Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
1.
mBio ; 12(6): e0275621, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34724828

RESUMEN

Outbreaks of emerging viral pathogens like severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are a major medical challenge. There is a pressing need for antivirals that can be rapidly deployed to curb infection and dissemination. We determined the efficacy of interferon lambda-1 (IFN-λ) as a broad-spectrum antiviral agent to inhibit SARS-CoV-2 infection and reduce pathology in a mouse model of disease. IFN-λ significantly limited SARS-CoV-2 production in primary human bronchial epithelial cells in culture. Pretreatment of human lung cells with IFN-λ completely blocked infectious virus production, and treatment with IFN-λ at the time of infection inhibited virus production more than 10-fold. To interrogate the protective effects of IFN-λ in response to SARS-CoV-2 infection, transgenic mice expressing the human angiotensin-converting enzyme 2 (ACE-2) were tested. One dose of IFN-λ administered intranasally was found to reduce animal morbidity and mortality. Our study with SARS-CoV-2 also revealed a sex differential in disease outcome. Male mice had higher mortality, reflecting the more severe symptoms and mortality found in male patients infected with SARS-CoV-2. The results indicate that IFN-λ potentially can treat early stages of SARS-CoV-2 infection and decrease pathology, and this murine model can be used to investigate the sex differential documented in COVID-19. IMPORTANCE The COVID-19 pandemic has claimed millions of lives worldwide. In this report, we used a preclinical mouse model to investigate the prophylactic and therapeutic value of intranasal IFN-λ for this acute respiratory disease. Specific vaccines have been responsible for curbing the transmission of SARS-CoV-2 in developed nations. However, vaccines require time to generate and keep pace with antigenic variants. There is a need for broad-spectrum prophylactic and therapeutic agents to combat new emerging viral pathogens. Our mouse model suggests IFN-λ has clinical utility, and it reflects the well-documented finding that male COVID-19 patients manifest more severe symptoms and mortality. Understanding this sex bias is critical for considering therapeutic approaches to COVID-19.


Asunto(s)
Antivirales/uso terapéutico , COVID-19/inmunología , COVID-19/terapia , Células Epiteliales/efectos de los fármacos , Interferones/inmunología , Interferones/farmacología , SARS-CoV-2/inmunología , Administración Intranasal , Enzima Convertidora de Angiotensina 2/genética , Animales , Antivirales/farmacología , Bronquios/citología , Modelos Animales de Enfermedad , Células Epiteliales/inmunología , Células Epiteliales/virología , Femenino , Células HEK293 , Humanos , Interferones/clasificación , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/virología , Masculino , Ratones , Ratones Transgénicos , Factores de Riesgo , SARS-CoV-2/efectos de los fármacos , Factores Sexuales
2.
Viruses ; 13(6)2021 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-34207487

RESUMEN

Hepatitis B virus (HBV) chronically infects over 250 million people worldwide and causes nearly 1 million deaths per year due to cirrhosis and liver cancer. Approved treatments for chronic infection include injectable type-I interferons and nucleos(t)ide reverse transcriptase inhibitors. A small minority of patients achieve seroclearance after treatment with type-I interferons, defined as sustained absence of detectable HBV DNA and surface antigen (HBsAg) antigenemia. However, type-I interferons cause significant side effects, are costly, must be administered for months, and most patients have viral rebound or non-response. Nucleos(t)ide reverse transcriptase inhibitors reduce HBV viral load and improve liver-related outcomes, but do not lower HBsAg levels or impart seroclearance. Thus, new therapeutics are urgently needed. Lambda interferons (IFNLs) have been tested as an alternative strategy to stimulate host antiviral pathways to treat HBV infection. IFNLs comprise an evolutionarily conserved innate immune pathway and have cell-type specific activity on hepatocytes, other epithelial cells found at mucosal surfaces, and some immune cells due to restricted cellular expression of the IFNL receptor. This article will review work that examined expression of IFNLs during acute and chronic HBV infection, the impact of IFNLs on HBV replication in vitro and in vivo, the association of polymorphisms in IFNL genes with clinical outcomes, and the therapeutic evaluation of IFNLs for the treatment of chronic HBV infection.


