Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 43(18): 3876-3894, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39112803

RESUMEN

The OAS-RNase L pathway is one of the oldest innate RNA sensing pathways that leads to interferon (IFN) signaling and cell death. OAS recognizes viral RNA and then activates RNase L, which subsequently cleaves both cellular and viral RNA, creating "processed RNA" as an endogenous ligand that further triggers RIG-I-like receptor signaling. However, the IFN response and antiviral activity of the OAS-RNase L pathway are weak compared to other RNA-sensing pathways. Here, we discover that the SKIV2L RNA exosome limits the antiviral capacity of the OAS-RNase L pathway. SKIV2L-deficient cells exhibit remarkably increased interferon responses to RNase L-processed RNA, resulting in heightened antiviral activity. The helicase activity of SKIV2L is indispensable for this function, acting downstream of RNase L. SKIV2L depletion increases the antiviral capacity of OAS-RNase L against RNA virus infection. Furthermore, SKIV2L loss exacerbates autoinflammation caused by human OAS1 gain-of-function mutations. Taken together, our results identify SKIV2L as a critical barrier to OAS-RNase L-mediated antiviral immunity that could be therapeutically targeted to enhance the activity of a basic antiviral pathway.


Asunto(s)
2',5'-Oligoadenilato Sintetasa , Endorribonucleasas , 2',5'-Oligoadenilato Sintetasa/metabolismo , 2',5'-Oligoadenilato Sintetasa/genética , Humanos , Endorribonucleasas/metabolismo , Endorribonucleasas/genética , ARN Helicasas/metabolismo , ARN Helicasas/genética , Animales , Inmunidad Innata , Transducción de Señal , Ratones , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/genética , ARN Viral/metabolismo , ARN Viral/genética , ARN Viral/inmunología , Infecciones por Virus ARN/inmunología , Infecciones por Virus ARN/metabolismo , Células HEK293
2.
J Virol ; 98(4): e0015924, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38499512

RESUMEN

Equine herpesvirus type 8 (EHV-8) causes abortion and respiratory disease in horses and donkeys, leading to serious economic losses in the global equine industry. Currently, there is no effective vaccine or drug against EHV-8 infection, underscoring the need for a novel antiviral drug to prevent EHV-8-induced latent infection and decrease the pathogenicity of this virus. The present study demonstrated that hyperoside can exert antiviral effects against EHV-8 infection in RK-13 (rabbit kidney cells), MDBK (Madin-Darby bovine kidney), and NBL-6 cells (E. Derm cells). Mechanistic investigations revealed that hyperoside induces heme oxygenase-1 expression by activating the c-Jun N-terminal kinase/nuclear factor erythroid-2-related factor 2/Kelch-like ECH-associated protein 1 axis, alleviating oxidative stress and triggering a downstream antiviral interferon response. Accordingly, hyperoside inhibits EHV-8 infection. Meanwhile, hyperoside can also mitigate EHV-8-induced injury in the lungs of infected mice. These results indicate that hyperoside may serve as a novel antiviral agent against EHV-8 infection.IMPORTANCEHyperoside has been reported to suppress viral infections, including herpesvirus, hepatitis B virus, infectious bronchitis virus, and severe acute respiratory syndrome coronavirus 2 infection. However, its mechanism of action against equine herpesvirus type 8 (EHV-8) is currently unknown. Here, we demonstrated that hyperoside significantly inhibits EHV-8 adsorption and internalization in susceptible cells. This process induces HO-1 expression via c-Jun N-terminal kinase/nuclear factor erythroid-2-related factor 2/Kelch-like ECH-associated protein 1 axis activation, alleviating oxidative stress and triggering an antiviral interferon response. These findings indicate that hyperoside could be very effective as a drug against EHV-8.


Asunto(s)
Antivirales , Infecciones por Herpesviridae , Herpesvirus Équido 1 , Sistema de Señalización de MAP Quinasas , Quercetina , Animales , Bovinos , Ratones , Conejos , Antivirales/farmacología , Caballos , Interferones/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/efectos de los fármacos , Quercetina/análogos & derivados , Quercetina/farmacología , Línea Celular
3.
J Virol ; 97(6): e0040023, 2023 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-37289084

RESUMEN

Induction of type I interferon (IFN) gene expression is among the first lines of cellular defense a virus encounters during primary infection. We previously identified the tegument protein M35 of murine cytomegalovirus (MCMV) as an essential antagonist of this antiviral system, showing that M35 interferes with type I IFN induction downstream of pattern-recognition receptor (PRR) activation. Here, we report structural and mechanistic details of M35's function. Determination of M35's crystal structure combined with reverse genetics revealed that homodimerization is a key feature for M35's immunomodulatory activity. In electrophoretic mobility shift assays (EMSAs), purified M35 protein specifically bound to the regulatory DNA element that governs transcription of the first type I IFN gene induced in nonimmune cells, Ifnb1. DNA-binding sites of M35 overlapped with the recognition elements of interferon regulatory factor 3 (IRF3), a key transcription factor activated by PRR signaling. Chromatin immunoprecipitation (ChIP) showed reduced binding of IRF3 to the host Ifnb1 promoter in the presence of M35. We furthermore defined the IRF3-dependent and the type I IFN signaling-responsive genes in murine fibroblasts by RNA sequencing of metabolically labeled transcripts (SLAM-seq) and assessed M35's global effect on gene expression. Stable expression of M35 broadly influenced the transcriptome in untreated cells and specifically downregulated basal expression of IRF3-dependent genes. During MCMV infection, M35 impaired expression of IRF3-responsive genes aside of Ifnb1. Our results suggest that M35-DNA binding directly antagonizes gene induction mediated by IRF3 and impairs the antiviral response more broadly than formerly recognized. IMPORTANCE Replication of the ubiquitous human cytomegalovirus (HCMV) in healthy individuals mostly goes unnoticed but can impair fetal development or cause life-threatening symptoms in immunosuppressed or -deficient patients. Like other herpesviruses, CMV extensively manipulates its hosts and establishes lifelong latent infections. Murine CMV (MCMV) presents an important model system as it allows the study of CMV infection in the host organism. We previously showed that during entry into host cells, MCMV virions release the evolutionary conserved protein M35 protein to immediately dampen the antiviral type I interferon (IFN) response induced by pathogen detection. Here, we show that M35 dimers bind to regulatory DNA elements and interfere with recruitment of interferon regulatory factor 3 (IRF3), a key cellular factor for antiviral gene expression. Thereby, M35 interferes with expression of type I IFNs and other IRF3-dependent genes, reflecting the importance for herpesviruses to avoid IRF3-mediated gene induction.


Asunto(s)
Infecciones por Citomegalovirus , Elementos de Facilitación Genéticos , Factor 3 Regulador del Interferón , Interferón Tipo I , Proteínas de la Matriz Viral , Animales , Humanos , Ratones , Infecciones por Citomegalovirus/genética , ADN/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Interferón beta/genética , Interferón beta/metabolismo , Muromegalovirus/genética , Muromegalovirus/metabolismo , Proteínas de la Matriz Viral/metabolismo
4.
Fish Shellfish Immunol ; 142: 109164, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37839543

RESUMEN

Stimulator of interferon genes (STING) serve as an endoplasmic reticulum (ER) protein and modulates innate immune responses to viral contagion. Most investigations involving teleost STING antiviral immunity have examined DNA viruses. Therefore, fish STING signaling events against RNA viruses require additional exploration. Here, common carp STING (named CcSTING) was cloned and characterized. The bioinformatics analyses of CcSTING showed evolutionary conservations and were most closely related to other cyprinid STINGs. Immunofluorescence staining discovered that the CcSTING was chiefly placed in the cytoplasm, specifically within the ER. CcSTING was ubiquitously generated in all analyzed organs, with especially strong expression in the gills and head kidney. Spring viremia of carp virus (SVCV) stimulation and poly(I:C) infection induced the generation of CcSTING in immune-associated organs, as well as in peripheral blood leukocytes. Additional investigations revealed that CcSTING overexpression strongly suppressed SVCV replication in EPC cells. Mechanistically, CcSTING enhanced IFN-1 and ISGs expression following SVCV infection. CcSTING also substantially increased both IFN and NF-κB promoter luciferase activity via a dosage-dependent fashion. Lastly, CcSTING significantly up-regulated both TBK1 and p65 phosphorylation. Collectively, these findings demonstrated the critical role and underlying mechanism of fish STING in response to RNA virus.


Asunto(s)
Carpas , Enfermedades de los Peces , Virus ARN , Infecciones por Rhabdoviridae , Rhabdoviridae , Animales , Viremia , Carpas/genética , Carpas/metabolismo , Rhabdoviridae/fisiología , Proteínas de Peces
5.
Mol Biol Rep ; 50(4): 3909-3917, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36662450

RESUMEN

BACKGROUND: IRF9 is a transcription factor that mediates the expression of interferon-stimulated genes (ISGs) through the Janus kinase-Signal transducer and activator of transcription (JAK-STAT) pathway. The JAK-STAT pathway is regulated through phosphorylation reactions, in which all components of the pathway are known to be phosphorylated except IRF9. The enigma surrounding IRF9 regulation by a phosphorylation event is intriguing. As IRF9 plays a major role in establishing an antiviral state in host cells, the topic of IRF9 regulation warrants deeper investigation. METHODS: Initially, total lysates of 2fTGH and U2A cells (transfected with recombinant IRF9) were filter-selected and concentrated using phosphoprotein enrichment assay. The phosphoprotein state of IRF9 was further confirmed using Phos-tag™ assay. All protein expression was determined using Western blotting. Tandem mass spectrometry was conducted on immunoprecipitated IRF9 to identify the phosphorylated amino acids. Finally, site-directed mutagenesis was performed and the effects of mutated IRF9 on relevant ISGs (i.e., USP18 and Mx1) was evaluated using qPCR. RESULTS: IRF9 is phosphorylated at S252 and S253 under IFNß-induced condition and R242 under non-induced condition. Site-directed mutagenesis of S252 and S253 to either alanine or aspartic acid has a modest effect on the upregulation of USP18 gene-a negative regulator of type I interferon (IFN) response-but not Mx1 gene. CONCLUSION: Our preliminary study shows that IRF9 is phosphorylated and possibly regulates USP18 gene expression. However, further in vivo studies are needed to determine the significance of IRF9 phosphorylation.


