Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 438
Filtrar
1.
Hepatology ; 69(3): 1004-1019, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30199576

RESUMEN

Long noncoding RNAs (lncRNAs) play a critical role in the regulation of many important cellular processes. However, the mechanisms by which lncRNAs regulate viral infection and host immune responses are not well understood. We sought to explore lncRNA regulation of hepatitis C virus (HCV) infection and interferon response. We performed RNA sequencing (RNAseq) in Huh7.5.1 cells with or without interferon alpha (IFNα) treatment. Clustered regularly interspaced short palindromic repeats/Cas9 guide RNA (gRNA) was used to knock out selected genes. The promoter clones were constructed, and the activity of related interferon-stimulated genes (ISGs) were detected by the secrete-pair dual luminescence assay. We constructed the full-length and four deletion mutants of an interferon-induced lncRNA RP11-288L9.4 (lncRNA-IFI6) based on predicted secondary structure. Selected gene mRNAs and their proteins, together with HCV infection, in Huh7.5.1 cells and primary human hepatocytes (PHHs) were monitored by quantitative real-time PCR (qRT-PCR) and western blot. We obtained 7,901 lncRNAs from RNAseq. A total of 1,062 host-encoded lncRNAs were significantly differentially regulated by IFNα treatment. We found that lncRNA-IFI6 gRNA significantly inhibited HCV infection compared with negative gRNA control. The expression of the antiviral ISG IFI6 was significantly increased following lncRNA-IFI6 gRNA editing compared with negative gRNA control in Japanese fulminant hepatitis 1 (JFH1)-infected Huh7.5.1 cells and PHHs. We observed that lncRNA-IFI6 regulation of HCV was independent of Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling. lncRNA-IFI6 negatively regulated IFI6 promoter function through histone modification. Overexpression of the truncated spatial domain or full-length lncRNA-IFI6 inhibited IFI6 expression and increased HCV replication. Conclusion: A lncRNA, lncRNA-IFI6, regulates antiviral innate immunity in the JFH1 HCV infection model. lncRNA-IFI6 regulates HCV infection independently of the JAK-STAT pathway. lncRNA-IFI6 exerts its regulatory function via promoter activation and histone modification of IFI6 through its spatial domain.


Asunto(s)
Hepacivirus/fisiología , Hepatitis C/virología , Interferón-alfa/fisiología , ARN Largo no Codificante/fisiología , Células Cultivadas , Humanos
2.
Scand J Rheumatol ; 49(2): 122-130, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31612777

RESUMEN

Objective: The aim of this study was to investigate whether incident proteinuria in patients with systemic lupus erythematosus (SLE) was preceded by changes in blood lymphocytes and neutrophil counts and/or neutrophil-lymphocyte ratio (NLR).Method: SLE patients with no proteinuria before or at the time of classification were included. Longitudinal data on SLE manifestations, vital status, and SLE-associated medications were collected during clinical visits and chart review. Laboratory data were collected through a nationwide database. Lymphopenia, severe lymphopenia, and neutropenia were defined as values below 0.8 × 109, 0.5 × 109, and 2.0 × 109 cells/L, respectively. High NLR was defined as values above the median. Proteinuria was defined by at least two measurements of elevated urine protein excretion (> 0.5 g/day). Hazard ratios (HRs) were calculated by Cox modelling using time-dependent continuous and binary covariates based on multiple laboratory measurements adjusted for use of immunosuppressants.Results: In total, 260 SLE patients were available for the analysis, of whom 30 (12%) developed incident proteinuria following the diagnosis of SLE. Median follow-up time was 73.5 months. Lymphocyte and neutrophil counts, but not NLR, were associated with incident proteinuria. HRs for incident proteinuria were 2.71 for lymphopenia [95% confidence interval (CI) 1.20-6.11], 4.73 for severe lymphopenia (95% CI 1.93-11.59), and 2.54 for neutropenia (95% CI 1.14-5.65).Conclusion: Lymphopenia and neutropenia predicted the risk of first-time proteinuria independently of immunosuppressants.


Asunto(s)
Lupus Eritematoso Sistémico/complicaciones , Linfopenia/complicaciones , Neutropenia/complicaciones , Proteinuria/etiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Femenino , Humanos , Inmunosupresores/uso terapéutico , Interferón-alfa/fisiología , Estudios Longitudinales , Nefritis Lúpica/etiología , Masculino , Persona de Mediana Edad , Modelos de Riesgos Proporcionales , Adulto Joven
3.
Brain Behav Immun ; 79: 174-185, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30711510

RESUMEN

Neuroinflammation occurs after germinal matrix hemorrhage (GMH) and induces secondary brain injury. Interferon-α (IFN-α) has been shown to exert anti-inflammatory effects in infectious diseases via activating IFNAR and its downstream signaling. We aimed to investigate the anti-inflammatory effects of Recombinant human IFN-α (rh-IFN-α) and the underlying mechanisms in a rat GMH model. Two hundred and eighteen P7 rat pups of both sexes were subjected to GMH by an intraparenchymal injection of bacterial collagenase. Rh-IFN-α was administered intraperitoneally. Small interfering RNA (siRNA) of IFNAR, and siRNA of tumor necrosis factor receptor associated factor 3 (TRAF3) were administered through intracerebroventricular (i.c.v.) injections. JAK1 inhibitor ruxolitinib was given by oral lavage. Post-GMH evaluation included neurobehavioral function, Nissl staining, Western blot analysis, and immunofluorescence. Our results showed that endogenous IFN-α and phosphorylated IFNAR levels were increased after GMH. Administration of rh-IFN-α improved neurological functions, attenuated neuroinflammation, inhibited microglial activation, and ameliorated post-hemorrhagic hydrocephalus after GMH. These observations were concomitant with IFNAR activation, increased expression of phosphorylated JAK1, phosphorylated STAT1 and TRAF3, and decreased levels of phosphorylated NF-κB, IL-6 and TNF-α. Specifically, knockdown of IFNAR, JAK1 and TRAF3 abolished the protective effects of rh-IFN-α. In conclusion, our findings demonstrated that rh-IFN-α treatment attenuated neuroinflammation, neurological deficits and hydrocephalus formation through inhibiting microglial activation after GMH, which might be mediated by IFNAR/JAK1-STAT1/TRAF3/NF-κB signaling pathway. Rh-IFN-α may be a promising therapeutic agent to attenuate brain injury via its anti-inflammatory effect.


Asunto(s)
Hemorragia Cerebral Intraventricular/inmunología , Interferón-alfa/metabolismo , Neuroinmunomodulación/fisiología , Animales , Animales Recién Nacidos , Lesiones Encefálicas/metabolismo , Hemorragia Cerebral Intraventricular/inducido químicamente , Hemorragia Cerebral Intraventricular/fisiopatología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Proteínas I-kappa B/metabolismo , Inflamación/metabolismo , Interferón-alfa/farmacología , Interferón-alfa/fisiología , Janus Quinasa 1/metabolismo , Janus Quinasa 1/fisiología , Masculino , Microglía/metabolismo , FN-kappa B/inmunología , FN-kappa B/metabolismo , Neuroinmunomodulación/inmunología , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT1/fisiología , Transducción de Señal/efectos de los fármacos , Factor 3 Asociado a Receptor de TNF/metabolismo
4.
J Biol Chem ; 291(12): 6158-68, 2016 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-26817845

RESUMEN

Adenosine deaminases acting on double-stranded RNA (ADARs) catalyze the deamination of adenosine (A) to produce inosine (I) in double-stranded (ds) RNA structures, a process known as A-to-I RNA editing. dsRNA is an important trigger of innate immune responses, including interferon (IFN) production and action. We examined the role of A-to-I RNA editing by two ADARs, ADAR1 and ADAR2, in the sensing of self-RNA in the absence of pathogen infection, leading to activation of IFN-induced, RNA-mediated responses in mouse embryo fibroblasts. IFN treatment of Adar1(-/-) cells lacking both the p110 constitutive and p150 IFN-inducible ADAR1 proteins induced formation of stress granules, whereas neither wild-type (WT) nor Adar2(-/-) cells displayed a comparable stress granule response following IFN treatment. Phosphorylation of protein synthesis initiation factor eIF2α at serine 51 was increased in IFN-treated Adar1(-/-) cells but not in either WT or Adar2(-/-) cells following IFN treatment. Analysis by deep sequencing of mouse exonic loci containing A-to-I-editing sites revealed that the majority of editing in mouse embryo fibroblasts was carried out by ADAR1. IFN treatment increased editing in both WT and Adar2(-/-) cells but not in either Adar1(-/-) or Adar1(-/-) (p150) cells or Stat1(-/-) or Stat2(-/-) cells. Hyper-edited sites found in predicted duplex structures showed strand bias of editing for some RNAs. These results implicate ADAR1 p150 as the major A-to-I editor in mouse embryo fibroblasts, acting as a feedback suppressor of innate immune responses otherwise triggered by self-RNAs possessing regions of double-stranded character.


Asunto(s)
Adenosina Desaminasa/fisiología , Inmunidad Innata , Edición de ARN , ARN Bicatenario/metabolismo , Animales , Células Cultivadas , Gránulos Citoplasmáticos/metabolismo , Desaminación , Factor 2 Eucariótico de Iniciación/metabolismo , Fibroblastos/metabolismo , Tolerancia Inmunológica , Interferón-alfa/fisiología , Ratones Noqueados , Fosforilación , Procesamiento Proteico-Postraduccional , ARN Bicatenario/genética , Proteínas de Unión al ARN/fisiología , Transducción de Señal
5.
Mediators Inflamm ; 2017: 4532409, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29403161

RESUMEN

The roles of pDC and IFN-α have not been well defined in IgA nephropathy (IgAN). In this study, we investigated the abundance of pDCs and IFN-α in IgAN patients and the response of peripheral blood mononuclear cells (PBMCs) after stimulation of the pDC-preferred TLR9 ligand CpG2216. The effects of IFN-α on plasma cell differentiation and leukocyte migration were also investigated. Here, we found that the percentages of pDCs were increased in PBMCs of IgAN patients, than in those of healthy controls. Plasma levels of IFN-α proteins and abundance of plasma cells were higher in IgAN patients than in healthy donors. Plasma IFN-α levels were positively associated with proteinuria, renal IgM deposition, and renal tubular atrophy/interstitial fibrosis grade in IgAN patients. Ex vivo activation of TLR9 on pDCs resulted in increased IFN-α production and enhanced plasma cell differentiation in IgAN patients as compared with healthy donors. IFN-α treatment led to increased plasma cell differentiation in vitro. IFN-α also significantly promoted expression of chemokines IP-10 and MCP-1 in human mesangial cells, which subsequently facilitated the transendothelial migration of human CD4+ and CD14+ cells. In conclusion, pDC and its secreted cytokine IFN-α may play important roles in pathological changes of IgA nephropathy.


Asunto(s)
Células Dendríticas/fisiología , Glomerulonefritis por IGA/patología , Interferón-alfa/fisiología , Células Plasmáticas/citología , Adulto , Linfocitos T CD4-Positivos/fisiología , Diferenciación Celular , Movimiento Celular , Femenino , Glomerulonefritis por IGA/inmunología , Humanos , Masculino , Receptor Toll-Like 9/fisiología
7.
J Biol Chem ; 290(45): 27345-27359, 2015 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-26370074

RESUMEN

Siglec-1 (sialoadhesin, CD169) is a surface receptor on human cells that mediates trans-enhancement of HIV-1 infection through recognition of sialic acid moieties in virus membrane gangliosides. Here, we demonstrate that mouse Siglec-1, expressed on the surface of primary macrophages in an interferon-α-responsive manner, captures murine leukemia virus (MLV) particles and mediates their transfer to proliferating lymphocytes. The MLV infection of primary B-cells was markedly more efficient than that of primary T-cells. The major structural protein of MLV particles, Gag, frequently co-localized with Siglec-1, and trans-infection, primarily of surface-bound MLV particles, efficiently occurred. To explore the role of sialic acid for MLV trans-infection at a submolecular level, we analyzed the potential of six sialic acid precursor analogs to modulate the sialylated ganglioside-dependent interaction of MLV particles with Siglec-1. Biosynthetically engineered sialic acids were detected in both the glycolipid and glycoprotein fractions of MLV producer cells. MLV released from cells carrying N-acyl-modified sialic acids displayed strikingly different capacities for Siglec-1-mediated capture and trans-infection; N-butanoyl, N-isobutanoyl, N-glycolyl, or N-pentanoyl side chain modifications resulted in up to 92 and 80% reduction of virus particle capture and trans-infection, respectively, whereas N-propanoyl or N-cyclopropylcarbamyl side chains had no effect. In agreement with these functional analyses, molecular modeling indicated reduced binding affinities for non-functional N-acyl modifications. Thus, Siglec-1 is a key receptor for macrophage/lymphocyte trans-infection of surface-bound virions, and the N-acyl side chain of sialic acid is a critical determinant for the Siglec-1/MLV interaction.


Asunto(s)
Virus de la Leucemia Murina de Moloney/patogenicidad , Lectina 1 Similar a Ig de Unión al Ácido Siálico/química , Lectina 1 Similar a Ig de Unión al Ácido Siálico/fisiología , Animales , Sitios de Unión , Línea Celular , Gangliósidos/química , Gangliósidos/metabolismo , Interacciones Huésped-Patógeno/fisiología , Humanos , Interferón-alfa/fisiología , Leucemia Experimental/fisiopatología , Leucemia Experimental/virología , Linfocitos/fisiología , Linfocitos/virología , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Moleculares , Virus de la Leucemia Murina de Moloney/genética , Virus de la Leucemia Murina de Moloney/fisiología , Ácido N-Acetilneuramínico/química , Receptores Virales/química , Receptores Virales/fisiología , Infecciones por Retroviridae/fisiopatología , Infecciones por Retroviridae/virología , Lectina 1 Similar a Ig de Unión al Ácido Siálico/genética , Infecciones Tumorales por Virus/fisiopatología , Infecciones Tumorales por Virus/virología
8.
Cytokine ; 80: 48-55, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26945996

RESUMEN

We aimed to investigate regulation of signal transducer and activator of transcription 3 (STAT3) and suppressor of cytokine signaling 3 (SOCS3) by interferon alpha (IFN-α) and to analyze the relationship between STAT3 and SOCS3 during hepatitis C virus (HCV) infection. Changes in STAT3 and SOCS3 were analyzed at both mRNA and protein levels in human hepatoma cells infected with HCV (J6/JFH1). At 72h of HCV infection, STAT3 expression was decreased with sustained phosphorylation, and IFN-α increased such decrease and phosphorylation. HCV increased SOCS3 expression, while IFN-α impaired such increase, indicating different regulation of STAT3 and SOCS3 by IFN-α. IFN-α-induced expression and phosphorylation of upstream kinases of the JAK/STAT pathway, Tyk2 and Jak1, were suppressed by HCV. Moreover, knockdown of STAT3 by RNA interference led to decreases in HCV RNA replication and viral protein expression, without affecting either the expression of Tyk2 and Jak1 or the SOCS3 induction in response to IFN-α. These results show that IFN-α antagonizes STAT3 and SOCS3 signaling triggered by HCV and that STAT3 regulation correlates inversely with SOCS3 induction by IFN-α, which may be important in better understanding the complex interplay between IFN-α and signal molecules during HCV infection.


Asunto(s)
Hepacivirus/fisiología , Interferón-alfa/fisiología , Factor de Transcripción STAT3/antagonistas & inhibidores , Transducción de Señal , Proteínas Supresoras de la Señalización de Citocinas/antagonistas & inhibidores , Carcinoma Hepatocelular , Línea Celular Tumoral , Regulación de la Expresión Génica , Humanos , Interferón alfa-2 , Interferón-alfa/farmacología , Neoplasias Hepáticas , Proteínas Recombinantes/farmacología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Replicación Viral
9.
Brain Behav Immun ; 58: 31-39, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26697999

RESUMEN

Interferon-alpha (IFN-α) is a key mediator of antiviral immune responses used to treat Hepatitis C infection. Though clinically effective, IFN-α rapidly impairs mood, motivation and cognition, effects that can appear indistinguishable from major depression and provide powerful empirical support for the inflammation theory of depression. Though inflammation has been shown to modulate activity within discrete brain regions, how it affects distributed information processing and the architecture of whole brain functional connectivity networks have not previously been investigated. Here we use a graph theoretic analysis of resting state functional magnetic resonance imaging (rfMRI) to investigate acute effects of systemic interferon-alpha (IFN-α) on whole brain functional connectivity architecture and its relationship to IFN-α-induced mood change. Twenty-two patients with Hepatitis-C infection, initiating IFN-α-based therapy were scanned at baseline and 4h after their first IFN-α dose. The whole brain network was parcellated into 110 cortical and sub-cortical nodes based on the Oxford-Harvard Atlas and effects assessed on higher-level graph metrics, including node degree, betweenness centrality, global and local efficiency. IFN-α was associated with a significant reduction in global network connectivity (node degree) (p=0.033) and efficiency (p=0.013), indicating a global reduction of information transfer among the nodes forming the whole brain network. Effects were similar for highly connected (hub) and non-hub nodes, with no effect on betweenness centrality (p>0.1). At a local level, we identified regions with reduced efficiency of information exchange and a sub-network with decreased functional connectivity after IFN-α. Changes in local and particularly global functional connectivity correlated with associated changes in mood measured on the Profile of Mood States (POMS) questionnaire. IFN-α rapidly induced a profound shift in whole brain network structure, impairing global functional connectivity and the efficiency of parallel information exchange. Correlations with multiple indices of mood change support a role for global changes in brain functional connectivity architecture in coordinated behavioral responses to IFN-α.


Asunto(s)
Encéfalo/fisiología , Interferón-alfa/fisiología , Adulto , Encéfalo/efectos de los fármacos , Mapeo Encefálico , Femenino , Humanos , Interferón-alfa/administración & dosificación , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad
10.
J Immunol ; 193(3): 1024-34, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24973447

RESUMEN

Increasing evidence suggests that type 1 IFN (IFN-αß) is associated with pathogenesis of Th1-mediated type 1 diabetes (T1D). A major source of IFN-αß is plasmacytoid dendritic cells (pDCs). In this study, we analyzed peripheral blood pDC numbers and functions in at-risk, new-onset, and established T1D patients and controls. We found that subjects at risk for T1D and new-onset and established T1D subjects possessed significantly increased pDCs but similar number of myeloid DCs when compared with controls. pDC numbers were not affected by age in T1D subjects but declined with increasing age in control subjects. It was demonstrated that IFN-α production by PBMCs stimulated with influenza viruses was significantly higher in T1D subjects than in controls, and IFN-α production was correlated with pDC numbers in PBMCs. Of interest, only T1D-associated Coxsackievirus serotype B4 but not B3 induced majority of T1D PBMCs to produce IFN-α, which was confirmed to be secreted by pDCs. Finally, in vitro studies demonstrated IFN-α produced by pDCs augmented Th1 responses, with significantly greater IFN-γ-producing CD4(+) T cells from T1D subjects. These findings indicate that increased pDCs and their IFN-αß production may be associated with this Th1-mediated autoimmune disease, especially under certain viral infections linked to T1D pathogenesis.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/patología , Diabetes Mellitus Tipo 1/inmunología , Interferón-alfa/biosíntesis , Células TH1/inmunología , Regulación hacia Arriba/inmunología , Adolescente , Adulto , Recuento de Células Sanguíneas , Diferenciación Celular/inmunología , Células Cultivadas , Niño , Preescolar , Células Dendríticas/virología , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/virología , Femenino , Humanos , Lactante , Interferón-alfa/fisiología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/patología , Leucocitos Mononucleares/virología , Masculino , Orthomyxoviridae/inmunología , Células TH1/patología , Células TH1/virología , Adulto Joven
11.
Cell Mol Life Sci ; 72(14): 2749-61, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25746225

RESUMEN

A new form of circuitry for gene regulation has been identified in which RNAs can crosstalk by competing for shared microRNAs (miRNAs). Such competing endogenous RNAs (ceRNAs) form a network via shared miRNA response elements (MREs) to antagonize miRNA function. We previously reported natural antisense RNA (AS) as an important modulator of interferon-α1 (IFN-α1) mRNA levels by promoting IFN-α1 mRNA stability. We show that IFN-α1 AS forms a ceRNA network with specific IFN-α AS (IFN-α7/-α8/-α10/-α14) and mRNA (IFN-α8/-α10/-α14/-α17) subtypes from the IFN-α gene (IFNA) family to antagonize miRNA-1270 (miR-1270), thereby modulating IFN-α1 mRNA levels. Bioinformatic analysis demonstrated that IFN-α1 AS harbors multiple miR-1270 MREs (MRE-1270s), whose presence was substantiated by miR-1270 overexpression and transfection of antimiR-1270. The antimiR-1270, complementary to the miR-1270 seed region, revealed that IFN-α1 AS likely shares the MRE-1270 with IFN-α1 mRNA and specific IFN-α AS and mRNA subtypes. Subsequent bioinformatic analysis for MRE-1270s showed that IFN-α1 AS and other RNA subtypes shared the 6-mer MRE-1270 site. Further MRE-mapping demonstrated that the total number of MRE-1270s in IFN-α1 AS accounted for approximately 30 % of the miR-1270 population. AntimiR-1270 transfection also caused specific de-repression of five cellular mRNAs, including that of CAPRIN1. These results suggest that IFN-α1 AS, together with specific IFN-α AS and mRNA subtypes, as well as the five cellular mRNAs, participate as competing molecules in the ceRNA network against miR-1270. This coordinated regulatory architecture suggests a vital function for the innate immune system in maintaining precise physiological type I IFN levels via post-transcriptional regulatory mechanisms.


Asunto(s)
Regulación de la Expresión Génica , Interferón-alfa/fisiología , MicroARNs/metabolismo , Sitios de Unión , Línea Celular , Silenciador del Gen , Humanos , Interferón-alfa/metabolismo , ARN sin Sentido/metabolismo , ARN Mensajero/metabolismo
12.
Biologicals ; 43(4): 225-31, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26073788

RESUMEN

Combined use of interferon (IFN) and thymosin (THY) holds a stronger antiviral effect than when applied individually because of their coordination and complementary action. In this study, prokaryotic expressed porcine IFNα1 (poIFNα1) or the porcine IFNα1-THYα1 fusion protein coding with the Escherichia coli preferred codon sequences connected by the three different linkers were gained in the unlabeled pRSFDDuet-1 expression systems and purified using the strong anion-exchange chromatography and hydrophobic chromatography (among which, one was digested by thrombin because the cleavage site was included in the linker). Then, the activities of IFN and THY in the fusion protein were detected using the cytopathic effect inhibition assay and T-cell activity assays. SDS PAGE and western blotting results showed that the poIFNα1 or the three poIFNα1-THYα1 fusion proteins with three different linkers were expressed solubly in E. coli. The poIFNα1 protein and three types of poIFNα1-THYα1 fusion proteins with >90% purity were gained. The poIFNα1-LinkerB-THYα1 fusion protein showed the highest interferon activity compared with the others (P < 0.001), and the poIFNα1-LinkerA-THYα1 fusion protein highest thymosin activity (P < 0.05). In this study, a preliminary experiment was conducted for the expression of the poIFNα1 and THYα1 fusion proteins.


Asunto(s)
Interferón-alfa/fisiología , Proteínas Recombinantes de Fusión/biosíntesis , Timosina/análogos & derivados , Animales , Técnicas In Vitro , Interferón-alfa/genética , Proteínas Recombinantes de Fusión/genética , Porcinos , Trombina/metabolismo , Timalfasina , Timosina/genética , Timosina/fisiología
13.
J Biol Chem ; 288(24): 17261-71, 2013 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-23649619

RESUMEN

Reoviruses are double-stranded RNA viruses that infect the mammalian respiratory and gastrointestinal tract. Reovirus infection elicits production of type I interferons (IFNs), which trigger antiviral pathways through the induction of interferon-stimulated genes (ISGs). Although hundreds of ISGs have been identified, the functions of many of these genes are unknown. The interferon-inducible transmembrane (IFITM) proteins are one class of ISGs that restrict the cell entry of some enveloped viruses, including influenza A virus. One family member, IFITM3, localizes to late endosomes, where reoviruses undergo proteolytic disassembly; therefore, we sought to determine whether IFITM3 also restricts reovirus entry. IFITM3-expressing cell lines were less susceptible to infection by reovirus, as they exhibited significantly lower percentages of infected cells in comparison to control cells. Reovirus replication was also significantly reduced in IFITM3-expressing cells. Additionally, cells expressing an shRNA targeting IFITM3 exhibited a smaller decrease in infection after IFN treatment than the control cells, indicating that endogenous IFITM3 restricts reovirus infection. However, IFITM3 did not restrict entry of reovirus infectious subvirion particles (ISVPs), which do not require endosomal proteolysis, indicating that restriction occurs in the endocytic pathway. Proteolysis of outer capsid protein µ1 was delayed in IFITM3-expressing cells in comparison to control cells, suggesting that IFITM3 modulates the function of late endosomal compartments either by reducing the activity of endosomal proteases or delaying the proteolytic processing of virions. These data provide the first evidence that IFITM3 restricts infection by a nonenveloped virus and suggest that IFITM3 targets an increasing number of viruses through a shared requirement for endosomes during cell entry.


Asunto(s)
Orthoreovirus Mamífero 3/fisiología , Proteínas de la Membrana/fisiología , Proteínas de Unión al ARN/fisiología , Internalización del Virus , Cápside/metabolismo , Endocitosis , Endosomas/virología , Expresión Génica , Técnicas de Silenciamiento del Gen , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Interferón-alfa/fisiología , Cinética , Orthoreovirus de los Mamíferos/fisiología , ARN Interferente Pequeño/genética , Virión/fisiología , Ensamble de Virus , Replicación Viral
14.
Infect Immun ; 82(4): 1698-709, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24491580

RESUMEN

Human monocytic ehrlichiosis (HME) is caused by a tick-borne obligate intracellular pathogen of the order Rickettsiales. HME disease can range from mild to a fatal, toxic shock-like syndrome, yet the mechanisms regulating pathogenesis are not well understood. We define a central role for type I interferons (alpha interferon [IFN-α] and IFN-ß) in severe disease in a mouse model of fatal ehrlichiosis caused by Ixodes ovatus Ehrlichia (IOE). IFN-α and IFN-ß were induced by IOE infection but not in response to a less virulent strain, Ehrlichia muris. The major sources of type I IFNs during IOE infection were plasmacytoid dendritic cells and monocytes. Mice lacking the receptor for type I IFNs (Ifnar deficient) or neutralization of IFN-α and IFN-ß resulted in a reduced bacterial burden. Ifnar-deficient mice exhibited significantly increased survival after IOE infection, relative to that of wild-type (WT) mice, that correlated with increased type II IFN (IFN-γ) production. Pathogen-specific antibody responses were also elevated in Ifnar-deficient mice, and this required IFN-γ. Remarkably, increased IFN-γ and IgM were not essential for protection in the absence of type I IFN signaling. The direct effect of type I IFNs on hematopoietic and nonhematopoietic cells was evaluated in bone marrow chimeric mice. We observed that chimeric mice containing Ifnar-deficient hematopoietic cells succumbed to infection early, whereas Ifnar-deficient mice containing WT hematopoietic cells exhibited increased survival, despite having a higher bacterial burden. These data demonstrate that IFN-α receptor signaling in nonhematopoietic cells is important for pathogenesis. Thus, type I IFNs are induced during a rickettsial infection in vivo and promote severe disease.


Asunto(s)
Ehrlichia/patogenicidad , Ehrlichiosis/inmunología , Interferón Tipo I/fisiología , Interferón-alfa/fisiología , Interferón gamma/fisiología , Análisis de Varianza , Animales , Linfocitos T CD8-positivos/metabolismo , Modelos Animales de Enfermedad , Inmunoglobulina M/fisiología , Ratones , Ratones Endogámicos C57BL
15.
Biochem Biophys Res Commun ; 450(4): 1462-8, 2014 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-25019997

RESUMEN

Sterile alpha motif and HD-domain containing protein 1 (SAMHD1) is a newly identified intracellular antiviral factor. By depleting the dNTPs pool of host cells to a low level that cannot support the efficient synthesis of viral cDNA, it restricts replication of some retroviruses. As a DNA virus, Hepatitis B virus (HBV) experiences a process of reverse transcription in its life cycle akin to that of retroviruses. However, whether SAMHD1 can restrict HBV replication in liver cells is unknown. Here, we reported that SAMHD1 expression was detectable in four liver cell lines. Exogenous expression of SAMHD1 in SMMC-7721 cells restrained HBV replication. Similarly, SAMHD1 impeded HBV replication in another liver cell line, BEL-7402. Remarkably, the catalytically inactive mutant, SAMHD1 HD/AA also hampered HBV replication. Additionally, HBV replication reduced SAMHD1 expression in HepG2 cells. Moreover, it was found that IFN-α induced expression of SAMHD1 in liver cells. Together, these findings suggested that IFN-α-inducible SAMHD1 inhibited HBV replication in liver cells.


Asunto(s)
Virus de la Hepatitis B/fisiología , Proteínas de Unión al GTP Monoméricas/fisiología , Replicación Viral/fisiología , Western Blotting , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Humanos , Interferón-alfa/fisiología , Proteína 1 que Contiene Dominios SAM y HD
16.
J Pharmacol Exp Ther ; 348(1): 96-105, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24133297

RESUMEN

GS-9620 [8-(3-(pyrrolidin-1-ylmethyl)benzyl)-4-amino-2-butoxy-7,8-dihydropteridin-6(5H)-one] is a potent, orally bioavailable small-molecule agonist of Toll-like receptor 7 (TLR7) developed for finite treatment of chronic hepatitis B viral (HBV) infection, with the goal of inducing a liver-targeted antiviral effect without inducing the adverse effects associated with current systemic interferon-α (IFN-α) therapies. We characterized the pharmacodynamic response of GS-9620 in CD-1 mice and cynomolgus monkeys following intravenous or oral administration and showed that GS-9620 induces the production of select chemokines and cytokines, including IFN-α and interferon-stimulated genes (ISGs). It is noteworthy that we also demonstrated that, in animals and healthy human volunteers, oral administration of GS-9620 can induce a type I interferon-dependent antiviral innate immune response, as measured by whole-blood mRNA of the ISGs 2'5'-oligoadenylate synthetase 1 (OAS1) and myxovirus resistance 1 (MX1), without the induction of detectable systemic IFN-α, i.e., a presystemic response. Additionally, presystemic induction of hepatic OAS1 and MX1 mRNA was observed in CD-1 mice in the absence of detectable systemic IFN-α. We propose that the mechanism of this presystemic response is likely its high intestinal absorption, which facilitates localized activation of TLR7, probably in plasmacytoid dendritic cells at the level of gut-associated lymphoid tissue and/or the liver. This localized response is further supported by data that indicate only minimal contributions of systemic immune stimulation to the overall pharmacodynamic response to orally administered GS-9620. These data demonstrate that GS-9620 can induce an antiviral innate immune response without inducing a systemic IFN-α response and thus suggest the therapeutic potential of this approach in the treatment of chronic HBV infection.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Interferón-alfa/fisiología , Pteridinas/farmacología , Pteridinas/farmacocinética , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/biosíntesis , Administración Oral , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Interferón-alfa/sangre , Macaca fascicularis , Ratones , Pteridinas/administración & dosificación , Receptor Toll-Like 7/genética
18.
Psychol Med ; 44(4): 789-95, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23659574

RESUMEN

BACKGROUND: The development of depressive symptomatology is a recognized complication of treatment with the cytokine interferon-α (IFN-α) and has been seen as supporting inflammatory theories of the pathophysiology of major depression. Major depression has been associated with changes in glutamatergic activity and recent formulations of IFN-induced depression have implicated neurotoxic influences that could also lead to changes in glutamate function. The present study used magnetic resonance spectroscopy (MRS) to measure glutamate and its major metabolite glutamine in patients with hepatitis C who received treatment with pegylated IFN-α and ribavirin. METHOD: MRS measurements of glutamate and glutamine were taken from a 25 × 20 × 20 mm voxel including the pregenual anterior cingulate cortex in 12 patients before and after 4-6 weeks of treatment with IFN. RESULTS: IFN treatment led to an increase in cortical levels of glutamine (p = 0.02) and a significant elevation in the ratio of glutamine to glutamate (p < 0.01). Furthermore, changes in glutamine level correlated significantly with ratings of depression and anxiety at the time of the second scan. CONCLUSIONS: We conclude that treatment with IFN-α is associated with MRS-visible changes in glutamatergic metabolism. However, the changes seen differ from those reported in major depression, which suggests that the pathophysiology of IFN-induced depression may be distinct from that of major depression more generally.


Asunto(s)
Corteza Cerebral , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Hepatitis C/metabolismo , Interferón-alfa/administración & dosificación , Espectroscopía de Resonancia Magnética , Adulto , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Depresión/diagnóstico , Depresión/tratamiento farmacológico , Depresión/metabolismo , Femenino , Ácido Glutámico/biosíntesis , Ácido Glutámico/fisiología , Glutamina/biosíntesis , Glutamina/fisiología , Hepatitis C/diagnóstico , Hepatitis C/tratamiento farmacológico , Humanos , Inflamación/diagnóstico , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Interferón-alfa/efectos adversos , Interferón-alfa/fisiología , Masculino , Persona de Mediana Edad , Protones
19.
J Immunol ; 189(7): 3299-310, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22925929

RESUMEN

Previous mouse and human studies have demonstrated that direct IFN-α/ß signaling on naive CD8 T cells is critical to support their expansion and acquisition of effector functions. In this study, we show that human naive CD8 T cells primed in the presence of IFN-α possess a heightened ability to respond to homeostatic cytokines and to secondary Ag stimulation, but rather than differentiating to effector or memory CTLs, they preserve nature-like phenotypic features. These are qualities associated with greater efficacy in adoptive immunotherapy. In a mouse model of adoptive transfer, CD8 T cells primed in the presence of IFN-α are able to persist and to mediate a robust recall response even after a long period of naturally driven homeostatic maintenance. The long-lasting persistence of IFN-α-primed CD8 T cells is favored by their enhanced responsiveness to IL-15 and IL-7, as demonstrated in IL-15(-/-) and IL-7(-/-) recipient mice. In humans, exposure to IFN-α during in vitro priming of naive HLA-A2(+) CD8 T cells with autologous dendritic cells loaded with MART1(26-35) peptide renders CD8 T cells with an improved capacity to respond to homeostatic cytokines and to specifically lyse MART1-expressing melanoma cells. Furthermore, in a mouse model of melanoma, adoptive transfer of tumor-specific CD8 T cells primed ex vivo in the presence of IFN-α exhibits an improved ability to contain tumor progression. Therefore, exposure to IFN-α during priming of naive CD8 T cells imprints decisive information on the expanded cells that can be exploited to improve the efficacy of adoptive T cell therapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Citocinas/fisiología , Homeostasis/inmunología , Inmunización Secundaria/métodos , Memoria Inmunológica , Interferón-alfa/fisiología , Activación de Linfocitos/inmunología , Linfocitos T Citotóxicos/inmunología , Traslado Adoptivo/métodos , Animales , Antígenos/fisiología , Linfocitos T CD8-positivos/trasplante , Células Cultivadas , Humanos , Interleucina-15/fisiología , Interleucina-17/fisiología , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Transducción de Señal/inmunología , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/trasplante
20.
J Immunol ; 189(2): 659-68, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22706089

RESUMEN

Autocrine IFN-γ signaling is important for CD4 differentiation to Th1 effector cells, but it has been unclear whether it contributes to CD8 T cell differentiation. We show in this paper that naive murine CD8 T cells rapidly and transiently produce low levels of IFN-γ upon stimulation with Ag and B7-1, with production peaking at ∼8 h and declining by 24 h. The autocrine IFN-γ signals for upregulation of expression of T-bet and granzyme B and induces weak cytolytic activity and effector IFN-γ production. IFN-α acts synergistically with IFN-γ to support development of strong effector functions, whereas IL-12 induces high T-bet expression and strong function in the absence of IFN-γ signaling. Thus, IFN-γ is not only an important CD8 T cell effector cytokine, it is an autocrine/paracrine factor whose contributions to differentiation vary depending on whether the response is supported by IL-12 or type I IFN.


Asunto(s)
Comunicación Autocrina/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/inmunología , Interferón-alfa/fisiología , Interferón gamma/fisiología , Traslado Adoptivo/métodos , Animales , Comunicación Autocrina/genética , Linfocitos T CD8-positivos/citología , Diferenciación Celular/genética , Línea Celular Tumoral , Células Cultivadas , Interferón gamma/deficiencia , Interferón gamma/genética , Interleucina-12/fisiología , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Comunicación Paracrina/genética , Comunicación Paracrina/inmunología , Proteínas de Dominio T Box/biosíntesis , Proteínas de Dominio T Box/fisiología , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda