Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Neuroimmunol ; 360: 577715, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34536787

RESUMEN

This post-hoc analysis evaluated candidate biomarkers of long-term efficacy of subcutaneous interferon beta-1a (sc IFN ß-1a) in REFLEX/REFLEXION studies of clinically isolated syndrome. Samples from 507 REFLEX and 287 REFLEXION study participants were analyzed. All investigated biomarkers were significantly upregulated 1.5-4-fold in response to sc IFN ß-1a treatment versus baseline (p ≤ 0.008). The validity of MX1, 2'5'OAS, and IL-1RA as biomarkers of response to sc IFN ß-1a was confirmed in this large patient cohort, with biomarkers consistently upregulated in a dose-dependent manner. Neopterin, TRAIL, and IP-10 were confirmed as biomarkers associated with long-term sc IFN ß-1a treatment efficacy over 5 years.


Asunto(s)
Interferón beta-1a/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , 2',5'-Oligoadenilato Sintetasa/biosíntesis , 2',5'-Oligoadenilato Sintetasa/sangre , 2',5'-Oligoadenilato Sintetasa/genética , Biomarcadores , Quimiocina CXCL10/biosíntesis , Quimiocina CXCL10/sangre , Quimiocina CXCL10/genética , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Estudios de Seguimiento , Humanos , Inyecciones Subcutáneas , Interferón beta-1a/administración & dosificación , Interferón beta-1a/farmacocinética , Proteína Antagonista del Receptor de Interleucina 1/biosíntesis , Proteína Antagonista del Receptor de Interleucina 1/sangre , Proteína Antagonista del Receptor de Interleucina 1/genética , Estudios Multicéntricos como Asunto , Esclerosis Múltiple/sangre , Proteínas de Resistencia a Mixovirus/biosíntesis , Proteínas de Resistencia a Mixovirus/sangre , Proteínas de Resistencia a Mixovirus/genética , Neopterin/biosíntesis , Neopterin/sangre , Neopterin/genética , Ensayos Clínicos Controlados Aleatorios como Asunto/estadística & datos numéricos , Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Ligando Inductor de Apoptosis Relacionado con TNF/sangre , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Regulación hacia Arriba
3.
J Biosci ; 462021.
Artículo en Inglés | MEDLINE | ID: mdl-34323222

RESUMEN

Membraneless organelles (MLOs) in the cytoplasm and nucleus in the form of phase-separated biomolecular condensates are increasingly viewed as critical in regulating diverse cellular functions. We summarize a paradigm shift over the last 3 years in the field of interferon (IFN)-inducible antiviral Mx-family GTPases. Expression of the 'myxovirus resistance proteins' MxA in human cells and its ortholog Mx1 in murine cells is increased 50- to 100-fold by Type I (IFN-α and -ß) and III IFNs (IFN-λ). Human MxA forms cytoplasmic structures, while murine Mx1 forms nuclear bodies. Since 2002, it has been widely thought that human (Hu) MxA is associated with the membraneous smooth endoplasmic reticulum (ER). In a paradigm shift, our recent data showed that HuMxA formed membraneless phase-separated biomolecular condensates in the cytoplasm. Some of the HuMxA condensates adhered to intermediate filaments generating a reticular pattern. Murine (Mu) Mx1, which was predominantly nuclear, was also confirmed to be in phase-separated nuclear biomolecular condensates. A subset of Huh7 cells showed association of GFP-MuMx1 with intermediate filaments in the cytoplasm. While cells with cytoplasmic GFP-HuMxA condensates and cytoplasmic GFP-MuMx1 filaments showed an antiviral phenotype towards vesicular stomatitis virus (VSV), those with only nuclear GFP-MuMx1 bodies did not. The new data bring forward the paradigm that both human MxA and murine Mx1 give rise to phase-separated biomolecular condensates, albeit in different subcellular compartments, and that differences in the subcellular localization of condensates of different Mx proteins determines the spectrum of their antiviral activity.


Asunto(s)
Condensados Biomoleculares/metabolismo , GTP Fosfohidrolasas/biosíntesis , Interferones/fisiología , Proteínas de Resistencia a Mixovirus/biosíntesis , Secuencia de Aminoácidos , Animales , GTP Fosfohidrolasas/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones , Proteínas de Resistencia a Mixovirus/química , Proteínas de Resistencia a Mixovirus/metabolismo , Fracciones Subcelulares/metabolismo
4.
Eur J Neurol ; 27(3): 514-521, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31571350

RESUMEN

BACKGROUND AND PURPOSE: Dermatomyositis (DM) with anti-nuclear matrix protein-2 (NXP-2) antibodies usually shows multifocal ischaemic lesions in muscle. Here, we aimed to investigate the microarteriopathy underlying muscle ischaemia in anti-NXP-2-positive DM. METHODS: A total of 16 patients diagnosed with anti-NXP-2-positive DM were investigated by muscle biopsy. A total of 13 patients with DM with other myositis-specific antibodies and 11 normal controls were included for comparison. Immunofluorescence assays were performed to localize endothelial cells, smooth muscle cells and pericytes, and to determine lesions in myofibers and microvessels by vascular endothelial growth factor and myxovirus resistance protein A (MxA). Electron microscopy was carried out to assess ultrastructure alterations. RESULTS: Subcutaneous edema, severe muscle weakness and dysphagia together with elevated creatine kinase, D-dimer and triglyceride levels, and decreased albumin levels were found in anti-NXP-2-positive DM. Muscle ischaemia included regional muscle edema, perifascicular atrophy, microinfarcts and focal punched-out vacuoles. The density of arterioles was higher in anti-NXP-2-positive DM (P ï¼œ 0.05). Perimysial arterioles with thickened vascular wall, thrombosis and lipid accumulation were found in the vascular wall of diseased perimysial arterioles. The frequency of diseased arterioles and thrombosis was higher in anti-NXP-2-positive DM (P < 0.05). Sarcoplasmic vascular endothelial growth factor and MxA expression was observed in multifocal ischaemic lesions. MxA was present in endothelial and smooth muscle cells of the diseased arterioles and pericytes. Electron microscopy confirmed damaged capillaries and tubuloreticular structures. CONCLUSIONS: Our research suggested that perimysial arterioles were most commonly involved in anti-NXP-2-positive DM, which led to muscle ischaemia.


Asunto(s)
Adenosina Trifosfatasas/inmunología , Anticuerpos Antinucleares/análisis , Proteínas de Unión al ADN/inmunología , Dermatomiositis/patología , Adolescente , Adulto , Arteriolas/patología , Biopsia , Capilares/patología , Niño , Preescolar , Dermatomiositis/complicaciones , Células Endoteliales/patología , Femenino , Humanos , Lactante , Masculino , Microcirculación , Persona de Mediana Edad , Fibras Musculares Esqueléticas/patología , Fibras Musculares Esqueléticas/ultraestructura , Miocitos del Músculo Liso/patología , Miocitos del Músculo Liso/ultraestructura , Proteínas de Resistencia a Mixovirus/biosíntesis , Proteínas de Resistencia a Mixovirus/genética , Pericitos/patología , Pericitos/ultraestructura
5.
J Neuroimmunol ; 337: 577047, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31520792

RESUMEN

The molecular mechanisms of antiviral innate immune reactions in brain microvascular endothelial cells remain unclear. Interferon (IFN)-induced transmembrane protein 1 (IFITM1) and Myxovirus resistance protein 1 (MX1), the members of IFN-stimulated genes, are known as antiviral molecules. IFITM1 inhibits virus entry into host cell cytoplasm, whereas MX1 antagonizes virus replication. Here we observed that IFITM1 and MX1, and a proinflammatory cytokine IL-6 expression was induced by polyinosinic-polycytidylic acid (poly IC) in hCMEC/D3 human brain microvascular endothelial cells. Poly IC-induced IFITM1 and MX1 expression were decreased by NF-κB inhibitor SN50, IFN regulatory factor 3 inhibitor MRT67307 and human type I IFN neutralizing antibody mixture. These findings suggest that IFITM1 and MX1 may help protect the brain from viruses.


Asunto(s)
Antígenos de Diferenciación/biosíntesis , Células Endoteliales/metabolismo , Microvasos/metabolismo , Proteínas de Resistencia a Mixovirus/biosíntesis , Poli I-C/farmacología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Microvasos/citología , Microvasos/efectos de los fármacos
6.
J Neuroimmunol ; 332: 73-77, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-30959341

RESUMEN

The most notable effect of prenatal Zika virus (ZIKV) infection is severe microcephaly. ZIKV has a selective tropism for neural progenitor cells; however, it is not clear what role the immune cells of the brain, microglia, may have in mitigating or exacerbating neuronal cell death following ZIKV infection. We cultured hippocampal and cortical neural cells from neonatal rat pups and infected them with ZIKV at various multiplicities of infection (MOI). We found that the neuroimmune response to ZIKV infection is composed of both pro-inflammatory and type I interferon responses and is largely dependent upon the viral dose.


Asunto(s)
Células-Madre Neurales/virología , Infección por el Virus Zika/inmunología , Virus Zika/patogenicidad , 2',5'-Oligoadenilato Sintetasa/biosíntesis , Animales , Animales Recién Nacidos , Células Cultivadas , Corteza Cerebral/citología , Femenino , Hipocampo/citología , Interferón beta/biosíntesis , Interleucina-6/biosíntesis , Masculino , Microglía/inmunología , Proteínas de Resistencia a Mixovirus/biosíntesis , Células-Madre Neurales/inmunología , Células-Madre Neurales/metabolismo , Ratas , Tropismo Viral
7.
Appl Microbiol Biotechnol ; 103(5): 2171-2180, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30637496

RESUMEN

The Chinese tree shrew holds a great potential as a viable animal model in biomedical research, especially for infectious diseases and neuropsychiatric disorders. A thorough understanding of the innate immunity, which represents the first line that defends the host against viral infection, of the Chinese tree shrew, is needed. However, the progress is hindered by the lack of a proper cell line for research usage. In this study, we established a cell line that is applicable to the study of tree shrew innate immune responses against viral infections. The Chinese tree shrew primary renal cells (TSPRCs) were immortalized by simian virus 40 large T antigen (SV40LT) transduction, and the immortalized cells were termed TSR6 (tree shrew renal cell #6). TSR6 showed a similar morphology to TSPRCs and expressed the epithelial cell-specific marker cytokeratin 18 (KRT18). In addition, TSR6 could be transfected by transfection reagent and was suitable for CRISPR/Cas9-mediated gene editing. Infection of Newcastle disease virus (NDV) or herpes simplex virus 1 (HSV-1) in TSR6 induced the mRNA expression of tree shrew interferon-ß (tIFNB1) and myxovirus resistance protein 1 (tMx1) in a dose- and time-dependent manner. Collectively, we successfully established a tree shrew renal cell line and demonstrated that this cell line was suitable for the study of the innate immune response to viral infections.


Asunto(s)
Células Epiteliales/metabolismo , Edición Génica/métodos , Inmunidad Innata/inmunología , Riñón/citología , Virosis/inmunología , Animales , Antígenos Transformadores de Poliomavirus/genética , Sistemas CRISPR-Cas , Técnicas de Cultivo de Célula , Línea Celular , Modelos Animales de Enfermedad , Células HEK293 , Herpesvirus Humano 1/crecimiento & desarrollo , Humanos , Interferón beta/biosíntesis , Queratina-18/biosíntesis , Proteínas de Resistencia a Mixovirus/biosíntesis , Virus de la Enfermedad de Newcastle/crecimiento & desarrollo , Cultivo Primario de Células , Tupaiidae
8.
Clin Chem ; 65(6): 739-750, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30593466

RESUMEN

BACKGROUND: In 2015, the 68th World Health Assembly declared that effective, rapid, low-cost diagnostic tools were needed for guiding optimal use of antibiotics in medicine. This review is devoted to interferon-inducible myxovirus resistance proteins as potential biomarkers for differentiating viral from bacterial infections. CONTENT: After viral infection, a branch of the interferon (IFN)-induced molecular reactions is triggered by the binding of IFNs with their receptors, a process leading to the activation of mx1 and mx2, which produce antiviral Mx proteins (MxA and MxB). We summarize current knowledge of the structures and functions of type I and III IFNs. Antiviral mechanisms of Mx proteins are discussed in reference to their structural and functional data to provide an in-depth picture of protection against viral attacks. Knowing such a mechanism may allow the development of countermeasures and the specific detection of any viral infection. Clinical research data indicate that Mx proteins are biomarkers for many virus infections, with some exceptions, whereas C-reactive protein (CRP) and procalcitonin have established positions as general biomarkers for bacterial infections. SUMMARY: Mx genes are not directly induced by viruses and are not expressed constitutively; their expression strictly depends on IFN signaling. MxA protein production in peripheral blood cells has been shown to be a clinically sensitive and specific marker for viral infection. Viral infections specifically increase MxA concentrations, whereas viruses have only a modest increase in CRP or procalcitonin concentrations. Therefore, comparison of MxA and CRP and/or procalcitonin values can be used for the differentiation of infectious etiology.


Asunto(s)
Interferones/fisiología , Proteínas de Resistencia a Mixovirus/biosíntesis , Virosis/diagnóstico , Biomarcadores/metabolismo , Diagnóstico Diferencial , Humanos , Virosis/metabolismo
9.
Int Immunopharmacol ; 62: 1-6, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29960044

RESUMEN

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


Asunto(s)
Anticuerpos Neutralizantes/sangre , Interferón beta/inmunología , Esclerosis Múltiple/sangre , Proteínas Recombinantes/inmunología , Células A549 , Bioensayo , Reacciones Cruzadas , Efecto Citopatogénico Viral , Virus de la Encefalomiocarditis/inmunología , Humanos , Esclerosis Múltiple/inmunología , Proteínas de Resistencia a Mixovirus/biosíntesis , Pruebas de Neutralización , Polietilenglicoles
10.
Med Hypotheses ; 112: 47-50, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29447938

RESUMEN

Human Herpes Virus type 6 (HHV-6) is a ubiquitous virus consisting of two viral species, HHV-6A and HHV-6B that have been associated with numerous and diverse pathologies. As many other viruses HHV-6 modulates the apoptotic machinery of its host to subvert immune response to infection, yet the exact mechanisms behind this process remain under investigation. The genes encoding the CTSS, PTX3, CHI3L1, Mx1, CXCL16, BIRC3 and BST2 proteins have been linked to HHV-6Α related neurologic diseases whilst also associated with apoptosis. This study aimed at the identification and functional analysis of the gene interaction network (interactome) of CTSS-PTX3-CHI3L1-Mx1-CXCL16-BIRC3-BST2 so as to evaluate the hypothesis of a probable link between the latter and host's immune response to HHV-6A infection.


Asunto(s)
Inmunidad Adaptativa/genética , Proteínas Reguladoras de la Apoptosis/genética , Redes Reguladoras de Genes , Herpesvirus Humano 6/fisiología , Interacciones Huésped-Patógeno/inmunología , Modelos Genéticos , Modelos Inmunológicos , Infecciones por Roseolovirus/inmunología , Antígenos CD/biosíntesis , Antígenos CD/genética , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteína 3 que Contiene Repeticiones IAP de Baculovirus/biosíntesis , Proteína 3 que Contiene Repeticiones IAP de Baculovirus/genética , Proteína C-Reactiva/biosíntesis , Proteína C-Reactiva/genética , Catepsinas/biosíntesis , Catepsinas/genética , Quimiocina CXCL16/biosíntesis , Quimiocina CXCL16/genética , Proteína 1 Similar a Quitinasa-3/biosíntesis , Proteína 1 Similar a Quitinasa-3/genética , Proteínas Ligadas a GPI/biosíntesis , Proteínas Ligadas a GPI/genética , Regulación Viral de la Expresión Génica/inmunología , Ontología de Genes , Redes Reguladoras de Genes/genética , Redes Reguladoras de Genes/inmunología , Interacciones Huésped-Patógeno/genética , Humanos , Evasión Inmune/genética , Proteínas de Resistencia a Mixovirus/biosíntesis , Proteínas de Resistencia a Mixovirus/genética , Infecciones por Roseolovirus/genética , Componente Amiloide P Sérico/biosíntesis , Componente Amiloide P Sérico/genética
11.
Arch Virol ; 162(5): 1387-1392, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28155193

RESUMEN

The expression of Mx1 in EPC cells after treatment with poly(I:C) or infection with viral hemorrhagic septicemia virus (VHSV) was significantly suppressed by treatment with dexamethasone. However, the titer of VHSV did not increase but instead decreased after dexamethasone treatment. This suggests that dexamethasone not only downregulates type I IFN but also affects certain factors that are necessary for VHSV replication. An important effect of HSP90 on replication of RNA viruses and downregulation of HSP90 by glucocorticoids have been reported. In this study, dexamethasone downregulated HSP90α expression in EPC cells that were stimulated with poly(I:C) or infected with VHSV. Furthermore, cells treated with an HSP90 inhibitor, geldanamycin, showed significantly decreased titers of VHSV, suggesting that HSP90 may be an important host component involved in VHSV replication, and HSP90 inhibition might be one of the causes for the observed reduction in viral titer caused by dexamethasone treatment.


Asunto(s)
Cyprinidae/virología , Dexametasona/farmacología , Enfermedades de los Peces/virología , Proteínas HSP90 de Choque Térmico/biosíntesis , Proteínas de Resistencia a Mixovirus/biosíntesis , Novirhabdovirus/genética , Poli I-C/farmacología , Animales , Benzoquinonas/farmacología , Carcinoma/metabolismo , Carcinoma/virología , Línea Celular Tumoral , Replicación del ADN/efectos de los fármacos , Regulación hacia Abajo , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Terapia de Inmunosupresión , Interferón Tipo I/biosíntesis , Lactamas Macrocíclicas/farmacología , Novirhabdovirus/inmunología , Replicación Viral/efectos de los fármacos
12.
Virus Genes ; 53(2): 233-239, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27866318

RESUMEN

In this study, we constructed for the first time a full-length cDNA clone of pig-original bovine viral diarrhea virus 2 (BVDV-2) strain SH-28, modified the cDNA clone (pASH28) for mutant pASHΔNpro and derived virus strain vASHΔNpro by deleting the genomic region encoding the Npro polypeptide, and examined significance of protein Npro for antiviral responses in vitro. Data showed that Npro-deletion mutant virus vASHΔNpro led to significant overexpression of oligo adenylate synthetase (OAS), myxovirus-resistant protein 1 (Mx1), and ubiquitin-like protein 15 (ISG15). Data also revealed that overexpression of Npro, but not NS2 and NS3 proteins, resulted in significant down-regulation of OAS, Mx1, and ISG15 production (p ≤ 0.05) in bovine cells as well as porcine cells transfected with Npro recombinant eukaryotic expression plasmids. Npro (but not NS2 and NS3) was also found to inhibit poly(IC) from inducing production of type I interferon (IFN-I). These results indicated that protein Npro may play multiple roles in regulating antiviral response in host cells interfered by pig BVDV-2 strain, and provided useful information to understand better the mechanism of BVDV-2 persistent infection in pigs.


Asunto(s)
Virus de la Diarrea Viral Bovina Tipo 2/genética , Proteínas de Resistencia a Mixovirus/genética , Infecciones por Pestivirus/genética , Proteínas no Estructurales Virales/genética , Animales , Bovinos , Línea Celular , Virus de la Diarrea Viral Bovina Tipo 2/patogenicidad , Regulación Viral de la Expresión Génica , Proteínas de Resistencia a Mixovirus/biosíntesis , Infecciones por Pestivirus/virología , Porcinos/virología , Ubiquitinas/biosíntesis , Ubiquitinas/genética , Proteínas no Estructurales Virales/biosíntesis
13.
J Neurol ; 263(4): 722-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26872667

RESUMEN

The aim of this study is to investigate whether induction of myxovirus resistance protein A (MxA) mRNA after 3 months of interferon-ß administration is related to the treatment response in multiple sclerosis (MS) patients. In this prospective study, MS patients were enrolled before starting treatment. Demographic, clinical and radiological variables were recorded. Blood samples were obtained before, and at 3 and 12 months after interferon-ß treatment. Real-time PCR was used to analyze MxA mRNA expression. Patients were classified as MxA-low or -high depending on MxA levels at baseline, and as MxA-induced or -non-induced according to whether an increase in MxA expression was detected at month 3. Time to the next relapse was investigated using Cox proportional hazards regression analysis. One hundred and four patients were selected and followed for a median of 2.2 years (IQR 1.6-3.5). On Cox regression analysis, a higher EDSS score before treatment (HR 1.57; 95 % CI 1.02-2.40; p = 0.039), MxA-high status at baseline (HR 2.71; 95 % CI 1.26-5.81; p = 0.010), and MxA-non-induced at month 3 (HR 2.49; 95 % CI 1.08-5.68; p = 0.031), were predictors of poor response to interferon-ß in naïve MS patients. Patients showing a lower capacity for MxA induction following 3 months of interferon-ß treatment are more likely to be non-responders to this therapy.


Asunto(s)
Factores Inmunológicos/uso terapéutico , Interferón beta/uso terapéutico , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Esclerosis Múltiple Recurrente-Remitente/metabolismo , Proteínas de Resistencia a Mixovirus/biosíntesis , Adulto , Femenino , Humanos , Masculino , Modelos de Riesgos Proporcionales , Estudios Prospectivos , Reacción en Cadena en Tiempo Real de la Polimerasa , Resultado del Tratamiento
14.
Eur J Immunol ; 46(1): 167-77, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26460926

RESUMEN

MicroRNAs are important posttranscriptional regulators of gene expression, which have been shown to fine-tune innate immune responses downstream of pattern recognition receptor (PRR) signaling. This study identifies miR-650 as a novel PRR-responsive microRNA that is downregulated upon stimulation of primary human monocyte-derived dendritic cells (MDDCs) with a variety of different microbe-associated molecular patterns. A comprehensive target search combining in silico analysis, transcriptional profiling, and reporter assays reveals that miR-650 regulates several well-known interferon-stimulated genes, including IFIT2 and MXA. In particular, downregulation of miR-650 in influenza A infected MDDCs enhances the expression of MxA and may therefore contribute to the establishment of an antiviral state. Together these findings reveal a novel link between miR-650 and the innate immune response in human MDDCs.


Asunto(s)
Células Dendríticas/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética , Virus de la Influenza A/inmunología , MicroARNs/inmunología , Proteínas de Resistencia a Mixovirus/biosíntesis , Células Cultivadas , Células Dendríticas/metabolismo , Regulación hacia Abajo , Citometría de Flujo , Regulación de la Expresión Génica/genética , Humanos , Inmunidad Innata/inmunología , Immunoblotting , MicroARNs/genética , Microscopía Confocal , Proteínas de Resistencia a Mixovirus/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de Reconocimiento de Patrones/inmunología , Transducción de Señal , Transfección
15.
Virol J ; 12: 150, 2015 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-26411318

RESUMEN

BACKGROUND: In viral disease, infection is controlled at the cellular level by type I interferon (IFN-I), but dengue virus (DENV) has the ability to inhibit this response. Type III interferon, also known as lambda IFN (IFN-III or IFN-λ), is a complementary pathway to the antiviral response by IFN-I. This work analyzed the IFN-λ (IFN-III) mediated antiviral response against DENV serotype 2 (DENV-2) infection. METHODS: Dengue fever patients were sampled to determine their IFN-λ levels by ELISA. To study the IFN-λ response during DENV infection we selected the epithelial cell line C33-A, and we demonstrated that it is permissive to DENV-2 infection. The effect of IFN-λ on virus replication was determined in these cells, in parallel to the expression of IFN-stimulated genes (ISGs), and Suppressor of Cytokine Signaling (SOCS), genes measured by RT-qPCR. RESULTS: We found increased (~1.8 times) serological IFN-λ in dengue fever patients compared to healthy blood donors. IFN-λ inhibited DENV-2 replication in a dose-dependent manner in vitro. The reduction of viral titer corresponded with increased ISG mRNA levels (MX1 and OAS1), with the highest inhibition occurring at ISG's peak expression. Presence of IFN-negative regulators, SOCS1 and SOCS3, during DENV-2 infection was associated with reduced IFN-λ1 expression. CONCLUSIONS: Evidence described here suggests that IFN-λ is a good candidate inhibitor of viral replication in dengue infection. Mechanisms for the cellular and organismal interplay between DENV and IFN- λ need to be further studied as they could provide insights into strategies to treat this disease. Furthermore, we report a novel epithelial model to study dengue infection in vitro.


Asunto(s)
Antivirales/metabolismo , Virus del Dengue/inmunología , Virus del Dengue/fisiología , Células Epiteliales/inmunología , Células Epiteliales/virología , Interleucinas/metabolismo , Replicación Viral/efectos de los fármacos , 2',5'-Oligoadenilato Sintetasa/biosíntesis , 2',5'-Oligoadenilato Sintetasa/genética , Antivirales/sangre , Perfilación de la Expresión Génica , Humanos , Interferones , Interleucinas/sangre , Proteínas de Resistencia a Mixovirus/biosíntesis , Proteínas de Resistencia a Mixovirus/genética , ARN Mensajero/análisis , ARN Mensajero/genética , Carga Viral
16.
J Invest Dermatol ; 135(12): 2935-2943, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26288353

RESUMEN

IFNλ is important for epidermal defense against viruses. It is produced by, and acts on, keratinocytes, whereas fibroblasts were previously considered to be unresponsive to this type III IFN. Herein we report findings revealing cell type-specific differences in IFNλ signaling and function in skin resident cells. In dermal fibroblasts, IFNλ induced the expression of myxovirus protein A (MxA), a potent antiviral factor, but not other IFN signature genes as it does in primary keratinocytes. In contrast to its effect on keratinocytes, IFNλ did not phosphorylate signal transducer and activator of transcription 1 in fibroblasts, but instead activated mitogen activated protein kinases (MAPK). Accordingly, inhibition of MAPK activation (p38 and p42/44) blocked the expression of MxA protein in fibroblasts but not in keratinocytes. Functionally, IFNλ inhibited proliferation in keratinocytes but not in fibroblasts. Moreover, IFNλ upregulated the expression of Tumor growth factor beta 1 (TGFß1)-induced collagens in fibroblasts. Taken together, our findings identify primary human dermal fibroblasts as responder cells to IFNλ. Our study shows cutaneous cell type-specific IFN signaling and suggests that IFNλ, although important for epidermal antiviral competence, may also have a regulatory role in the dermal compartment balancing type I IFN-induced inhibition of tissue repair processes.


Asunto(s)
Interferón gamma/farmacología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Proteínas de Resistencia a Mixovirus/biosíntesis , Factor de Transcripción STAT1/fisiología , Piel/metabolismo , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Interferón-alfa/farmacología , Receptores de Interferón/fisiología , Piel/citología , Factor de Crecimiento Transformador beta1/farmacología , Receptor de Interferón gamma
17.
Dev Comp Immunol ; 53(1): 55-62, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26133072

RESUMEN

Although several studies have demonstrated the ability of some endocrine disruptive chemicals (EDCs) to alter the physiology of zebrafish, the immune-reproductive interaction has received little attention in this species. In this study, we used a homozygous line carrying an insertion of 8 amino acids in the ligand-binding domain of the estrogen receptor 2b gene (esr2b) to further understand the role of estrogen signaling on innate immunity. Adult mutant fish showed distorted sexual ratios related with alterations in testicular morphology and supraphysiological testosterone and 17ß-estradiol (E2) levels. Immunity-wise, although esr2b mutant fish showed unaltered antibacterial responses, they were unable to mount an effective antiviral response upon viral challenge. RT-qPCR analysis demonstrated that mutant fish were able to induce the genes encoding major antiviral molecules, including Ifnphi1, Ifnphi2, Infphi3, Mxb and Mxc, and the negative feedback regulator of cytokine signaling Socs1. Notably, although esr2b mutant larvae showed a similar resistance to SVCV infection to their wild type siblings, waterborne E2 increased their viral susceptibility. Similarly, the exposure of adult wild type zebrafish to E2 also resulted in increased susceptibility to SVCV infection. Finally, the administration of recombinant Ifnphi1 hardly reversed the higher viral susceptibility of esr2b mutant zebrafish, suggesting that elevated socs1 levels impair Ifn signaling. All together, these results uncover an important role for E2 and Esr signaling in the fine-tuning of sexual hormone balance and the antiviral response of vertebrates.


Asunto(s)
Receptor beta de Estrógeno/genética , Enfermedades de los Peces/inmunología , Rhabdoviridae/inmunología , Vibrio/inmunología , Proteínas de Pez Cebra/genética , Pez Cebra/inmunología , Animales , Estradiol/metabolismo , Receptor beta de Estrógeno/deficiencia , Enfermedades de los Peces/microbiología , Enfermedades de los Peces/virología , Inmunidad Innata/inmunología , Interferones/biosíntesis , Larva/inmunología , Proteínas de Resistencia a Mixovirus/biosíntesis , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Pez Cebra/genética , Proteínas de Pez Cebra/biosíntesis , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/metabolismo
18.
Discov Med ; 19(107): 419-25, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26175399

RESUMEN

OBJECTIVES: Elevated type I interferon (IFN) is believed to be one of the crucial factors involved in the pathogenesis of systemic lupus erythematosus (SLE). Its expression was recently found to be governed by the transcription factor E2F1 which is involved in an autoregulatory triad along with c-Myc and the microRNA polycistron miR-17-92. However, this intricate triad has seldom been investigated in SLE patients. Therefore, the current study was undertaken to investigate the expression pattern of the E2F1/c-Myc/miR-17-5p triad in peripheral blood of SLE patients as well as to examine the impact of manipulating this triad using miR-17-5p mimics and inhibitors on IFN signature in SLE patients. METHODS: Expression of the E2F1/c-Myc/miR-17-5p triad and the IFN-stimulated gene MxA was analyzed using real time qPCR. Peripheral blood mononuclear cells from SLE patients and controls were transfected with miR-17-5p mimics and antagomirs using the HiPerfect transfection reagent. RESULTS: E2F1 transcripts and miR-17-5p were significantly downregulated while c-Myc and MxA transcripts were significantly upregulated in SLE. Also, transfection of SLE PBMCs with miR-17-5p mimics led to a substantial repression of E2F1 and c-Myc expression. The overall change in this triad upon miR-17-5p mimicking resulted in lowering the transcript levels of the IFN-inducible gene MxA in SLE. CONCLUSION: This may advocate the manipulation/use of the E2F1/c-Myc/miR-17-5p trinity to effectively control the aberrantly high levels of type I IFN activity in lupus patients.


Asunto(s)
Factor de Transcripción E2F1/biosíntesis , Regulación de la Expresión Génica , Leucocitos Mononucleares/metabolismo , Lupus Eritematoso Sistémico/metabolismo , MicroARNs/metabolismo , Proteínas de Resistencia a Mixovirus/biosíntesis , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Adolescente , Células Cultivadas , Niño , Femenino , Humanos , Leucocitos Mononucleares/patología , Lupus Eritematoso Sistémico/patología , Masculino
19.
J Virol ; 89(17): 8880-96, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26085147

RESUMEN

UNLABELLED: Zika virus (ZIKV) is an emerging arbovirus of the Flaviviridae family, which includes dengue, West Nile, yellow fever, and Japanese encephalitis viruses, that causes a mosquito-borne disease transmitted by the Aedes genus, with recent outbreaks in the South Pacific. Here we examine the importance of human skin in the entry of ZIKV and its contribution to the induction of antiviral immune responses. We show that human dermal fibroblasts, epidermal keratinocytes, and immature dendritic cells are permissive to the most recent ZIKV isolate, responsible for the epidemic in French Polynesia. Several entry and/or adhesion factors, including DC-SIGN, AXL, Tyro3, and, to a lesser extent, TIM-1, permitted ZIKV entry, with a major role for the TAM receptor AXL. The ZIKV permissiveness of human skin fibroblasts was confirmed by the use of a neutralizing antibody and specific RNA silencing. ZIKV induced the transcription of Toll-like receptor 3 (TLR3), RIG-I, and MDA5, as well as several interferon-stimulated genes, including OAS2, ISG15, and MX1, characterized by strongly enhanced beta interferon gene expression. ZIKV was found to be sensitive to the antiviral effects of both type I and type II interferons. Finally, infection of skin fibroblasts resulted in the formation of autophagosomes, whose presence was associated with enhanced viral replication, as shown by the use of Torin 1, a chemical inducer of autophagy, and the specific autophagy inhibitor 3-methyladenine. The results presented herein permit us to gain further insight into the biology of ZIKV and to devise strategies aiming to interfere with the pathology caused by this emerging flavivirus. IMPORTANCE: Zika virus (ZIKV) is an arbovirus belonging to the Flaviviridae family. Vector-mediated transmission of ZIKV is initiated when a blood-feeding female Aedes mosquito injects the virus into the skin of its mammalian host, followed by infection of permissive cells via specific receptors. Indeed, skin immune cells, including dermal fibroblasts, epidermal keratinocytes, and immature dendritic cells, were all found to be permissive to ZIKV infection. The results also show a major role for the phosphatidylserine receptor AXL as a ZIKV entry receptor and for cellular autophagy in enhancing ZIKV replication in permissive cells. ZIKV replication leads to activation of an antiviral innate immune response and the production of type I interferons in infected cells. Taken together, these results provide the first general insights into the interaction between ZIKV and its mammalian host.


Asunto(s)
Células Dendríticas/virología , Flaviviridae/fisiología , Queratinocitos/virología , Internalización del Virus , Replicación Viral , Aedes/virología , Animales , Autofagia/inmunología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Chlorocebus aethiops , Citocinas/biosíntesis , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Células Dendríticas/inmunología , Fibroblastos/virología , Flaviviridae/inmunología , Infecciones por Flaviviridae/inmunología , Infecciones por Flaviviridae/virología , Células HEK293 , Receptor Celular 1 del Virus de la Hepatitis A , Humanos , Insectos Vectores/virología , Helicasa Inducida por Interferón IFIH1 , Interferón beta/biosíntesis , Interferón beta/inmunología , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de Resistencia a Mixovirus/biosíntesis , Fagosomas/inmunología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores Inmunológicos , Receptores Virales/genética , Receptores Virales/metabolismo , Piel/inmunología , Piel/virología , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/inmunología , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 7/inmunología , Ubiquitinas/biosíntesis , Células Vero , Tirosina Quinasa del Receptor Axl
20.
Dev Comp Immunol ; 48(1): 95-101, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25265425

RESUMEN

The retinoic acid-inducible gene I (RIG-I) is a critical sensor for host recognition of RNA virus infection and initiation of antiviral signaling pathways in mammals. However, data on the occurrence and functions of this molecule in lower vertebrates are limited. In this study, we characterized an RIG-I homolog (DrRIG-I) from zebrafish. Structurally, this DrRIG-I shares a number of conserved functional domains/motifs with its mammalian counterparts, namely, caspase activation and recruitment domain, DExD/H box, a helicase domain, and a C-terminal domain. Functionally, stimulation with DrRIG-I CARD in zebrafish embryos significantly activated the NF-κB and IFN signaling pathways, leading to the expression of TNF-α, IL-8 and IFN-induced Mx, ISG15, and viperin. However, knockdown of TRIM25 (a pivotal activator for RIG-I receptors) significantly suppressed the induced activation of IFN signaling. Results suggested the functional conservation of RIG-I receptors in the NF-κB and IFN signaling pathways between teleosts and mammals, providing a perspective into the evolutionary history of RIG-I-mediated antiviral innate immunity.


Asunto(s)
Interferones/metabolismo , FN-kappa B/metabolismo , Infecciones por Virus ARN/inmunología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/inmunología , Secuencia de Aminoácidos , Animales , Antivirales/metabolismo , Secuencia de Bases , Proteínas Adaptadoras de Señalización CARD/biosíntesis , Proteínas Adaptadoras de Señalización CARD/metabolismo , ARN Helicasas DEAD-box , Femenino , Enfermedades de los Peces/inmunología , Técnicas de Silenciamiento del Gen , Inmunidad Innata/inmunología , Interleucina-8/biosíntesis , Masculino , Datos de Secuencia Molecular , Morfolinos/genética , Proteínas de Resistencia a Mixovirus/biosíntesis , Infecciones por Virus ARN/virología , Virus ARN/inmunología , Alineación de Secuencia , Análisis de Secuencia de ADN , Factores de Transcripción/genética , Factor de Necrosis Tumoral alfa/biosíntesis , Pez Cebra/virología , Proteínas de Pez Cebra/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...