Asunto(s)
Interferón Tipo I , Quinasas Janus , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/genética , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/metabolismo , Fosforilación , Quinasas Janus/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Factor de Transcripción STAT2/genética , Factor de Transcripción STAT2/metabolismo , Interferón Tipo I/metabolismo
6.
Int J Mol Sci ; 24(5)2023 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-36902023

RESUMEN

In humans, four small HERCs (HERC3-6) exhibit differential degrees of antiviral activity toward HIV-1. Recently we revealed a novel member HERC7 of small HERCs exclusively in non-mammalian vertebrates and varied copies of herc7 genes in distinct fish species, raising a question of what is the exact role for a certain fish herc7 gene. Here, a total of four herc7 genes (named HERC7a-d sequentially) are identified in the zebrafish genome. They are transcriptionally induced by a viral infection, and detailed promoter analyses indicate that zebrafish herc7c is a typical interferon (IFN)-stimulated gene. Overexpression of zebrafish HERC7c promotes SVCV (spring viremia of carp virus) replication in fish cells and concomitantly downregulates cellular IFN response. Mechanistically, zebrafish HERC7c targets STING, MAVS, and IRF7 for protein degradation, thus impairing cellular IFN response. Whereas the recently-identified crucian carp HERC7 has an E3 ligase activity for the conjugation of both ubiquitin and ISG15, zebrafish HERC7c only displays the potential to transfer ubiquitin. Considering the necessity for timely regulation of IFN expression during viral infection, these results together suggest that zebrafish HERC7c is a negative regulator of fish IFN antiviral response.


Asunto(s)
Enfermedades de los Peces , Infecciones por Rhabdoviridae , Animales , Humanos , Pez Cebra/genética , Interferones/metabolismo , Proteínas de Pez Cebra/metabolismo , Antivirales , Ubiquitinas
7.
J Virol ; 95(9)2021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33568513

RESUMEN

Negative-sense RNA viruses (NSVs) rely on prepackaged viral RNA-dependent RNA polymerases (RdRp) to replicate and transcribe their viral genomes. Their replication machinery consists of an RdRp bound to viral RNA which is wound around a nucleoprotein (NP) scaffold, forming a viral ribonucleoprotein complex. NSV NP is known to regulate transcription and replication of genomic RNA; however, its role in maintaining and protecting the viral genetic material is unknown. Here, we exploited host microRNA expression to target NP of influenza A virus and Sendai virus to ascertain how this would impact genomic levels and the host response to infection. We find that in addition to inducing a drastic decrease in genome replication, the antiviral host response in the absence of NP is dramatically enhanced. Additionally, our data show that insufficient levels of NP prevent the replication machinery of these NSVs to process full-length genomes, resulting in aberrant replication products which form pathogen-associated molecular patterns in the process. These dynamics facilitate immune recognition by cellular pattern recognition receptors leading to a strong host antiviral response. Moreover, we observe that the consequences of limiting NP levels are universal among NSVs, including Ebola virus, Lassa virus, and measles virus. Overall, these results provide new insights into viral genome replication of negative-sense RNA viruses and highlight novel avenues for developing effective antiviral strategies, adjuvants, and/or live-attenuated vaccines.IMPORTANCE Negative-sense RNA viruses comprise some of the most important known human pathogens, including influenza A virus, measles virus, and Ebola virus. These viruses possess RNA genomes that are unreadable to the host, as they require specific viral RNA-dependent RNA polymerases in conjunction with other viral proteins, such as nucleoprotein, to be replicated and transcribed. As this process generates a significant amount of pathogen-associated molecular patterns, this phylum of viruses can result in a robust induction of the intrinsic host cellular response. To circumvent these defenses, these viruses form tightly regulated ribonucleoprotein replication complexes in order to protect their genomes from detection and to prevent excessive aberrant replication. Here, we demonstrate the balance that negative-sense RNA viruses must achieve both to replicate efficiently and to avoid induction of the host defenses.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/virología , Proteínas de la Nucleocápside/fisiología , Infecciones por Respirovirus/virología , Virus Sendai/fisiología , Replicación Viral , Células A549 , Animales , Chlorocebus aethiops , Perros , Células HEK293 , Células HeLa , Humanos , Células de Riñón Canino Madin Darby , Células Vero , Tropismo Viral
8.
Am J Kidney Dis ; 80(5): 677-682, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35817275

RESUMEN

Adenosine deaminase 2 (ADA2) deficiency is a rare autosomal recessive disease that is caused by loss-of-function mutations in the ADA2 gene. It is considered a monogenic form of polyarteritis nodosa and frequently is positive for a type I interferon (IFN) signature. Renal manifestations in ADA2 deficiency are poorly characterized. We herein report 2 cases of ADA2 deficiency with different kidney patterns due, respectively, to a predominantly macroscopic and microscopic vasculopathy, and review the literature on kidney disease in ADA2 deficiency. Patient 1 presented with a spontaneous perirenal hematoma; angiography demonstrated multiple microaneurysms but no further defects of the renal parenchyma; his kidney function remained normal. Patient 2 experienced slowly deteriorating kidney function and proteinuria. No major angiographic abnormalities were detected, while kidney biopsy revealed massive vasculopathy resembling chronic thrombotic microangiopathy (TMA) of the small and medium-sized vessels. Both patients had a positive peripheral type I IFN signature. In immunofluorescence staining of a kidney biopsy sample from patient 2, we observed marked expression of the type I IFN-induced protein MXA within endothelial cells, especially in vessels with TMA, and in infiltrating T cells. Our findings confirm that the kidney phenotype of ADA2 deficiency results from small and medium-sized vessel vasculopathy and suggest that type I IFN may be involved in the pathogenesis of kidney lesions.


Asunto(s)
Interferón Tipo I , Poliarteritis Nudosa , Enfermedades Vasculares , Humanos , Poliarteritis Nudosa/genética , Adenosina Desaminasa/genética , Células Endoteliales , Péptidos y Proteínas de Señalización Intercelular/genética , Fenotipo , Mutación , Riñón
9.
J Immunoassay Immunochem ; 43(4): 347-364, 2022 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-35227165

RESUMEN

Alteration of micro-RNAs (miRNAs) expression, including miRNA-122a, -146a and -205 family members, can have profound effects on inflammatory and IFN pathways (miRNA-146a), known as hallmarks of COVID-19. SARS-CoV-2-infected patients were recruited at Policlinico Umberto I Hospital of Sapienza University of Rome (Italy). MiRNA-122a, -146a, -205 and IFI27 (Interferon Alpha Inducible Protein 27) levels were screened in SARS-CoV-2 patients (n = 14) and healthy controls (n = 10) by real-time RT-PCR assays. Then, miRNA-146a rs2910164 GC single-nucleotide polymorphism (SNP) was genotyped in a larger group of COVID-19 patients (n = 129), and its relationship with severe disease [Intensive Care Unit (ICU) support or survival/death] was assessed. SARS-CoV-2-positive patients had increased PCR, D-Dimer and Fibrinogen levels compared to healthy controls (p < .05 for all measurements). MiRNA-122a and -146a serum levels were upregulated in COVID-19 patients (miRNA-122a: p = .002; miRNA-146a: p < .001). Decreased IFI27 levels were observed in COVID-19 patients with higher miRNA-146a levels (p = .047). Moreover, miRNA-146a rs2910164 C/G genotypes distributions were similar in COVID-19 patients and in validated European healthy subjects (n = 37,214). MiRNA-146a SNP was not associated with severe COVID-19 outcome (ICU or death). MiRNA-122a and -146a levels were elevated in SARS-CoV-2 infected patients, with miRNA-146a upregulation possibly contributing to IFN pathways dysregulation (e.g., reduced IFI27 levels) observed in severe COVID-19, although there is no evidence for the involvement of rs2910164 SNP.


Asunto(s)
COVID-19 , MicroARN Circulante , MicroARNs , Humanos , Estudios de Casos y Controles , COVID-19/genética , Predisposición Genética a la Enfermedad , Genotipo , MicroARNs/genética , Polimorfismo de Nucleótido Simple , SARS-CoV-2
10.
Int J Mol Sci ; 23(19)2022 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-36232993

RESUMEN

Coronavirus nonstructural protein 3 (nsp3) is a multi-functional protein, playing a critical role in viral replication and in regulating host antiviral innate immunity. In this study, we demonstrate that nsp3 from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and avian coronavirus infectious bronchitis virus (IBV) directly interacts with melanoma differentiation-associated gene 5 (MDA5), rendering an inhibitory effect on the MDA5-mediated type I interferon (IFN) response. By the co-expression of MDA5 with wild-type and truncated nsp3 constructs, at least three interacting regions mapped to the papain-like protease (PLpro) domain and two other domains located at the N- and C-terminal regions were identified in SARS-CoV-2 nsp3. Furthermore, by introducing point mutations to the catalytic triad, the deubiquitylation activity of the PLpro domain from both SARS-CoV-2 and IBV nsp3 was shown to be responsible for the suppression of the MDA5-mediated type I IFN response. It was also demonstrated that both MDA5 and nsp3 were able to interact with ubiquitin and ubiquitinated proteins, contributing to the interaction between the two proteins. This study confirms the antagonistic role of nsp3 in the MDA5-mediated type I IFN signaling, highlighting the complex interaction between a multi-functional viral protein and the innate immune response.


Asunto(s)
Infecciones por Coronavirus , Virus de la Bronquitis Infecciosa , Interferón Tipo I , Helicasa Inducida por Interferón IFIH1 , SARS-CoV-2 , Proteínas no Estructurales Virales , COVID-19 , Infecciones por Coronavirus/inmunología , Humanos , Virus de la Bronquitis Infecciosa/metabolismo , Interferón Tipo I/inmunología , Helicasa Inducida por Interferón IFIH1/metabolismo , SARS-CoV-2/metabolismo , Ubiquitina/metabolismo , Proteínas Ubiquitinadas , Proteínas no Estructurales Virales/metabolismo
11.
J Biol Chem ; 295(8): 2385-2397, 2020 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-31915247

RESUMEN

Viral infection is an environmental trigger that has been suggested to initiate pancreatic ß-cell damage, leading to the development of autoimmune diabetes. Viruses potently activate the immune system and can damage ß cells by either directly infecting them or stimulating the production of secondary effector molecules (such as proinflammatory cytokines) during bystander activation. However, how and where ß cells recognize viruses is unclear, and the antiviral responses that are initiated following virus recognition are incompletely understood. In this study, we show that the ß-cell response to dsRNA, a viral replication intermediate known to activate antiviral responses, is determined by the cellular location of sensing (intracellular versus extracellular) and differs from the cellular response to cytokine treatment. Using biochemical and immunological methods, we show that ß cells selectively respond to intracellular dsRNA by expressing type I interferons (IFNs) and inducing apoptosis, but that they do not respond to extracellular dsRNA. These responses differ from the activities of cytokines on ß cells, which are mediated by inducible nitric oxide synthase expression and ß-cell production of nitric oxide. These findings provide evidence that the antiviral activities of type I IFN production and apoptosis are elicited in ß cells via the recognition of intracellular viral replication intermediates and that ß cells lack the capacity to respond to extracellular viral intermediates known to activate innate immune responses.


Asunto(s)
Células Secretoras de Insulina/virología , ARN Bicatenario/metabolismo , Animales , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Supervivencia Celular/efectos de los fármacos , Daño del ADN , Activación Enzimática/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/patología , Interferón Tipo I/metabolismo , Masculino , Óxido Nítrico Sintasa de Tipo II/metabolismo , Poli I-C/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 3/metabolismo
12.
Clin Exp Allergy ; 51(2): 273-283, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33091192

RESUMEN

BACKGROUND: Upper respiratory tract infections (URTIs) are important triggers for asthma exacerbations. We hypothesized that inhalation of the anti-viral cytokine, interferon (IFN)-ß, during URTI, could prevent these exacerbations. OBJECTIVE: To evaluate the efficacy of on-demand inhaled IFN-ß1a (AZD9412) to prevent severe asthma exacerbations following symptomatic URTI. METHODS: This was a randomized, double-blind, placebo-controlled trial in which patients with severe asthma (GINA 4-5; n = 121) reporting URTI symptoms were randomized to 14 days of once-daily nebulized AZD9412 or placebo. The primary endpoint was severe exacerbations during treatment. Secondary endpoints included 6-item asthma control questionnaire (ACQ-6) and lung function. Exploratory biomarkers included IFN-response markers in serum and sputum, blood leucocyte counts and serum inflammatory cytokines. RESULTS: Following a pre-planned interim analysis, the trial was terminated early due to an unexpectedly low exacerbation rate. Asthma worsenings were generally mild and tended to peak at randomization, possibly contributing to the lack of benefit of AZD9412 on other asthma endpoints. Numerically, AZD9412 did not reduce severe exacerbation rate, ACQ-6, asthma symptom scores or reliever medication use. AZD9412 improved lung function (morning peak expiratory flow; mPEF) by 19.7 L/min. Exploratory post hoc analyses indicated a greater mPEF improvement by AZD9412 in patients with high blood eosinophils (>0.3 × 109 /L) at screening and low serum interleukin-18 relative change at pre-treatment baseline. Pharmacodynamic effect of AZD9412 was confirmed using IFN-response markers. CONCLUSIONS & CLINICAL RELEVANCE: Colds did not have the impact on asthma patients that was expected and, due to the low exacerbation rate, the trial was stopped early. On-demand AZD9412 treatment did not numerically reduce the number of exacerbations, but did attenuate URTI-induced worsening of mPEF. Severe asthma patients with high blood eosinophils or low serum interleukin-18 response are potential subgroups for further investigation of inhaled IFN-ß1a.


Asunto(s)
Antivirales/uso terapéutico , Asma/tratamiento farmacológico , Interferón beta-1a/uso terapéutico , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Administración por Inhalación , Adulto , Asma/sangre , Asma/complicaciones , Asma/fisiopatología , Citocinas/sangre , Progresión de la Enfermedad , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Ápice del Flujo Espiratorio/fisiología , Infecciones del Sistema Respiratorio/sangre , Infecciones del Sistema Respiratorio/complicaciones , Infecciones del Sistema Respiratorio/fisiopatología , Índice de Severidad de la Enfermedad
13.
Methods ; 183: 21-29, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31682923

RESUMEN

Mammalian cells express different types of RNA molecules that can be classified as protein coding RNAs (mRNA) and non-coding RNAs (ncRNAs) the latter of which have housekeeping and regulatory functions in cells. Cellular RNAs are not recognized by cellular pattern recognition receptors (PRRs) and innate immunity is not activated. RNA viruses encode and express RNA molecules that usually differ from cell-specific RNAs and they include for instance 5'capped and 5'mono- and triphosphorylated RNAs, small viral RNAs and viral RNA-protein complexes called vRNPs. These molecules are recognized by certain members of Toll-like receptor (TLR) and RIG-I-like receptor (RLR) families leading to activation of innate immune responses and the production of antiviral cytokines, such as type I and type III interferons (IFNs). Virus-specific ssRNA and dsRNA molecules that mimic the viral genomic RNAs or their replication intermediates can efficiently be produced by bacteriophage T7 DNA-dependent RNA polymerase and bacteriophage phi6 RNA-dependent RNA polymerase, respectively. These molecules can then be delivered into mammalian cells and the mechanisms of activation of innate immune responses can be studied. In addition, synthetic viral dsRNAs can be processed to small interfering RNAs (siRNAs) by a Dicer enzyme to produce a swarm of antiviral siRNAs. Here we describe the biology of RNAs, their in vitro production and delivery into mammalian cells as well as how these molecules can be used to inhibit virus replication and to study the mechanisms of activation of the innate immune system.


Asunto(s)
Inmunidad Innata/genética , Técnicas In Vitro/métodos , ARN Interferente Pequeño/metabolismo , ARN Viral/metabolismo , Transfección/métodos , Animales , Células Cultivadas , ARN Polimerasas Dirigidas por ADN/metabolismo , Genoma Viral , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/inmunología , Humanos , Interferones/inmunología , Interferones/metabolismo , Cultivo Primario de Células/métodos , Interferencia de ARN , Virus ARN/genética , Virus ARN/inmunología , ARN Bicatenario , ARN Viral/inmunología , ARN Polimerasa Dependiente del ARN/metabolismo , Ribonucleasa III/metabolismo , Proteínas Virales/metabolismo , Replicación Viral/genética , Replicación Viral/inmunología
14.
Adv Exp Med Biol ; 1304: 53-71, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34019263

RESUMEN

Innate immunity is the first defense line of the host against various infectious pathogens, environmental insults, and other stimuli causing cell damages. Upon stimulation, pattern recognition receptors (PRRs) act as sensors to activate innate immune responses, containing NF-κB signaling, IFN response, and inflammasome activation. Toll-like receptors (TLRs), retinoic acid-inducible gene I-like receptors (RLRs), NOD-like receptors (NLRs), and other nucleic acid sensors are involved in innate immune responses. The activation of innate immune responses can facilitate the host to eliminate pathogens and maintain tissue homeostasis. However, the activity of innate immune responses needs to be tightly controlled to ensure the optimal intensity and duration of activation under various contexts. Uncontrolled innate immune responses can lead to various disorders associated with aberrant inflammatory response, including pulmonary diseases such as COPD, asthma, and COVID-19. In this chapter, we will have a broad overview of how innate immune responses function and the regulation and activation of innate immune response at molecular levels as well as their contribution to various pulmonary diseases. A better understanding of such association between innate immune responses and pulmonary diseases may provide potential therapeutic strategies.


Asunto(s)
COVID-19 , Humanos , Inmunidad Innata , Receptores de Reconocimiento de Patrones , SARS-CoV-2 , Receptores Toll-Like/genética
15.
J Cell Mol Med ; 24(18): 10803-10815, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32757451

RESUMEN

Some studies suggested the prognosis value of immune gene in lower grade glioma (LGG). Recurrence in LGG is a tough clinical problem for many LGG patients. Therefore, prognosis biomarker is required. Multivariate prognosis Cox model was constructed and then calculated the risk score. And differential expressed transcription factors (TFs) and differential expressed immune genes (DEIGs) were co-analysed. Besides, significant immune cells/pathways were identified by single sample gene set enrichment analysis (ssGSEA). Moreover, gene set variation analysis (GSVA) and univariate Cox regression were applied to filter prognostic signalling pathways. Additionally, significant DEIG and immune cells/pathways, and significant DEIG and pathways were co-analysed. Further, differential enriched pathways were identified by GSEA. In sum, a scientific hypothesis for recurrence LGG including TF, immune gene and immune cell/pathway was established. In our study, a total of 536 primary LGG samples, 2,498 immune genes and 318 TFs were acquired. Based on edgeR method, 2,164 DEGs, 2,498 DEIGs and 31 differentials expressed TFs were identified. A total of 106 DEIGs were integrated into multivariate prognostic model. Additionally, the AUC of the ROC curve was 0.860, and P value of Kaplan-Meier curve < 0.001. GATA6 (TF) and COL3A1 (DEIG) were selected (R = 0.900, P < 0.001, positive) as significant TF-immune gene links. Type II IFN response (P < 0.001) was the significant immune pathway. Propanoate metabolism (P < 0.001) was the significant KEGG pathway. We proposed that COL3A1 was positively regulated by GATA6, and by effecting type II IFN response and propanoate metabolism, COL3A1 involved in LGG recurrence.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Colágeno Tipo III/fisiología , Factor de Transcripción GATA6/fisiología , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , Interferón alfa-2/biosíntesis , Proteínas de Neoplasias/fisiología , Recurrencia Local de Neoplasia/metabolismo , Propionatos/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Femenino , Redes Reguladoras de Genes , Glioma/genética , Glioma/inmunología , Glioma/patología , Humanos , Interferón alfa-2/genética , Masculino , Redes y Vías Metabólicas/genética , Redes y Vías Metabólicas/inmunología , Persona de Mediana Edad , Clasificación del Tumor , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Recurrencia Local de Neoplasia/patología , Pronóstico , Modelos de Riesgos Proporcionales , Riesgo , Adulto Joven
16.
BMC Vet Res ; 16(1): 450, 2020 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-33213475

RESUMEN

BACKGROUND: Interferon (IFN) regulatory factors (IRFs), as transcriptional regulatory factors, play important roles in regulating the expression of type I IFN and IFN- stimulated genes (ISGs) in innate immune responses. In addition, they participate in cell growth and development and regulate oncogenesis. RESULTS: In the present study, the cDNA sequence of IRF10 in common carp (Cyprinus carpio L.) was characterized (abbreviation, CcIRF10). The predicted protein sequence of CcIRF10 shared 52.7-89.2% identity with other teleost IRF10s and contained a DNA-binding domain (DBD), a nuclear localization signal (NLS) and an IRF-associated domain (IAD). Phylogenetic analysis showed that CcIRF10 had the closest relationship with IRF10 of Ctenopharyngodon idella. CcIRF10 transcripts were detectable in all examined tissues, with the highest expression in the gonad and the lowest expression in the head kidney. CcIRF10 expression was upregulated in the spleen, head kidney, foregut and hindgut upon polyinosinic:polycytidylic acid (poly I:C) and Aeromonas hydrophila stimulation and induced by poly I:C, lipopolysaccharide (LPS) and peptidoglycan (PGN) in peripheral blood leucocytes (PBLs) and head kidney leukocytes (HKLs) of C. carpio. In addition, overexpression of CcIRF10 was able to decrease the expression of the IFN and IFN-stimulated genes PKR and ISG15. CONCLUSIONS: These results indicate that CcIRF10 participates in antiviral and antibacterial immunity and negatively regulates the IFN response, which provides new insights into the IFN system of C. carpio.


Asunto(s)
Carpas/genética , Carpas/inmunología , Factores Reguladores del Interferón/genética , Aeromonas hydrophila/inmunología , Animales , Carpas/metabolismo , ADN Complementario , Proteínas de Peces , Lipopolisacáridos/inmunología , Peptidoglicano/inmunología , Filogenia , Poli I-C/inmunología , Análisis de Secuencia de ADN , Distribución Tisular
17.
Int J Mol Sci ; 21(4)2020 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-32079277

RESUMEN

RNA silencing is a posttranscriptional gene silencing mechanism directed by endogenous small non-coding RNAs called microRNAs (miRNAs). By contrast, the type-I interferon (IFN) response is an innate immune response induced by exogenous RNAs, such as viral RNAs. Endogenous and exogenous RNAs have typical structural features and are recognized accurately by specific RNA-binding proteins in each pathway. In mammalian cells, both RNA silencing and the IFN response are induced by double-stranded RNAs (dsRNAs) in the cytoplasm, but have long been considered two independent pathways. However, recent reports have shed light on crosstalk between the two pathways, which are mutually regulated by protein-protein interactions triggered by viral infection. This review provides brief overviews of RNA silencing and the IFN response and an outline of the molecular mechanism of their crosstalk and its biological implications. Crosstalk between RNA silencing and the IFN response may reveal a novel antiviral defense system that is regulated by miRNAs in mammalian cells.


Asunto(s)
Antivirales/farmacología , Interferones/metabolismo , Interferones/farmacología , Interferencia de ARN/fisiología , Animales , Citoplasma/metabolismo , Citoplasma/virología , Silenciador del Gen , Humanos , Inmunidad Innata , Interferón Tipo I/farmacología , Interferones/genética , Interferones/inmunología , MicroARNs/biosíntesis , MicroARNs/genética , ARN Bicatenario , ARN Viral/efectos de los fármacos , Proteínas de Unión al ARN/metabolismo , Virus
18.
Fish Shellfish Immunol ; 86: 1058-1063, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30593899

RESUMEN

In mammals, virus infection of host cells triggers innate immune response, characterized by induction of interferon (IFN) and downstream IFN-stimulated genes (ISGs). The initiation of IFN antiviral response is dependent on host recognition of virus infection. In fish, similar IFN antiviral response is induced in response to RNA or DNA virus infection; however, the detailed mechanisms underlying recognition of a given virus and activation of downstream signaling remain largely unexplored. Using an infection model with Epithelioma papulosum cyprini (EPC) cells and spring viremia of carp virus (SVCV), a negative sense single-stranded RNA virus, we reported that fish RLR signaling pathway was involved in SVCV-triggered fish IFN response. IFN response was significantly initiated in EPC cells when infected with SVCV, as evidenced by activation of fish IFN promoters, upregulation of IFN and ISGs at mRNA and protein levels. However, function blockade of RIG-I and MDA5, two cytosolic receptors of fish RLR family, significantly attenuated the activation of fish IFN promoters and also the induction of fish IFN and ISGs by SVCV infection. Consistently, SVCV infection-triggered IFN response were blocked in EPC cells when transfected with the dominant negative mutants of pivotal RLR signaling factors, including MAVS, MITA, TBK1, IRF3 and IRF7. These results together shed light on the conservation of RLR-mediated IFN signaling that contributes to fish cells responding to RNA virus infection.


Asunto(s)
Enfermedades de los Peces/inmunología , Infecciones por Rhabdoviridae/veterinaria , Rhabdoviridae/fisiología , Transducción de Señal , Animales , Línea Celular Tumoral , Cyprinidae/inmunología , Proteína 58 DEAD Box/metabolismo , Enfermedades de los Peces/virología , Inmunidad Innata , Interferones/inmunología , Regiones Promotoras Genéticas , Infecciones por Rhabdoviridae/inmunología
19.
Fish Shellfish Immunol ; 86: 152-159, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30448445

RESUMEN

Teleost fish possess two groups of type I interferons (IFNs) with two (group I IFNs) or four (group II IFNs) conserved cysteines, which are further classified into seven subgroups. In our previous study, two group I type I IFNs, LcIFNd and LcIFNh (a new subgroup member), were identified in the perciform fish, large yellow croaker (Larimichthys crocea). Here, we identified a group II type I IFN, LcIFNc, in this species. The deduced LcIFNc contained six cysteines, four of which are highly conserved (C1: C28, C2:C53, C3: C130, and C4:C159) in the fish group II type I IFNs, and a typical type I IFN signature motif was also found in it. Phylogenetic analysis indicated that LcIFNc belongs to the IFNc subgroup of fish group II type I IFNs. LcIFNc was constitutively expressed in all examined tissues, and was rapidly up-regulated in spleen and head kidney by poly(I:C) and Aeromonas hydrophila. Recombinant LcIFNc protein (rLcIFNc) could increase the expression of antiviral genes, Mx1, PKR and ISG15, in large yellow croaker peripheral blood leukocytes (PBLs). The rLcIFNc also exhibited obvious antiviral activity based on less cytopathic effect (CPE) and decreased expression levels of several viral genes in the rLcIFNc-treated grouper spleen (GS) cells following Singapore grouper iridovirus (SGIV) infection. Additionally, rLcIFNc was able to induce the expression of LcIFNc, as well as LcIFNd and LcIFNh in the PBLs and primary head kidney cells (HKCs) from large yellow croaker. These results therefore indicated that LcIFNc not only had antiviral activity, but also mediated the regulation of type I IFN response.


Asunto(s)
Interferón Tipo I/metabolismo , Perciformes/metabolismo , Secuencia de Aminoácidos , Animales , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Interferón Tipo I/química , Interferón Tipo I/genética , Filogenia , Especificidad de la Especie , Bazo/citología , Distribución Tisular
20.
Rev Med Virol ; 28(6): e2008, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30209859

RESUMEN

Increased sensitivity of cancer cells to viruses is a prerequisite for the success of oncolytic virotherapy. One of the major causes of such a phenotype is the disruption of innate antiviral defenses associated with dysfunction of type 1 interferons (IFNs) that permits unlimited replication of viruses in cancer cells. Defects in IFN pathways help cancer progression by providing additional advantages to tumor cells. However, while these defects promote the survival and accelerated proliferation of malignant cells, they facilitate viral replication and thus enhance the efficiency of viral oncolysis. This review describes a broad spectrum of defects in genes that participate in IFN induction and IFN response pathways. Expression levels and/or functional activities of these genes are frequently low or absent in cancer cells, making them sensitive to virus infection. Therefore, certain specific defects in IFN signaling cascades might serve as potential biomarkers to help in identifying individual cancer patients who are likely to benefit from oncolytic virotherapy.


Asunto(s)
Antineoplásicos/inmunología , Biomarcadores/análisis , Interferones/deficiencia , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/inmunología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA