Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 438
Filtrar
1.
Front Immunol ; 12: 748423, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34691060

RESUMO

The Type I Interferon family of cytokines all act through the same cell surface receptor and induce phosphorylation of the same subset of response regulators of the STAT family. Despite their shared receptor, different Type I Interferons have different functions during immune response to infection. In particular, they differ in the potency of their induced anti-viral and anti-proliferative responses in target cells. It remains not fully understood how these functional differences can arise in a ligand-specific manner both at the level of STAT phosphorylation and the downstream function. We use a minimal computational model of Type I Interferon signaling, focusing on Interferon-α and Interferon-ß. We validate the model with quantitative experimental data to identify the key determinants of specificity and functional plasticity in Type I Interferon signaling. We investigate different mechanisms of signal discrimination, and how multiple system components such as binding affinity, receptor expression levels and their variability, receptor internalization, short-term negative feedback by SOCS1 protein, and differential receptor expression play together to ensure ligand specificity on the level of STAT phosphorylation. Based on these results, we propose phenomenological functional mappings from STAT activation to downstream anti-viral and anti-proliferative activity to investigate differential signal processing steps downstream of STAT phosphorylation. We find that the negative feedback by the protein USP18, which enhances differences in signaling between Interferons via ligand-dependent refractoriness, can give rise to functional plasticity in Interferon-α and Interferon-ß signaling, and explore other factors that control functional plasticity. Beyond Type I Interferon signaling, our results have a broad applicability to questions of signaling specificity and functional plasticity in signaling systems with multiple ligands acting through a bottleneck of a small number of shared receptors.


Assuntos
Interferon-alfa/fisiologia , Interferon beta/fisiologia , Modelos Imunológicos , Receptor Cross-Talk/fisiologia , Receptor de Interferon alfa e beta/fisiologia , Transdução de Sinais/fisiologia , Animais , Simulação por Computador , Dimerização , Retroalimentação Fisiológica , Feminino , Humanos , Concentração Inibidora 50 , Cinética , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Mapeamento de Interação de Proteínas , Fatores de Transcrição STAT/metabolismo , Baço/citologia , Proteína 1 Supressora da Sinalização de Citocina/fisiologia , Linfócitos T/imunologia , Ubiquitina Tiolesterase
2.
Elife ; 92020 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-32452770

RESUMO

Junín virus (JUNV) is one of five New World mammarenaviruses (NWMs) that causes fatal hemorrhagic disease in humans and is the etiological agent of Argentine hemorrhagic fever (AHF). The pathogenesis underlying AHF is poorly understood; however, a prolonged, elevated interferon-α (IFN-α) response is associated with a negative disease outcome. A feature of all NWMs that cause viral hemorrhagic fever is the use of human transferrin receptor 1 (hTfR1) for cellular entry. Here, we show that mice expressing hTfR1 develop a lethal disease course marked by an increase in serum IFN-α concentration when challenged with JUNV. Further, we provide evidence that the type I IFN response is central to the development of severe JUNV disease in hTfR1 mice. Our findings identify hTfR1-mediated entry and the type I IFN response as key factors in the pathogenesis of JUNV infection in mice.


Assuntos
Antígenos CD/fisiologia , Febre Hemorrágica Americana/virologia , Interações Hospedeiro-Patógeno , Interferon-alfa/fisiologia , Vírus Junin/fisiologia , Receptores da Transferrina/fisiologia , Animais , Camundongos
3.
Scand J Rheumatol ; 49(2): 122-130, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31612777

RESUMO

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.


Assuntos
Lúpus Eritematoso Sistêmico/complicações , Linfopenia/complicações , Neutropenia/complicações , Proteinúria/etiologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Pré-Escolar , Feminino , Humanos , Imunossupressores/uso terapêutico , Interferon-alfa/fisiologia , Estudos Longitudinais , Nefrite Lúpica/etiologia , Masculino , Pessoa de Meia-Idade , Modelos de Riscos Proporcionais , Adulto Jovem
4.
Brain Behav Immun ; 79: 174-185, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30711510

RESUMO

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.


Assuntos
Hemorragia Cerebral Intraventricular/imunologia , Interferon-alfa/metabolismo , Neuroimunomodulação/fisiologia , Animais , Animais Recém-Nascidos , Lesões Encefálicas/metabolismo , Hemorragia Cerebral Intraventricular/induzido quimicamente , Hemorragia Cerebral Intraventricular/fisiopatologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Proteínas I-kappa B/metabolismo , Inflamação/metabolismo , Interferon-alfa/farmacologia , Interferon-alfa/fisiologia , Janus Quinase 1/metabolismo , Janus Quinase 1/fisiologia , Masculino , Microglia/metabolismo , NF-kappa B/imunologia , NF-kappa B/metabolismo , Neuroimunomodulação/imunologia , Ratos , Ratos Sprague-Dawley , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT1/fisiologia , Transdução de Sinais/efeitos dos fármacos , Fator 3 Associado a Receptor de TNF/metabolismo
5.
Hepatology ; 69(3): 1004-1019, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30199576

RESUMO

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.


Assuntos
Hepacivirus/fisiologia , Hepatite C/virologia , Interferon-alfa/fisiologia , RNA Longo não Codificante/fisiologia , Células Cultivadas , Humanos
7.
Vet Immunol Immunopathol ; 191: 51-59, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28895867

RESUMO

Porcine parvovirus type 1 (PPV1) and porcine circovirus type 2 (PCV2) are small single-stranded DNA viruses with high prevalence in the global pig population. The aim of this study was to compare and contrast PCV2 and PPV1 infections in high-health status pigs and to describe PCV2 long-term infection dynamics. Six caesarian-derived colostrum-deprived pigs were randomly divided into two groups and were experimentally infected with PCV2 or PPV1 at 5 weeks of age. All pigs had detectable viremia by day (D) 3 post-infection. Pigs infected with PPV1 had a detectable INF-α response by D3 followed by a high IFN-γ response by D6. The PPV1 pigs developed antibodies against PPV1 by D6 resulting in decreasing virus titers until PPV1 DNA became undetectable from D28 until D42. In contrast, PCV2-infected pigs had no detectable INF-α or IFN-γ response after PCV2 infection. PCV2-infected pigs had no detectable anti-PCV2 humoral response until D49 and had a sustained high level of PCV2 DNA for the duration of the study. While PPV1-infected pigs were clinically normal, PCV2-infected pigs developed severe clinical illness including fatal systemic porcine circovirus associated disease (PCVAD) by D28, fatal enteric PCVAD by D56 and chronic PCVAD manifested as decreased weight gain and periods of diarrhea. Microscopically, all three PCV2-infected pigs had lymphoid lesions consistent with PCVAD and associated with low (chronic disease) to high (acute disease) levels of PCV2 antigen. Under the study conditions, there was a lack of early IFN-γ and INF-α activation followed by a delayed and low humoral immune response and persisting viremia with PCV2 infection. In contrast, PPV1-infected pigs had IFN-γ and INF-α activation and an effective immune response to the PPV1 infection.


Assuntos
Infecções por Circoviridae/veterinária , Circovirus/imunologia , Doenças dos Suínos/virologia , Animais , Anticorpos Antivirais/imunologia , Formação de Anticorpos/fisiologia , Infecções por Circoviridae/imunologia , Infecções por Circoviridae/virologia , Citocinas/fisiologia , DNA Viral/genética , Interferon-alfa/fisiologia , Interferon gama/fisiologia , Masculino , Suínos , Doenças dos Suínos/imunologia , Viremia/imunologia , Viremia/veterinária , Viremia/virologia
8.
Vet Immunol Immunopathol ; 191: 80-93, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28895871

RESUMO

Type I interferons (IFNs) such as IFN-α, IFN-ß, IFN-ε, IFN-κ, and IFN-ω represent cytokines, which are deeply involved in the regulation and activation of innate and adaptive immune responses. They possess strong antiviral, antiproliferative, and immunomodulatory activities allowing their use in the therapy of different viral diseases, neoplasms, and immune-mediated disorders, respectively. Initially, treatment strategies were based on nonspecific inducers of type I IFNs, which were soon replaced by different recombinant proteins. Drugs with type I IFNs as active agents are currently used in the treatment of hepatitis B and C virus infection, lymphoma, myeloid leukemia, renal carcinoma, malignant melanoma, and multiple sclerosis in humans. In addition, recombinant feline IFN-ω has been approved for the treatment of canine parvovirus, feline leukemia virus, and feline immunodeficiency virus infections. However, the role of type I IFNs in the pathogenesis of canine diseases remains largely undetermined so far, even though some share pathogenic mechanisms and clinical features with their human counterparts. This review summarizes the present knowledge of type I IFNs and down-stream targets such as Mx and 2',5'-oligoadenylate synthetase proteins in the pathogenesis of infectious and immune-mediated canine diseases. Moreover, studies investigating the potential use of type I IFNs in the treatment of canine lymphomas, melanomas, sarcomas, and carcinomas, canine distemper virus, parvovirus, and papillomavirus infections as well as immune-mediated keratoconjunctivitis sicca and atopic dermatitis are presented. A separate chapter is dedicated to the therapeutic potential of IFN-λ, a type III IFN, in canine diseases. However, further future studies are still needed to unravel the exact functions of the different subtypes of type I IFNs and their target genes in healthy and diseased dogs and the full potential action of type I IFNs as treatment strategy.


Assuntos
Doenças do Cão/tratamento farmacológico , Interferon Tipo I/uso terapêutico , Animais , Doenças do Cão/imunologia , Cães , Interferon Tipo I/fisiologia , Interferon-alfa/fisiologia , Interferon-alfa/uso terapêutico , Interferon beta/fisiologia , Interferon beta/uso terapêutico , Interferon gama/fisiologia , Interferon gama/uso terapêutico
9.
Arthritis Rheumatol ; 69(12): 2328-2337, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28859258

RESUMO

OBJECTIVE: Systemic lupus erythematosus (SLE) is frequently characterized by activation of the type I interferon (IFN) pathway. We previously observed that a missense single-nucleotide polymorphism (rs1049564) in the purine nucleoside phosphorylase (PNP) gene was associated with high levels of IFN in SLE. PNP is a key enzyme involved in purine metabolism. In this study, we performed functional follow-up of this polymorphism in human cells. METHODS: Type I IFN was measured in patient sera, using a reporter cell assay. Structural modeling of the PNP variant was performed using PyMOL software. PNP messenger RNA (mRNA) and protein levels and type I IFN-induced gene expression were measured in lymphoblastoid cell lines with known PNP rs1049564 genotypes. The cell cycle was assayed using flow cytometry. RESULTS: Structural modeling indicated no major disruption in folding related to rs1049564. We observed that homozygous rs1049564 TT lymphoblastoid cells had decreased PNP mRNA expression and protein levels, and that cells with the TT genotype had reduced PNP enzymatic activity even when the amount of PNP was controlled. Cells with the TT genotype had a 2-fold increase in S-phase block as compared with cells with the homozygous CC phenotype. The S-phase block could be pharmacologically reversed with hypoxanthine and adenosine, supporting the notion that relative PNP deficiency is the cause of the S-phase block. Type I IFN-induced transcripts were increased in a dose-response manner related to the rs1049564 T allele, at both baseline and after type I IFN stimulation. CONCLUSION: The PNP rs1049564 T allele is a loss-of-function variant that induces S-phase block and IFN pathway activation in lymphocytes. The S-phase block could be rescued in our in vitro experiments, suggesting the potential for personalized treatment.


Assuntos
Ciclo Celular/genética , Interferon-alfa/fisiologia , Lúpus Eritematoso Sistêmico/genética , Polimorfismo de Nucleotídeo Único/fisiologia , Purina-Núcleosídeo Fosforilase/genética , Alelos , Ciclo Celular/imunologia , Expressão Gênica , Genótipo , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/fisiopatologia , Fenótipo , Purina-Núcleosídeo Fosforilase/sangue , Transdução de Sinais/genética , Transdução de Sinais/imunologia
11.
Eur J Pharm Sci ; 97: 200-207, 2017 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-27845233

RESUMO

This study is to investigate whether ectoines (ectoine and hydroxyectoine) can reduce aggregation of rhIFNα2b in aqueous solutions on thermal stress. The effect of thermal stress condition on the stability was therefore investigated using size exclusion-high performance liquid chromatography (SE-HPLC), different spectroscopic measurements, dynamic light scattering (DLS), electrophoresis, and differential scanning calorimetry (DSC). All experiments were performed in a sodium phosphate buffer system (100mM, pH7). The protein samples (100µg/ml) were incubated at 50°C for 14days in the absence or presence (1, 10, 20, and 100mM) of ectoines. In summary, thermal-induced aggregation was reduced in the presence of ectoines, regardless of the ectoines concentration in different periods of incubation time by analyzing with SE-HPLC and turbidity measurement. The inhibitory effect of ectoines on the aggregation was shown by other techniques used. The optimal ectoines concentration was 10mM for aggregation reduction, so samples containing of 10mM of ectoines were selected for further evaluation. Secondary structural and conformational stability increased in presence of ectoines as measured by far-UV circular dichroism and fluorescence spectroscopy, respectively. DSC showed slight increase in Tm of interferon in the presence of ectoines. Hydroxyectoine had superior protein-stabilizing properties than ectoine. In conclusion, this study demonstrates that ectoine and hydroxyectoine are highly effective excipients which can significantly reduce the thermal-induced aggregation of rhIFNα2b at low concentration.


Assuntos
Diamino Aminoácidos/farmacologia , Temperatura Alta/efeitos adversos , Interferon-alfa , Agregados Proteicos/efeitos dos fármacos , Diamino Aminoácidos/química , Linhagem Celular , Estabilidade de Medicamentos , Humanos , Interferon alfa-2 , Interferon-alfa/fisiologia , Agregados Proteicos/fisiologia , Estabilidade Proteica/efeitos dos fármacos , Proteínas Recombinantes
12.
Mediators Inflamm ; 2017: 4532409, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29403161

RESUMO

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.


Assuntos
Células Dendríticas/fisiologia , Glomerulonefrite por IGA/patologia , Interferon-alfa/fisiologia , Plasmócitos/citologia , Adulto , Linfócitos T CD4-Positivos/fisiologia , Diferenciação Celular , Movimento Celular , Feminino , Glomerulonefrite por IGA/imunologia , Humanos , Masculino , Receptor Toll-Like 9/fisiologia
13.
J Biotechnol ; 233: 6-16, 2016 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-27346232

RESUMO

Type I Interferons (IFNs-I) are species-specific glycoproteins which play an important role as primary defence against viral infections and that can also modulate the adaptive immune system. In some autoimmune diseases, interferons (IFNs) are over-produced. IFNs are widely used as biopharmaceuticals for a variety of cancer indications, chronic viral diseases, and for their immuno-modulatory action in patients with multiple sclerosis; therefore, increasing their therapeutic efficiency and decreasing their side effects is of high clinical value. In this sense, it is interesting to find molecules that can modulate the activity of IFNs. In order to achieve that, it was necessary to establish a simple, fast and robust assay to analyze numerous compounds simultaneously. We developed four reporter gene assays (RGAs) to identify IFN activity modulator compounds by using WISH-Mx2/EGFP, HeLa-Mx2/EGFP, A549-Mx2/EGFP, and HEp2-Mx2/EGFP reporter cell lines (RCLs). All of them present a Z' factor higher than 0.7. By using these RGAs, natural and synthetic compounds were analyzed simultaneously. A total of 442 compounds were studied by the Low Throughput Screening (LTS) assay using the four RCLs to discriminate between their inhibitory or enhancing effects on IFN activity. Some of them were characterized and 15 leads were identified. Finally, one promising candidate with enhancing effect on IFN-α/-ß activity and five compounds with inhibitory effect were described.


Assuntos
Descoberta de Drogas/métodos , Genes Reporter/genética , Interferon-alfa/efeitos dos fármacos , Interferon-alfa/fisiologia , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Técnicas Genéticas , Células HeLa , Humanos , Reprodutibilidade dos Testes
14.
Cytokine ; 80: 48-55, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26945996

RESUMO

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.


Assuntos
Hepacivirus/fisiologia , Interferon-alfa/fisiologia , Fator de Transcrição STAT3/antagonistas & inibidores , Transdução de Sinais , Proteínas Supressoras da Sinalização de Citocina/antagonistas & inibidores , Carcinoma Hepatocelular , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Humanos , Interferon alfa-2 , Interferon-alfa/farmacologia , Neoplasias Hepáticas , Proteínas Recombinantes/farmacologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Replicação Viral
15.
J Biol Chem ; 291(12): 6158-68, 2016 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-26817845

RESUMO

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.


Assuntos
Adenosina Desaminase/fisiologia , Imunidade Inata , Edição de RNA , RNA de Cadeia Dupla/metabolismo , Animais , Células Cultivadas , Grânulos Citoplasmáticos/metabolismo , Desaminação , Fator de Iniciação 2 em Eucariotos/metabolismo , Fibroblastos/metabolismo , Tolerância Imunológica , Interferon-alfa/fisiologia , Camundongos Knockout , Fosforilação , Processamento de Proteína Pós-Traducional , RNA de Cadeia Dupla/genética , Proteínas de Ligação a RNA/fisiologia , Transdução de Sinais
16.
PLoS One ; 11(1): e0146325, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26735690

RESUMO

INTRODUCTION: Interferon alpha (IFNα) is routinely used in the clinical practice for adjuvant systemic melanoma therapy. Understanding the molecular mechanism of IFNα effects and prediction of response in the IFNα therapy regime allows initiation and continuation of IFNα treatment for responder and exclusion of non-responder to avoid therapy inefficacy and side-effects. The transporter protein associated with antigen processing-1 (TAP1) is part of the MHC class I peptide-loading complex, and important for antigen presentation in tumor and antigen presenting cells. In the context of personalized medicine, we address this potential biomarker TAP1 as a target of IFNα signalling. RESULTS: We could show that IFNα upregulates TAP1 expression in peripheral blood mononuclear cells (PBMCs) of patients with malignant melanoma receiving adjuvant high-dose immunotherapy. IFNα also induced expression of TAP1 in mouse blood and tumor tissue and suppressed the formation of melanoma metastasis in an in vivo B16 tumor model. Besides its expression, TAP binding affinity and transport activity is induced by IFNα in human monocytic THP1 cells. Furthermore, our data revealed that IFNα clearly activates phosphorylation of STAT1 and STAT3 in THP1 and A375 melanoma cells. Inhibition of Janus kinases abrogates the IFNα-induced TAP1 expression. These results suggest that the JAK/STAT pathway is a crucial mediator for TAP1 expression elicited by IFNα treatment. CONCLUSION: We suppose that silencing of TAP1 expression provides tumor cells with a mechanism to escape cytotoxic T-lymphocyte recognition. The observed benefit of IFNα treatment could be mediated by the shown dual effect of TAP1 upregulation in antigen presenting cells on the one hand, and of TAP1 upregulation in 'silent' metastatic melanoma cells on the other hand. In conclusion, this work contributes to a better understanding of the mode of action of IFNα which is essential to identify markers to predict, assess and monitor therapeutic response of IFNα treatment in the future.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Interferon-alfa/fisiologia , Melanoma/imunologia , Neoplasias Cutâneas/imunologia , Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Adulto , Idoso , Animais , Apresentação de Antígeno , Humanos , Imunoterapia , Interferon-alfa/farmacologia , Janus Quinases , Leucócitos Mononucleares , Masculino , Melanoma/tratamento farmacológico , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Transplante de Neoplasias , Fosforilação , Processamento de Proteína Pós-Traducional , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Neoplasias Cutâneas/tratamento farmacológico , Regulação para Cima
17.
Brain Behav Immun ; 58: 31-39, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26697999

RESUMO

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.


Assuntos
Encéfalo/fisiologia , Interferon-alfa/fisiologia , Adulto , Encéfalo/efeitos dos fármacos , Mapeamento Encefálico , Feminino , Humanos , Interferon-alfa/administração & dosagem , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade
18.
J Biol Chem ; 290(45): 27345-27359, 2015 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-26370074

RESUMO

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.


Assuntos
Vírus da Leucemia Murina de Moloney/patogenicidade , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/química , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/fisiologia , Animais , Sítios de Ligação , Linhagem Celular , Gangliosídeos/química , Gangliosídeos/metabolismo , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Interferon-alfa/fisiologia , Leucemia Experimental/fisiopatologia , Leucemia Experimental/virologia , Linfócitos/fisiologia , Linfócitos/virologia , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Moleculares , Vírus da Leucemia Murina de Moloney/genética , Vírus da Leucemia Murina de Moloney/fisiologia , Ácido N-Acetilneuramínico/química , Receptores Virais/química , Receptores Virais/fisiologia , Infecções por Retroviridae/fisiopatologia , Infecções por Retroviridae/virologia , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/genética , Infecções Tumorais por Vírus/fisiopatologia , Infecções Tumorais por Vírus/virologia
19.
Mol Med Rep ; 12(5): 7176-80, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26397446

RESUMO

Sterile α motif and HD domain-containing protein 1 (SAMHD1) is a novel intrinsic restriction factor that inhibits the replication of certain retroviruses and DNA viruses through its deoxynucleoside triphosphate triphosphohydrolase activity. A previous study by our group showed that SAMHD1 restrained hepatitis B virus replication and interferon (IFN)­α induced SAMHD1 expression in liver cells. However the mechanisms of SAMHD1 upregulation by IFN­α in liver cells have remained elusive. The present study demonstrated that IFN­α treatment increased SAMHD1 mRNA levels in SMMC­7721 cells in a time­dependent manner. Knockdown of STAT1 inhibited the induction of SAMHD1 expression by IFN­α in SMMC­7721 cells. STAT2 silencing also suppressed the induction of SAMHD1 expression by IFN­α in SMMC­7721 cells. Furthermore, the induction of SAMHD1 expression in SMMC­7721 cells by IFN­α was found to be dependent on IFN­regulatory factor 9 (IRF9). In conclusion, these results suggested that the interferon­stimulated gene factor 3 complex, which consists of STAT1, STAT2 and IRF9, is required for the induction of SAMHD1 expression by IFN-α in SMMC-7721 cells.


Assuntos
Fator Gênico 3 Estimulado por Interferon/fisiologia , Interferon-alfa/fisiologia , Proteínas Monoméricas de Ligação ao GTP/genética , Linhagem Celular Tumoral , Expressão Gênica , Humanos , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Proteína 1 com Domínio SAM e Domínio HD , Ativação Transcricional
20.
Biologicals ; 43(4): 225-31, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26073788

RESUMO

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.


Assuntos
Interferon-alfa/fisiologia , Proteínas Recombinantes de Fusão/biossíntese , Timosina/análogos & derivados , Animais , Técnicas In Vitro , Interferon-alfa/genética , Proteínas Recombinantes de Fusão/genética , Suínos , Trombina/metabolismo , Timalfasina , Timosina/genética , Timosina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...