Asunto(s)
Antivirales/uso terapéutico , Virus de la Hepatitis B/inmunología , Hepatitis B Crónica/inmunología , Hepatitis B/tratamiento farmacológico , Hepatitis B/genética , Interferones/genética , Interferones/uso terapéutico , Antivirales/inmunología , Hepatitis B/inmunología , Hepatitis B Crónica/tratamiento farmacológico , Hepatitis B Crónica/genética , Hepatocitos/efectos de los fármacos , Hepatocitos/virología , Humanos , Inmunidad Innata/genética , Interferones/clasificación , Hígado/virología
3.
Vet Microbiol ; 259: 109150, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34144506

RESUMEN

Interferon (IFN)-mediated antiviral responses are central to host defense against viral infection. Porcine viral infection has emerged as a serious hazard for the pig industry. The construction of an engineered Saccharomyces cerevisiae strain that efficiently produces porcine IFN has demonstrated several advantages. It can be easily fed to pigs, which helps in reducing antibiotic residues in pork and improve meat quality. In this study, the stable expression of several porcine IFN molecules (pIFN-α1, pIFN-ß, pIFN-λ1, pIFN-λ1-ß, pIFN-λ1-ß-α1) were determined using an engineered S. cerevisiae system. With the YeastFab assembly method, the complete transcriptional units containing promoter (GPD), secretory peptide (α-mating factor), target gene (IFN) and terminator (ADH1) were successfully constructed using the characteristics of type II restriction endonuclease, and then integrated into the chromosomes Ⅳ and XVI of ST1814 yeast host strain, respectively. The expression kinetics of recombinant pIFNs were further analyzed. Synergism in the expression level of IFN receptor, antiviral protein, and viral loading was observed in viral-cell infection model treated with different porcine IFN subtypes. The porcine reproductive and respiratory syndrome viral load and antibody titer in serum decreased significantly after oral administration of IFN expression yeast fermentation broth. These findings indicate the potential efficacy of multi-valent pIFNs expressing S. cerevisiae as a potent feed material to prevent viral infections of pigs.


Asunto(s)
Expresión Génica , Interferones/administración & dosificación , Interferones/inmunología , Saccharomyces cerevisiae/genética , Virosis/prevención & control , Virosis/veterinaria , Animales , Antivirales/administración & dosificación , Antivirales/inmunología , Línea Celular , Medios de Cultivo/análisis , Fermentación , Interferones/clasificación , Interferones/genética , Síndrome Respiratorio y de la Reproducción Porcina/prevención & control , Virus del Síndrome Respiratorio y Reproductivo Porcino/inmunología , Saccharomyces cerevisiae/crecimiento & desarrollo , Saccharomyces cerevisiae/inmunología , Porcinos/inmunología , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/prevención & control , Enfermedades de los Porcinos/virología , Virosis/inmunología , Replicación Viral
4.
Cell Mol Life Sci ; 78(4): 1423-1444, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33084946

RESUMEN

Antiviral responses of interferons (IFNs) are crucial in the host immune response, playing a relevant role in controlling viralw infections. Three types of IFNs, type I (IFN-α, IFN-ß), II (IFN-γ) and III (IFN-λ), are classified according to their receptor usage, mode of induction, biological activity and amino acid sequence. Here, we provide a comprehensive review of type I IFN responses and different mechanisms that viruses employ to circumvent this response. In the first part, we will give an overview of the different induction and signaling cascades induced in the cell by IFN-I after virus encounter. Next, highlights of some of the mechanisms used by viruses to counteract the IFN induction will be described. And finally, we will address different mechanism used by viruses to interference with the IFN signaling cascade and the blockade of IFN induced antiviral activities.


Asunto(s)
Interacciones Huésped-Patógeno/genética , Inmunidad Innata/genética , Interferones/genética , Virosis/genética , Humanos , Evasión Inmune/genética , Interferón-alfa/genética , Interferón-alfa/inmunología , Interferones/clasificación , Interferones/inmunología , Transducción de Señal , Virosis/inmunología
5.
Vet Immunol Immunopathol ; 230: 110145, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33160262

RESUMEN

Interferon lambda (IFN-λ) plays an important role in inducing an antiviral state in mucosal surfaces and has been used as an effective biotherapeutic against several viral diseases. Here we performed a proof of concept study on the activity of a biologically active recombinant bovine IFN-λ (rIFN-λ) produced in eukaryotic cells against Bovine Viral Diarrhea Virus (BVDV) in cattle. We first confirmed the lack of toxicity of different concentrations of rIFN-λ in bovine peripheral blood cells and the safety of its subcutaneous application in calves in doses up to 12 IU/kg. The antiviral activity of the rIFN-λ against BVDV was assessed in calves that were inoculated with 6 IU/kg of rIFN-λ (n = 4) or mock-treated (n = 2) two days before and after challenge with a BVDV type-2 non-cytopathic strain. Mock-treated animals developed respiratory disease, shedded the virus from 4 to 7 days post-infection (dpi) and had viremia between 4 and 14 dpi. Conversely, calves treated with rIFN-λ did not develop clinical symptoms. The virus was not found in nasal secretions or sera. Only one animal had a positive viral RNA detection in serum at 7 dpi. All infected animals treated with rIFN-λ increased systemic type-I IFNs levels at 4 dpi. The antiviral treatment induced an earlier onset of the anti-BVDV neutralizing antibodies. Altogether, these results constitute the proof-of-principle of bovine IFN-λ as an antiviral biotherapeutic to protect cattle against the clinical disease caused by BVDV.


Asunto(s)
Diarrea Mucosa Bovina Viral/inmunología , Diarrea Mucosa Bovina Viral/prevención & control , Enfermedades de los Bovinos/prevención & control , Virus de la Diarrea Viral Bovina/inmunología , Diarrea/veterinaria , Inmunización Pasiva , Interferones/administración & dosificación , Factores de Edad , Animales , Bovinos , Enfermedades de los Bovinos/inmunología , Enfermedades de los Bovinos/virología , Diarrea/prevención & control , Diarrea/virología , Virus de la Diarrea Viral Bovina Tipo 1/inmunología , Virus de la Diarrea Viral Bovina Tipo 2/inmunología , Femenino , Inmunización Pasiva/veterinaria , Interferones/clasificación , Interferones/genética , Interferones/inmunología , Prueba de Estudio Conceptual , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/inmunología , Esparcimiento de Virus
6.
Sci Rep ; 10(1): 20393, 2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-33230288

RESUMEN

Brain embryonic periventricular endothelial cells (PVEC) crosstalk with neural progenitor cells (NPC) promoting mutual proliferation, formation of tubular-like structures in the former and maintenance of stemness in the latter. To better characterize this interaction, we conducted a comparative transcriptome analysis of mouse PVEC vs. adult brain endothelial cells (ABEC) in mono-culture or NPC co-culture. We identified > 6000 differentially expressed genes (DEG), regardless of culture condition. PVEC exhibited a 30-fold greater response to NPC than ABEC (411 vs. 13 DEG). Gene Ontology (GO) analysis of DEG that were higher or lower in PVEC vs. ABEC identified "Nervous system development" and "Response to Stress" as the top significantly different biological process, respectively. Enrichment in canonical pathways included HIF1A, FGF/stemness, WNT signaling, interferon signaling and complement. Solute carriers (SLC) and ABC transporters represented an important subset of DEG, underscoring PVEC's implication in blood-brain barrier formation and maintenance of nutrient-rich/non-toxic environment. Our work characterizes the gene signature of PVEC and their important partnership with NPC, underpinning their unique role in maintaining a healthy neurovascular niche, and in supporting brain development. This information may pave the way for additional studies to explore their therapeutic potential in neuro-degenerative diseases, such as Alzheimer's and Parkinson's disease.


Asunto(s)
Envejecimiento/genética , Proteínas del Sistema Complemento/genética , Células Endoteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Interferones/genética , Células Madre Embrionarias de Ratones/metabolismo , Células-Madre Neurales/metabolismo , Envejecimiento/metabolismo , Animales , Encéfalo/irrigación sanguínea , Encéfalo/citología , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Proteínas Portadoras/clasificación , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , Técnicas de Cocultivo , Proteínas del Sistema Complemento/clasificación , Proteínas del Sistema Complemento/metabolismo , Embrión de Mamíferos , Células Endoteliales/citología , Femenino , Factor 1 de Crecimiento de Fibroblastos/genética , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Ontología de Genes , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Interferones/clasificación , Interferones/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Anotación de Secuencia Molecular , Células Madre Embrionarias de Ratones/citología , Células-Madre Neurales/citología , Cultivo Primario de Células , Proteínas Wnt/clasificación , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
7.
Fish Shellfish Immunol ; 98: 564-573, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32001354

RESUMEN

A relatively large repertoire of type I interferon (IFN) genes is apparent in rainbow trout/Atlantic salmon, that includes six different IFN subgroups (IFNa-IFNf) belonging to the three known type I IFN groups (1-3) in bony fish. Whether this is true for other salmonids, and how the various type I subgroups evolved in teleost fish was studied using the extensive genomic resources available for fish. This confirmed that salmonids, at least the Salmoninae, indeed have a complex (in terms of IFN subgroups present) and large (number of genes) IFN repertoire relative to other teleost fish. This is in part a consequence of the salmonid 4 R WGD that duplicated the growth hormone (GH) locus in which type I IFNs are generally located. Divergence of the IFN genes at the two GH loci was apparent but was not seen in common carp, a species that also underwent an independent 4 R WGD. However, expansion of IFN gene number can be found at the CD79b locus of some perciform fish (both freshwater and marine), with expansion of the IFNd gene repertoire. Curiously the primordial gene order of GH-IFNc-IFNb-IFNa-IFNe is largely retained in many teleost lineages and likely reflects the tandem duplications that are taking place to increase IFN gene number. With respect to the evolution of the IFN subgroups, a complex acquisition and/or loss has occurred in different teleost lineages, with complete loss of IFN genes at the GH or CD79b locus in some species, and reduction to a single IFN subgroup in others. It becomes clear that there are many variations to be discovered regarding the mechanisms by which fish elicit protective (antiviral) immune responses.


Asunto(s)
Evolución Biológica , Genoma , Interferones/genética , Salmonidae/genética , Animales , Duplicación de Gen , Interferones/clasificación , Salmonidae/inmunología
8.
Viral Immunol ; 33(1): 3-11, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31545139

RESUMEN

Immune regulation at the maternal-fetal interface is complex due to conflicting immunological objectives: protection of the fetus from maternal pathogens and prevention of immune-mediated rejection of the semiallogeneic fetus and placenta. Interferon (IFN) signaling plays an important role in restricting congenital infections as well as in the physiology of healthy pregnancies. In this review, we discuss the antiviral and pathogenic effects of type I IFN (IFN-α, IFN-ß), type II IFN (IFN-γ), and type III IFN (IFN-λ) during pregnancy, with an emphasis on mouse and non-human primate models of congenital Zika virus infection. In the context of these animal model systems, we examine the role of IFN signaling during healthy pregnancy. Finally, we review mechanisms by which dysregulated type I IFN responses contribute to poor pregnancy outcomes in humans with autoimmune disease, including interferonopathies and systemic lupus erythematosus.


Asunto(s)
Inmunidad Innata , Interferones/inmunología , Resultado del Embarazo , Transducción de Señal/inmunología , Animales , Antivirales/inmunología , Enfermedades Autoinmunes/complicaciones , Modelos Animales de Enfermedad , Femenino , Humanos , Interferones/clasificación , Ratones , Placenta/inmunología , Placenta/virología , Embarazo , Complicaciones Infecciosas del Embarazo/virología , Primates , Infección por el Virus Zika/inmunología
9.
J Virol ; 94(5)2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-31776283

RESUMEN

Interferon lambda 4 (IFN-λ4) is a recently identified enigmatic member of the interferon (IFN) lambda family. Genetic data suggest that the IFNL4 gene acts in a proviral and anti-inflammatory manner in patients. However, the protein is indistinguishable in vitro from the other members of the interferon lambda family. We have investigated the gene regulation of IFNL4 in detail and found that it differs radically from that of canonical antiviral interferons. Being induced by viral infection is a defining characteristic of interferons, but viral infection or overexpression of members of the interferon regulatory factor (IRF) family of transcription factors only leads to a minute induction of IFNL4 This behavior is evolutionarily conserved and can be reversed by inserting a functional IRF3 binding site into the IFNL4 promoter. Thus, the regulation of the IFNL4 gene is radically different and might explain some of the atypical phenotypes associated with the IFNL4 gene in humans.IMPORTANCE Recent genetic evidence has highlighted how the IFNL4 gene acts in a counterintuitive manner, as patients with a nonfunctional IFNL4 gene exhibit increased clearance of hepatitis C virus (HCV) but also increased liver inflammation. This suggests that the IFNL4 gene acts in a proviral and anti-inflammatory manner. These surprising but quite clear genetic data have prompted an extensive examination of the basic characteristics of the IFNL4 gene and its gene product, interferon lambda 4 (IFN-λ4). We have investigated the expression of the IFNL4 gene and found it to be poorly induced by viral infections. A thorough investigation of the IFNL4 promoter revealed a highly conserved and functional promoter, but also one that lacks the defining characteristic of interferons (IFNs), i.e., the ability to be effectively induced by viral infections. We suggest that the unique function of the IFNL4 gene is related to its noncanonical transcriptional regulation.


Asunto(s)
Evolución Molecular , Interferones/genética , Interferones/metabolismo , Células A549 , Animales , Antivirales/farmacología , Secuencia de Bases , Regulación de la Expresión Génica , Células HEK293 , Células Hep G2 , Hepacivirus/fisiología , Hepatitis C/metabolismo , Humanos , Inflamación , Interferones/clasificación , Interleucinas/clasificación , Interleucinas/genética , Interleucinas/farmacología , Hígado/patología , ARN Mensajero/metabolismo , Alineación de Secuencia , Células THP-1
10.
Fish Shellfish Immunol ; 95: 163-170, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31626921

RESUMEN

The present study helps clarify when the fish type I IFN groups/subgroups evolved, by examination of the IFN genes present in the Holostean spotted gar, Lepisosteus oculatus, in relation to the IFN genes present in the Chondrostea (sturgeon). It confirms that all three IFN groups (I-III), and group II subgroups, existed prior to the appearance of teleost fish. Preliminary expression analysis in a gar cell line (GARL) suggests these IFN genes will have a role in antiviral defence in Holostean fish, in that they are induced by poly(I:C). A refined model of IFN evolution within the actinopterygian fish is proposed.


Asunto(s)
Evolución Molecular , Peces/genética , Peces/inmunología , Interferones/genética , Rajidae/genética , Rajidae/inmunología , Animales , Línea Celular , Interferones/clasificación , Poli I-C/inmunología , Poli I-C/farmacología
11.
Cell ; 163(7): 1808-1808.e1, 2015 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-26687364

RESUMEN

Interferons (IFNs) are crucial cytokines of antimicrobial, antitumor, and immunomodulatory activity. The three types of IFN (I, II, and III) are classified by their receptor specificity and sequence homology. IFNs are produced and secreted by cells in response to specific stimuli. Here, we review the subsequent IFN signaling events occurring through unique receptors leading to regulation of gene expression for modulation of innate and adaptive immunity. To view this SnapShot, open or download the PDF.


Asunto(s)
Interferones/metabolismo , Transducción de Señal , Animales , Humanos , Interferones/clasificación , Receptores de Interferón
13.
Vestn Ross Akad Med Nauk ; (3-4): 31-5, 2014.
Artículo en Ruso | MEDLINE | ID: mdl-25306593

RESUMEN

BACKGROUND: To study impact of interferon (IFN) alpha, beta and gamma on the Herpes simplex virus type 1 (HSV-1) infected endothelial cells functional activity related with participation in the inflammation development. MATERIALS AND METHODS: In the work endothelial cells isolated from umbilical vein were used. Intact and infected cultures were treated by interferon and in the dynamics of cultivation tested mediators in the cultural medium. RESULTS: All investigated interferons activated the production of IL-6. IFN alpha, beta activated the production of IL-8, while IFN gamma inhibited her. IFN alpha and gamma increased synthesis of nitrogen oxides and reduced the synthesis of endothelin-1, while IFN beta activated the production of endothelin-1. CONCLUSION: Infection of endothelial cells isolated from umbilical vein with HSV-1 does not alter the ability of interferon in modulating of proinflammatory cytokines, nitric oxide and endothelin-1 synthesis. It is obvious in the body modulation manifestations of innate immunity under the influence of exogenous interferon is implemented both intact and infected with HSV-1-vascular endothelium and nature modulation is determined by the type of IFN.


Asunto(s)
Endotelio Vascular/inmunología , Herpesvirus Humano 1/patogenicidad , Inmunidad Innata/efectos de los fármacos , Interferones , Células Cultivadas , Endotelina-1/metabolismo , Humanos , Mediadores de Inflamación/fisiología , Interferones/clasificación , Interferones/inmunología , Interferones/farmacología , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Óxido Nítrico/biosíntesis , Venas Umbilicales
14.
J Clin Invest ; 124(8): 3352-63, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24983321

RESUMEN

BACKGROUND. Hepatitis C virus (HCV) infects approximately 170 million people worldwide and may lead to cirrhosis and hepatocellular carcinoma in chronically infected individuals. Treatment is rapidly evolving from IFN-α-based therapies to IFN-α-free regimens that consist of directly acting antiviral agents (DAAs), which demonstrate improved efficacy and tolerability in clinical trials. Virologic relapse after DAA therapy is a common cause of treatment failure; however, it is not clear why relapse occurs or whether certain individuals are more prone to recurrent viremia. METHODS. We conducted a clinical trial using the DAA sofosbuvir plus ribavirin (SOF/RBV) and performed detailed mRNA expression analysis in liver and peripheral blood from patients who achieved either a sustained virologic response (SVR) or relapsed. RESULTS. On-treatment viral clearance was accompanied by rapid downregulation of IFN-stimulated genes (ISGs) in liver and blood, regardless of treatment outcome. Analysis of paired pretreatment and end of treatment (EOT) liver biopsies from SVR patients showed that viral clearance was accompanied by decreased expression of type II and III IFNs, but unexpectedly increased expression of the type I IFN IFNA2. mRNA expression of ISGs was higher in EOT liver biopsies of patients who achieved SVR than in patients who later relapsed. CONCLUSION. These results suggest that restoration of type I intrahepatic IFN signaling by EOT may facilitate HCV eradication and prevention of relapse upon withdrawal of SOF/RBV. TRIAL REGISTRATION. ClinicalTrials.gov NCT01441180.


Asunto(s)
Antivirales/administración & dosificación , Hepatitis C Crónica/tratamiento farmacológico , Hepatitis C Crónica/inmunología , Interferones/metabolismo , Hígado/inmunología , Ribavirina/administración & dosificación , Uridina Monofosfato/análogos & derivados , Quimiocina CXCL10/sangre , Quimioterapia Combinada , Endopeptidasas/genética , Expresión Génica/efectos de los fármacos , Hepatitis C Crónica/genética , Humanos , Interferón-alfa/genética , Interferón-alfa/metabolismo , Interferones/clasificación , Interferones/genética , Interleucinas/genética , Hígado/metabolismo , ARN Mensajero/sangre , ARN Mensajero/genética , ARN Mensajero/metabolismo , Recurrencia , Sofosbuvir , Resultado del Tratamiento , Ubiquitina Tiolesterasa , Uridina Monofosfato/administración & dosificación
15.
J Interferon Cytokine Res ; 34(11): 848-57, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24950142

RESUMEN

In this study, we provide the first comprehensive annotation of canine interferon-λ (CaIFN-λ, type III IFN). Phylogenetic analysis based on genomic sequences indicated that CaIFN-λ is located in the same branch with Swine IFN-λ1 (SwIFN-λ), Bat IFN-λ1 (BaIFN-λ), and human IFN-λ1 (HuIFN-λ1). CaIFN-λ was cloned, expressed in Escherichia coli, and purified to further investigate the biological activity in vitro. The recombinant CaIFN-λ (rCaIFN-λ) displayed potent antiviral activity on both homologous and heterologous animal cells in terms of inhibiting the replication of the New Jersey serotype of vesicular stomatitis virus (VSV), canine parvovirus, and influenza virus A/WSN/33 (H1N1), respectively. In addition, we also found that rCaIFN-λ exhibits a significant antiproliferative response against A72 canine tumor cells and MDCK cells in a dose-dependent manner. Furthermore, CaIFN-λ activated the JAK-STAT signaling pathway. To evaluate the expression of CaIFN-λ induced by virus and the expression of IFN-stimulated genes (ISGs) induced by rCaIFN-λ in the MDCK cells, we measured the relative mRNA level of CaIFN-λ and ISGs (ISG15, Mx1, and 2'5'-OAS) by quantitative real-time PCR and found that the mRNA level of CaIFN-λ and the ISGs significantly increased after treating the MDCK cells with viruses and rCaIFN-λ protein, respectively. Finally, to evaluate the binding activity of rCaIFN-λ to its receptor, we expressed the extracellular domain of the canine IFN-λ receptor 1 (CaIFN-λR1-EC) and determined the binding activity via ELISA. Our results demonstrated that rCaIFN-λ bound tightly to recombinant CaIFN-λR1-EC (rCaIFN-λR1-EC).


Asunto(s)
Inhibidores de Crecimiento/metabolismo , Interferones/clasificación , Interferones/inmunología , Interleucinas/clasificación , Interleucinas/inmunología , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Clonación Molecular , Perros , Humanos , Interferones/genética , Interleucinas/genética , Quinasas Janus/metabolismo , Células de Riñón Canino Madin Darby , Datos de Secuencia Molecular , Filogenia , Unión Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Virosis/inmunología , Replicación Viral
16.
Postepy Hig Med Dosw (Online) ; 68: 428-40, 2014 May 06.
Artículo en Polaco | MEDLINE | ID: mdl-24864095

RESUMEN

Interferons are a family of proteins that are released by a variety of cells in response to infections caused by viruses. Currently, we distinguish three types of interferons. They are classified based on the nucleotide sequence, interaction with specific receptors, chromosomal location, structure and physicochemical properties. The following interferons are classified as type I: α, ß, ω, κ, ε, ζ, τ, δ, ν. They are recognized and bound by a receptor formed by two peptides, IFN-αR1 and IFN-αR2. Representative of type II interferons is interferon-γ. It binds to a receptor composed of chains IFNGR-1 and IFNGR-2. The recently classified type III interferons comprise IFN-λ1, IFN-λ2, and IFN-λ3. They act on receptors formed by λR1 IFN-and IL-10R2 subunits. A high level of antiviral protection is achieved by IFN-α, IFN-ß and IFN-λ. Antiviral activity of interferons is based on the induction and regulation of innate and acquired immune mechanisms. By binding to transmembrane receptors, IFN interacts with target cells mainly by activating the JAK/STAT, but also other signaling pathways. This leads to induction and activation of many antiviral agents, such as protein kinase RNA-activated (PKR), ribonuclease 2-5A pathway, and Mx proteins, as well as numerous apoptotic pathways. As a result of the protective effect of interferons, the virus binding to cells and viral particles penetration into cells is stopped, and the release of the nucleocapsid from an envelope is suppressed. Disruption of transcription and translation processes of the structural proteins prevents the formation of virions or budding of viruses, and as a result degradation of the viral mRNA; the started processes inhibit the chain synthesis of viral proteins and therefore further stimulate the immune system cells.


Asunto(s)
Interferones/química , Interferones/inmunología , Activación Metabólica/inmunología , Animales , Antivirales/farmacología , Apoptosis/fisiología , Humanos , Interferón-alfa/química , Interferón-alfa/inmunología , Interferón gamma/química , Interferón gamma/inmunología , Interferones/clasificación , Sustancias Protectoras/metabolismo , ARN Viral/metabolismo , Transducción de Señal/fisiología , Proteínas Virales/química , Proteínas Virales/metabolismo
17.
Gynecol Endocrinol ; 30(1): 1-6, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24188446

RESUMEN

The interferons (IFNs) form part of the large family of glycoproteins known as cytokines. They are secreted by host cells as a line of defence against pathogens and certain tumours. IFNs affect cell proliferation and differentiation and also play a very important role in the functioning of the immune system. Miscarriage in both humans has been associated with higher levels of IFN, particularly IFN-γ. However, this cytokine is evidently vital in successful murine pregnancies since it is involved in maintaining the decidual layer in addition to remodelling of the vasculature in the uterus. The effects of IFN on human pregnancies are more difficult to study. Hence, there is still a lot more to be discovered in the hope of reaching a definite conclusion regarding the impact of IFN.


Asunto(s)
Interferones/fisiología , Primer Trimestre del Embarazo/inmunología , Aborto Espontáneo/genética , Aborto Espontáneo/inmunología , Animales , Implantación del Embrión/genética , Implantación del Embrión/inmunología , Femenino , Humanos , Interferones/química , Interferones/clasificación , Polimorfismo Genético , Embarazo/genética , Embarazo/inmunología , Embarazo/metabolismo , Receptores de Interferón/clasificación , Receptores de Interferón/fisiología , Transducción de Señal/genética , Transducción de Señal/inmunología
19.
Gerontology ; 59(5): 421-6, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23615484

RESUMEN

Initial secretion of interferons by innate immune cells such as dendritic cells is crucial for protection against infections as well as for alerting and activating the downstream immune responses. The secretion of innate interferons, both type I and type III, by dendritic cells is severely impaired in aged subjects. This review focuses on the mechanisms responsible for the reduced interferon secretion by dendritic cells and the role this plays in the increased susceptibility of the elderly to infections, particularly of the respiratory mucosa.


Asunto(s)
Envejecimiento/inmunología , Células Dendríticas/inmunología , Interferones/metabolismo , Animales , Células Dendríticas/clasificación , Humanos , Inmunidad Innata , Interferón Tipo I/clasificación , Interferón Tipo I/metabolismo , Interferones/clasificación , Interleucinas/clasificación , Interleucinas/metabolismo , Modelos Inmunológicos
20.
Mol Immunol ; 52(2): 61-70, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22578383

RESUMEN

MicroRNAs (miRNAs) are involved in the regulation of immunity via targeting of mRNA encoding immune response elements. In this report, alterations in the expression of microRNAs as autoantibody levels increase was investigated. The (NZB×NZW)F1 or B/W mouse model of systemic lupus erythematosus (SLE) naturally has increased autoantibodies with aging. IFNα (type I IFN) accelerates B/W disease, however, the effects of a related IFN, IFNλ, which is a type III IFN, is largely unknown. The purpose of the study was to investigate the relationship between IFN-accelerated disease, microRNAs, immunoregulatory B cell subsets and autoantibody production in the autoimmune-prone environment in vivo. B/W mice received osmotic pumps to chronically deliver IFNα and IFNλ for up to 16 weeks. Urine protein level was monitored weekly by urine strips and proteinuria was used as the disease marker. Splenic cells were taken for flow analysis of B cell subsets and levels of microRNAs determined. Plasma were analyzed for autoantibodies and microRNA levels. As a result of treatment, IFNα accelerated proteinuria in a dose dependent manner, while IFNλ single treatment did not show a significant effect, but greatly enhanced low dose IFNα effects in the combination treatment. Both the splenic cellular and plasma miR-15a were elevated in diseased compared to pre-diseased mice as well as autoantibody levels. Autoantibodies and miR-15a levels were significantly correlated. The immunosuppressive B subpopulation, B-10, was reduced following IFNα treatment. In addition in diseased mice, B-10 versus B-2 ratios were reduced in IFN-treated B/W compared to the control PBS treated group. In all B/W the miR-15a was highly expressed in the B-10 subset and this increased with disease development, suggesting that miR-15a has a specific negative effect on the B-10 subpopulation. In conclusion, our data support the involvement of elevated miR-15a in autoimmune disease development in B/W mice and suggest that decreasing this microRNA might be beneficial in B/W mice.


Asunto(s)
Autoanticuerpos/biosíntesis , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , MicroARNs/genética , MicroARNs/inmunología , Animales , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/patología , Secuencia de Bases , Cartilla de ADN/genética , Modelos Animales de Enfermedad , Femenino , Interferón-alfa/administración & dosificación , Interferones/administración & dosificación , Interferones/clasificación , Lupus Eritematoso Sistémico/etiología , Lupus Eritematoso Sistémico/patología , Ratones , Ratones Endogámicos NZB , MicroARNs/sangre , MicroARNs/metabolismo , Proteínas Recombinantes/administración & dosificación , Bazo/inmunología , Bazo/patología , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...