD Box/antagonistas & inhibidores , Proteína 58 DEAD Box/inmunología , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Humanos , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/inmunología , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/inmunología , Helicasa Inducida por Interferón IFIH1/antagonistas & inhibidores , Helicasa Inducida por Interferón IFIH1/inmunología , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/genética , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/inmunología , Interferón gamma/farmacología , Interleucina-6/farmacología , Factor Inhibidor de Leucemia/farmacología , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/genética , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/inmunología , Lipopolisacáridos/farmacología , Pulmón/citología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Cultivo Primario de Células , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores Inmunológicos , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología , Transducción de Señal , Piel/citología , Piel/efectos de los fármacos , Piel/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/inmunología
15.
J Virol ; 91(13)2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28424289

RESUMEN

Coxsackievirus A16 (CV-A16), CV-A6, and enterovirus D68 (EV-D68) belong to the Picornaviridae family and are major causes of hand, foot, and mouth disease (HFMD) and pediatric respiratory disease worldwide. The biological characteristics of these viruses, especially their interplay with the host innate immune system, have not been well investigated. In this study, we discovered that the 3Cpro proteins from CV-A16, CV-A6, and EV-D68 bind melanoma differentiation-associated gene 5 (MDA5) and inhibit its interaction with MAVS. Consequently, MDA5-triggered type I interferon (IFN) signaling in the retinoic acid-inducible gene I-like receptor (RLR) pathway was blocked by the CV-A16, CV-A6, and EV-D68 3Cpro proteins. Furthermore, the CV-A16, CV-A6, and EV-D68 3Cpro proteins all cleave transforming growth factor ß-activated kinase 1 (TAK1), resulting in the inhibition of NF-κB activation, a host response also critical for Toll-like receptor (TLR)-mediated signaling. Thus, our data demonstrate that circulating HFMD-associated CV-A16 and CV-A6, as well as severe respiratory disease-associated EV-D68, have developed novel mechanisms to subvert host innate immune responses by targeting key factors in the RLR and TLR pathways. Blocking the ability of 3Cpro proteins from diverse enteroviruses and coxsackieviruses to interfere with type I IFN induction should restore IFN antiviral function, offering a potential novel antiviral strategy.IMPORTANCE CV-A16, CV-A6, and EV-D68 are emerging pathogens associated with hand, foot, and mouth disease and pediatric respiratory disease worldwide. The pathogenic mechanisms of these viruses are largely unknown. Here we demonstrate that the CV-A16, CV-A6, and EV-D68 3Cpro proteins block MDA5-triggered type I IFN induction. The 3Cpro proteins of these viruses bind MDA5 and inhibit its interaction with MAVS. In addition, the CV-A16, CV-A6, and EV-D68 3Cpro proteins cleave TAK1 to inhibit the NF-κB response. Thus, our data demonstrate that circulating HFMD-associated CV-A16 and CV-A6, as well as severe respiratory disease-associated EV-D68, have developed a mechanism to subvert host innate immune responses by simultaneously targeting key factors in the RLR and TLR pathways. These findings indicate the potential merit of targeting the CV-A16, CV-A6, and EV-D68 3Cpro proteins as an antiviral strategy.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Enterovirus/inmunología , Enterovirus/patogenicidad , Interacciones Huésped-Patógeno , Evasión Inmune , Inmunidad Innata , Helicasa Inducida por Interferón IFIH1/antagonistas & inhibidores , Proteínas Virales/metabolismo , Proteasas Virales 3C , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Proteolisis , Transducción de Señal
16.
Artículo en Inglés | MEDLINE | ID: mdl-28194372

RESUMEN

Avian leukosis virus subgroup J (ALV-J) is an oncogenic retrovirus that has a similar replication cycle to multiple viruses and therefore can be used as a model system for viral entry into host cells. However, there are few reports on the genes or microRNAs (miRNAs) that are responsible for the replication of ALV-J. Our previous miRNA and RNA sequencing data showed that the expression of miR-34b-5p was significantly upregulated in ALV-J-infected chicken spleens compared to non-infected chicken spleens, but melanoma differentiation-associated gene 5 (MDA5) had the opposite expression pattern. In this study, a dual-luciferase reporter assay showed that MDA5 is a direct target of miR-34b-5p. In vitro, overexpression of miR-34b-5p accelerated the proliferation of ALV-J-infected cells by inducing the progression from G2 to S phase and it promoted cell migration. Ectopic expression of MDA5 inhibited ALV-J-infected cell proliferation, the cell cycle and cell migration, and knockdown of MDA5 promoted proliferation, the cell cycle and migration. In addition, during ALV-J infections, MDA5 can detect virus invasion and it triggers the MDA5 signaling pathway. MDA5 overexpression can activate the MDA5 signaling pathway, and thus it can inhibit the mRNA and protein expression of the ALV-J env gene and it can suppress virion secretion. In contrast, in response to the knockdown of MDA5 by small interfering RNA (siRNA) or an miR-34b-5p mimic, genes in the MDA5 signaling pathway were significantly downregulated (P < 0.05), but the mRNA and protein expression of ALV-J env and the sample-to-positive ratio of virion in the supernatants were increased. This indicates that miR-34b-5p is able to trigger the MDA5 signaling pathway and affect ALV-J infections. Together, these results suggest that miR-34b-5p targets MDA5 to accelerate the proliferation and migration of ALV-J-infected cells, and it promotes ALV-J replication, via the MDA5 signaling pathway.


Asunto(s)
Virus de la Leucosis Aviar/fisiología , Proliferación Celular , Interacciones Huésped-Patógeno , Helicasa Inducida por Interferón IFIH1/antagonistas & inhibidores , MicroARNs/metabolismo , Transducción de Señal , Replicación Viral , Animales , Movimiento Celular , Pollos
17.
PLoS One ; 11(4): e0154285, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27115353

RESUMEN

OBJECTIVE: Autoantibodies to melanoma differentiation-associated gene 5 (MDA5) are specifically expressed in patients with dermatomyositis (DM) and are associated with a subset of DM patients with rapidly progressive interstitial lung disease (RP-ILD). Here, we examined the clinical utility of a newly developed enzyme-linked immunosorbent assay (ELISA) system for detecting these antibodies. METHODS: Here we developed an improved ELISA for detecting anti-MDA5 antibodies. We then performed a multicenter clinical study involving 8 medical centers and enrolled 242 adult patients with polymyositis (PM)/DM, 190 with non-PM/DM connective tissue disease (CTD), 154 with idiopathic interstitial pneumonia (IIP), and 123 healthy controls. Anti-MDA5 antibodies in the patients' serum samples were quantified using our newly developed ELISA, and the results were compared to those obtained using the gold-standard immunoprecipitation (IP) assay. In addition, correlations between the ELISA-quantified anti-MDA5 antibodies and clinical characteristics were evaluated. RESULTS: In patients with PM/DM, the anti-MDA5 antibody measurements obtained from the ELISA and IP assay were highly concordant; the ELISA exhibited an analytical sensitivity of 98.2%, specificity of 100%, positive predictive value of 100%, and negative predictive value of 99.5% (compared to the IP assay). Anti-MDA5 antibodies were detected in 22.7% of the DM patients, but not in any of the patients with PM, non-PM/DM CTD, or IIP. Clinically amyopathic DM, RP-ILD, arthritis, and fever were more prevalent in DM patients who were anti-MDA5 antibody-positive than in those who were antibody-negative (P ≤ 0.0002 for all comparisons). In addition, anti-MDA5 antibody-positive patients with RP-ILD exhibited higher antibody levels than those without RP-ILD (P = 0.006). CONCLUSION: Our newly developed ELISA can detect anti-MDA5 antibodies as efficiently as the gold standard IP assay and has the potential to facilitate the routine clinical measurement of anti-MDA5 antibodies in patients who suspected to have DM.


Asunto(s)
Autoanticuerpos/sangre , Enfermedades del Tejido Conjuntivo/diagnóstico , Dermatomiositis/diagnóstico , Ensayo de Inmunoadsorción Enzimática/normas , Neumonías Intersticiales Idiopáticas/diagnóstico , Helicasa Inducida por Interferón IFIH1/inmunología , Polimiositis/diagnóstico , Adulto , Anciano , Estudios de Casos y Controles , Enfermedades del Tejido Conjuntivo/sangre , Enfermedades del Tejido Conjuntivo/inmunología , Dermatomiositis/sangre , Dermatomiositis/inmunología , Diagnóstico Diferencial , Femenino , Expresión Génica , Humanos , Neumonías Intersticiales Idiopáticas/sangre , Neumonías Intersticiales Idiopáticas/inmunología , Inmunoprecipitación , Helicasa Inducida por Interferón IFIH1/antagonistas & inhibidores , Helicasa Inducida por Interferón IFIH1/genética , Masculino , Persona de Mediana Edad , Polimiositis/sangre , Polimiositis/inmunología , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA