Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.229
Filtrar
1.
Curr Opin Immunol ; 87: 102430, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38824869

RESUMEN

Lambda interferons (IFNλs), also termed type III interferons (IFNs) or interleukins-28/29, have been in the shadow of type I IFNs for a long time. Their common induction mechanisms and signalling cascades with type I IFNs have made difficult the unwinding of their unique nonredundant functions. However, this is now changing with mounting evidence supporting a major role of IFNλs as a specialized antiviral defense system in the body, mediating protection at mucosal barrier surfaces while limiting immunopathology. Here, we review the latest progress on the complex activities of IFNλs in the respiratory tract, focusing on their multiple effects in IFNλ receptor-expressing cells, the modulation of innate and adaptive immune responses in the context of infections and respiratory diseases, and their similarities and differences with type I IFNs. We also discuss their potential in therapeutic applications and the most recent developments in that direction.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Interferón lambda , Interferones , Sistema Respiratorio , Humanos , Animales , Interferones/metabolismo , Interferones/inmunología , Sistema Respiratorio/inmunología , Sistema Respiratorio/metabolismo , Transducción de Señal/inmunología , Interferón Tipo I/metabolismo , Interferón Tipo I/inmunología
2.
Nat Commun ; 15(1): 4920, 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38858353

RESUMEN

The differentiation of the stroma is a hallmark event during postnatal uterine development. However, the spatiotemporal changes that occur during this process and the underlying regulatory mechanisms remain elusive. Here, we comprehensively delineated the dynamic development of the neonatal uterus at single-cell resolution and characterized two distinct stromal subpopulations, inner and outer stroma. Furthermore, single-cell RNA sequencing revealed that uterine ablation of Pr-set7, the sole methyltransferase catalyzing H4K20me1, led to a reduced proportion of the inner stroma due to massive cell death, thus impeding uterine development. By combining RNA sequencing and epigenetic profiling of H4K20me1, we demonstrated that PR-SET7-H4K20me1 either directly repressed the transcription of interferon stimulated genes or indirectly restricted the interferon response via silencing endogenous retroviruses. Declined H4K20me1 level caused viral mimicry responses and ZBP1-mediated apoptosis and necroptosis in stromal cells. Collectively, our study provides insight into the epigenetic machinery governing postnatal uterine stromal development mediated by PR-SET7.


Asunto(s)
Epigénesis Genética , N-Metiltransferasa de Histona-Lisina , Células del Estroma , Útero , Femenino , Animales , Útero/metabolismo , Células del Estroma/metabolismo , Ratones , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Interferones/metabolismo , Interferones/genética , Retrovirus Endógenos/genética , Apoptosis/genética , Ratones Endogámicos C57BL , Muerte Celular/genética , Necroptosis/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Histonas/metabolismo , Análisis de la Célula Individual , Ratones Noqueados , Diferenciación Celular/genética
3.
Int J Mol Sci ; 25(11)2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38891862

RESUMEN

RNA processing is a highly conserved mechanism that serves as a pivotal regulator of gene expression. Alternative processing generates transcripts that can still be translated but lead to potentially nonfunctional proteins. A plethora of respiratory viruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), strategically manipulate the host's RNA processing machinery to circumvent antiviral responses. We integrated publicly available omics datasets to systematically analyze isoform-level expression and delineate the nascent peptide landscape of SARS-CoV-2-infected human cells. Our findings explore a suggested but uncharacterized mechanism, whereby SARS-CoV-2 infection induces the predominant expression of unproductive splicing isoforms in key IFN signaling, interferon-stimulated (ISGs), class I MHC, and splicing machinery genes, including IRF7, HLA-B, and HNRNPH1. In stark contrast, cytokine and chemokine genes, such as IL6 and TNF, predominantly express productive (protein-coding) splicing isoforms in response to SARS-CoV-2 infection. We postulate that SARS-CoV-2 employs an unreported tactic of exploiting the host splicing machinery to bolster viral replication and subvert the immune response by selectively upregulating unproductive splicing isoforms from antigen presentation and antiviral response genes. Our study sheds new light on the molecular interplay between SARS-CoV-2 and the host immune system, offering a foundation for the development of novel therapeutic strategies to combat COVID-19.


Asunto(s)
Empalme Alternativo , COVID-19 , Interferones , Isoformas de Proteínas , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , COVID-19/virología , COVID-19/genética , COVID-19/inmunología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferones/metabolismo , Interferones/genética , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo
4.
Virulence ; 15(1): 2359470, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38918890

RESUMEN

Influenza A virus (IAV) is the leading cause of highly contagious respiratory infections, which poses a serious threat to public health. The non-structural protein 1 (NS1) is encoded by segment 8 of IAV genome and is expressed in high levels in host cells upon IAV infection. It is the determinant of virulence and has multiple functions by targeting type Ι interferon (IFN-I) and type III interferon (IFN-III) production, disrupting cell apoptosis and autophagy in IAV-infected cells, and regulating the host fitness of influenza viruses. This review will summarize the current research on the NS1 including the structure and related biological functions of the NS1 as well as the interaction between the NS1 and host cells. It is hoped that this will provide some scientific basis for the prevention and control of the influenza virus.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Proteínas no Estructurales Virales , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/patogenicidad , Virus de la Influenza A/fisiología , Gripe Humana/virología , Animales , Autofagia , Virulencia , Interacciones Huésped-Patógeno , Apoptosis , Interferones/metabolismo , Interferones/inmunología , Interferones/genética
6.
New Microbiol ; 47(1): 60-67, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38700885

RESUMEN

Acute respiratory tract infection (ARTI) is common in all age groups, especially in children and the elderly. About 85% of children who present with bronchiolitis are infected with respiratory syncytial virus (RSV); however, nearly one-third are coinfected with another respiratory virus, such as human rhinovirus (HRV). Therefore, it is necessary to explore the immune response to coinfection to better understand the molecular and cellular pathways involving virus-virus interactions that might be modulated by innate immunity and additional host cell response mechanisms. This study aims to investigate the host innate immune response against RSV-HRV coinfection compared with monoinfection. Human primary bronchial/tracheal epithelial cells (HPECs) were infected with RSV, HRV, or coinfected with both viruses, and the infected cells were collected at 48 and 72 hours. Gene expression profiles of IL-6, CCL5, TNF-α, IFN-ß, IFN-λ1, CXCL10, IL-10, IL-13, IRF3, and IRF7 were investigated using real-time quantitative PCR, which revealed that RSV-infected cells exhibited increased expression of IL-10, whereas HRV infection increased the expression of CXCL10, IL-10, and CCL5. IFN-λ1 and CXCL10 expression was significantly different between the coinfection and monoinfection groups. In conclusion, our study revealed that two important cytokines, IFN-λ1 and CXCL10, exhibited increased expression during coinfection.


Asunto(s)
Bronquios , Quimiocina CXCL10 , Coinfección , Células Epiteliales , Interferón lambda , Interferones , Interleucinas , Infecciones por Picornaviridae , Infecciones por Virus Sincitial Respiratorio , Rhinovirus , Humanos , Rhinovirus/fisiología , Coinfección/virología , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Células Epiteliales/virología , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/virología , Bronquios/virología , Bronquios/citología , Infecciones por Picornaviridae/virología , Infecciones por Picornaviridae/inmunología , Interferones/genética , Interferones/metabolismo , Virus Sincitial Respiratorio Humano/fisiología , Virus Sincitial Respiratorio Humano/genética , Células Cultivadas , Virus Sincitiales Respiratorios/fisiología
7.
Biotechnol J ; 19(5): e2300672, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38719621

RESUMEN

The production of recombinant adeno-associated virus (rAAV) for gene therapy applications relies on the use of various host cell lines, with suspension-grown HEK293 cells being the preferred expression system due to their satisfactory rAAV yields in transient transfections. As the field of gene therapy continues to expand, there is a growing demand for efficient rAAV production, which has prompted efforts to optimize HEK293 cell line productivity through engineering. In contrast to other cell lines like CHO cells, the transcriptome of HEK293 cells during rAAV production has remained largely unexplored in terms of identifying molecular components that can enhance yields. In our previous research, we analyzed global regulatory pathways and mRNA expression patterns associated with increased rAAV production in HEK293 cells. Our data revealed substantial variations in the expression patterns between cell lines with low (LP) and high-production (HP) rates. Moving to a deeper layer for a more detailed analysis of inflammation-related transcriptome data, we detected an increased expression of interferon-related genes in low-producing cell lines. Following upon these results, we investigated the use of Ruxolitinib, an interferon pathway inhibitor, during the transient production of rAAV in HEK293 cells as potential media additive to boost rAAV titers. Indeed, we find a two-fold increase in rAAV titers compared to the control when the interferon pathways were inhibited. In essence, this work offers a rational design approach for optimization of HEK293 cell line productivity and potential engineering targets, ultimately paving the way for more cost-efficient and readily available gene therapies for patients.


Asunto(s)
Dependovirus , Interferones , Transducción de Señal , Humanos , Células HEK293 , Dependovirus/genética , Interferones/metabolismo , Interferones/genética , Nitrilos/farmacología , Pirimidinas/farmacología , Transfección , Pirazoles/farmacología
8.
Viruses ; 16(5)2024 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-38793622

RESUMEN

The pathogenesis of viral infection is attributed to two folds: intrinsic cell death pathway activation due to the viral cytopathic effect, and immune-mediated extrinsic cellular injuries. The immune system, encompassing both innate and adaptive immunity, therefore acts as a double-edged sword in viral infection. Insufficient potency permits pathogens to establish lifelong persistent infection and its consequences, while excessive activation leads to organ damage beyond its mission to control viral pathogens. The innate immune response serves as the front line of defense against viral infection, which is triggered through the recognition of viral products, referred to as pathogen-associated molecular patterns (PAMPs), by host cell pattern recognition receptors (PRRs). The PRRs-PAMPs interaction results in the induction of interferon-stimulated genes (ISGs) in infected cells, as well as the secretion of interferons (IFNs), to establish a tissue-wide antiviral state in an autocrine and paracrine manner. Cumulative evidence suggests significant variability in the expression patterns of PRRs, the induction potency of ISGs and IFNs, and the IFN response across different cell types and species. Hence, in our understanding of viral hepatitis pathogenesis, insights gained through hepatoma cell lines or murine-based experimental systems are uncertain in precisely recapitulating the innate antiviral response of genuine human hepatocytes. Accordingly, this review article aims to extract and summarize evidence made possible with bona fide human hepatocytes-based study tools, along with their clinical relevance and implications, as well as to identify the remaining gaps in knowledge for future investigations.


Asunto(s)
Virus de la Hepatitis Delta , Hepatocitos , Inmunidad Innata , Interferones , Receptores de Reconocimiento de Patrones , Humanos , Hepatitis D/inmunología , Hepatitis D/virología , Virus de la Hepatitis Delta/inmunología , Virus de la Hepatitis Delta/fisiología , Hepatocitos/virología , Hepatocitos/inmunología , Interacciones Huésped-Patógeno/inmunología , Interferones/inmunología , Interferones/metabolismo , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Receptores de Reconocimiento de Patrones/metabolismo , Receptores de Reconocimiento de Patrones/inmunología
9.
Viruses ; 16(5)2024 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-38793616

RESUMEN

Interferons (IFNs) are antiviral cytokines that defend against viral infections by inducing the expression of interferon-stimulated genes (ISGs). Interferon-inducible transmembrane proteins (IFITMs) 1, 2, and 3 are crucial ISG products and members of the CD225 protein family. Compelling evidence shows that IFITMs restrict the infection of many unrelated viruses by inhibiting the virus-cell membrane fusion at the virus entry step via the modulation of lipid composition and membrane properties. Meanwhile, viruses can evade IFITMs' restrictions by either directly interacting with IFITMs via viral glycoproteins or by altering the native entry pathway. At the same time, cumulative evidence suggests context-dependent and multifaceted roles of IFITMs in modulating virus infections and cell signaling. Here, we review the diverse antiviral mechanisms of IFITMs, the viral antagonizing strategies, and the regulation of IFITM activity in host cells. The mechanisms behind the antiviral activity of IFITMs could aid the development of broad-spectrum antivirals and enhance preparedness for future pandemics.


Asunto(s)
Interferones , Proteínas de la Membrana , Internalización del Virus , Humanos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/inmunología , Interferones/inmunología , Interferones/metabolismo , Internalización del Virus/efectos de los fármacos , Antivirales/farmacología , Evasión Inmune , Animales , Virosis/inmunología , Virosis/virología , Virus/inmunología , Virus/efectos de los fármacos , Interacciones Huésped-Patógeno/inmunología , Transducción de Señal , Antígenos de Diferenciación/metabolismo , Antígenos de Diferenciación/inmunología
10.
Nat Commun ; 15(1): 4177, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755196

RESUMEN

Plasma RNAemia, delayed antibody responses and inflammation predict COVID-19 outcomes, but the mechanisms underlying these immunovirological patterns are poorly understood. We profile 782 longitudinal plasma samples from 318 hospitalized patients with COVID-19. Integrated analysis using k-means reveals four patient clusters in a discovery cohort: mechanically ventilated critically-ill cases are subdivided into good prognosis and high-fatality clusters (reproduced in a validation cohort), while non-critical survivors segregate into high and low early antibody responders. Only the high-fatality cluster is enriched for transcriptomic signatures associated with COVID-19 severity, and each cluster has distinct RBD-specific antibody elicitation kinetics. Both critical and non-critical clusters with delayed antibody responses exhibit sustained IFN signatures, which negatively correlate with contemporaneous RBD-specific IgG levels and absolute SARS-CoV-2-specific B and CD4+ T cell frequencies. These data suggest that the "Interferon paradox" previously described in murine LCMV models is operative in COVID-19, with excessive IFN signaling delaying development of adaptive virus-specific immunity.


Asunto(s)
Anticuerpos Antivirales , COVID-19 , Interferones , SARS-CoV-2 , Transducción de Señal , Humanos , COVID-19/inmunología , SARS-CoV-2/inmunología , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Transducción de Señal/inmunología , Interferones/metabolismo , Interferones/inmunología , Femenino , Masculino , Persona de Mediana Edad , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Linfocitos T CD4-Positivos/inmunología , Anciano , Adulto , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Glicoproteína de la Espiga del Coronavirus/genética
11.
Front Immunol ; 15: 1383358, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38779657

RESUMEN

Introduction: Immune cells that contribute to the pathogenesis of systemic lupus erythematosus (SLE) derive from adult hematopoietic stem and progenitor cells (HSPCs) within the bone marrow (BM). For this reason, we reasoned that fundamental abnormalities in SLE can be traced to a BM-derived HSPC inflammatory signature. Methods: BM samples from four SLE patients, six healthy controls, and two umbilical cord blood (CB) samples were used. CD34+ cells were isolated from BM and CB samples, and single-cell RNA-sequencing was performed. Results: A total of 426 cells and 24,473 genes were used in the analysis. Clustering analysis resulted in seven distinct clusters of cell types. Mutually exclusive markers, which were characteristic of each cell type, were identified. We identified three HSPC subpopulations, one of which consisted of proliferating cells (MKI67 expressing cells), one T-like, one B-like, and two myeloid-like progenitor subpopulations. Differential expression analysis revealed i) cell cycle-associated signatures, in healthy BM of HSPC clusters 3 and 4 when compared with CB, and ii) interferon (IFN) signatures in SLE BM of HSPC clusters 3 and 4 and myeloid-like progenitor cluster 5 when compared with healthy controls. The IFN signature in SLE appeared to be deregulated following TF regulatory network analysis and differential alternative splicing analysis between SLE and healthy controls in HSPC subpopulations. Discussion: This study revealed both quantitative-as evidenced by decreased numbers of non-proliferating early progenitors-and qualitative differences-characterized by an IFN signature in SLE, which is known to drive loss of function and depletion of HSPCs. Chronic IFN exposure affects early hematopoietic progenitors in SLE, which may account for the immune aberrancies and the cytopenias in SLE.


Asunto(s)
Perfilación de la Expresión Génica , Células Madre Hematopoyéticas , Interferones , Lupus Eritematoso Sistémico , Análisis de la Célula Individual , Transcriptoma , Humanos , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Células Madre Hematopoyéticas/metabolismo , Interferones/metabolismo , Interferones/genética , Femenino , Adulto , Reprogramación Celular/genética , Masculino
12.
Mol Immunol ; 170: 156-169, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38692097

RESUMEN

Type-I and -III interferons play a central role in immune rejection of pathogens and tumors, thus promoting immunogenicity and suppressing tumor recurrence. Double strand RNA is an important ligand that stimulates tumor immunity via interferon responses. Differentiation of embryonic stem cells to pluripotent epithelial cells activates the interferon response during development, raising the question of whether epithelial vs. mesenchymal gene signatures in cancer potentially regulate the interferon pathway as well. Here, using genomics and signaling approaches, we show that Grainyhead-like-2 (GRHL2), a master programmer of epithelial cell identity, promotes type-I and -III interferon responses to double-strand RNA. GRHL2 enhanced the activation of IRF3 and relA/NF-kB and the expression of IRF1; a functional GRHL2 binding site in the IFNL1 promoter was also identified. Moreover, time to recurrence in breast cancer correlated positively with GRHL2 protein expression, indicating that GRHL2 is a tumor recurrence suppressor, consistent with its enhancement of interferon responses. These observations demonstrate that epithelial cell identity supports interferon responses in the context of cancer.


Asunto(s)
Neoplasias de la Mama , Proteínas de Unión al ADN , Factores de Transcripción , Humanos , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Neoplasias de la Mama/genética , Femenino , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Factor 3 Regulador del Interferón/genética , Recurrencia Local de Neoplasia/inmunología , Interferones/metabolismo , Interferones/inmunología , Interferones/genética , Línea Celular Tumoral , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Animales , ARN Bicatenario/inmunología , Factor de Transcripción ReIA/metabolismo , Ratones , Regulación Neoplásica de la Expresión Génica , Transducción de Señal/inmunología , Factor 1 Regulador del Interferón/metabolismo , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/inmunología
13.
Front Cell Infect Microbiol ; 14: 1365221, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38711929

RESUMEN

Bunyaviruses are a large group of important viral pathogens that cause significant diseases in humans and animals worldwide. Bunyaviruses are enveloped, single-stranded, negative-sense RNA viruses that infect a wide range of hosts. Upon entry into host cells, the components of viruses are recognized by host innate immune system, leading to the activation of downstream signaling cascades to induce interferons (IFNs) and other proinflammatory cytokines. IFNs bind to their receptors and upregulate the expression of hundreds of interferon-stimulated genes (ISGs). Many ISGs have antiviral activities and confer an antiviral state to host cells. For efficient replication and spread, viruses have evolved different strategies to antagonize IFN-mediated restriction. Here, we discuss recent advances in our understanding of the interactions between bunyaviruses and host innate immune response.


Asunto(s)
Infecciones por Bunyaviridae , Inmunidad Innata , Orthobunyavirus , Infecciones por Bunyaviridae/inmunología , Infecciones por Bunyaviridae/virología , Humanos , Animales , Orthobunyavirus/inmunología , Interacciones Huésped-Patógeno/inmunología , Interferones/inmunología , Interferones/metabolismo , Transducción de Señal , Citocinas/metabolismo , Citocinas/inmunología , Enfermedades Transmitidas por Vectores/inmunología , Enfermedades Transmitidas por Vectores/virología , Enfermedades Transmitidas por Vectores/prevención & control , Replicación Viral
14.
Nat Commun ; 15(1): 4067, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38744958

RESUMEN

The complexity of the tumor microenvironment poses significant challenges in cancer therapy. Here, to comprehensively investigate the tumor-normal ecosystems, we perform an integrative analysis of 4.9 million single-cell transcriptomes from 1070 tumor and 493 normal samples in combination with pan-cancer 137 spatial transcriptomics, 8887 TCGA, and 1261 checkpoint inhibitor-treated bulk tumors. We define a myriad of cell states constituting the tumor-normal ecosystems and also identify hallmark gene signatures across different cell types and organs. Our atlas characterizes distinctions between inflammatory fibroblasts marked by AKR1C1 or WNT5A in terms of cellular interactions and spatial co-localization patterns. Co-occurrence analysis reveals interferon-enriched community states including tertiary lymphoid structure (TLS) components, which exhibit differential rewiring between tumor, adjacent normal, and healthy normal tissues. The favorable response of interferon-enriched community states to immunotherapy is validated using immunotherapy-treated cancers (n = 1261) including our lung cancer cohort (n = 497). Deconvolution of spatial transcriptomes discriminates TLS-enriched from non-enriched cell types among immunotherapy-favorable components. Our systematic dissection of tumor-normal ecosystems provides a deeper understanding of inter- and intra-tumoral heterogeneity.


Asunto(s)
Neoplasias , Análisis de la Célula Individual , Transcriptoma , Microambiente Tumoral , Humanos , Microambiente Tumoral/inmunología , Microambiente Tumoral/genética , Neoplasias/genética , Neoplasias/patología , Neoplasias/metabolismo , Regulación Neoplásica de la Expresión Génica , Inmunoterapia/métodos , Perfilación de la Expresión Génica , Interferones/metabolismo
15.
Curr Opin Immunol ; 87: 102427, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38781720

RESUMEN

The essential and redundant functions of human type I and II interferons (IFNs) have been delineated over the last three decades by studies of patients with inborn errors of immunity or their autoimmune phenocopies, but much less is known about type III IFNs. Patients with cells that do not respond to type III IFNs due to inherited IL10RB deficiency display no overt viral disease, and their inflammatory disease phenotypes can be explained by defective signaling via other interleukine10RB-dependent pathways. Moreover, patients with inherited deficiencies of interferon-stimulated gene factor 3 (ISGF-3) (STAT1, STAT2, IRF9) present viral diseases also seen in patients with inherited deficiencies of the type I IFN receptor (IFNAR1/2). Finally, patients with autoantibodies neutralizing type III IFNs have no obvious predisposition to viral disease. Current findings thus suggest that type III IFNs are largely redundant in humans. The essential functions of human type III IFNs, particularly in antiviral defenses, remain to be discovered.


Asunto(s)
Interferón lambda , Interferones , Virosis , Humanos , Interferones/metabolismo , Interferones/inmunología , Virosis/inmunología , Animales , Transducción de Señal/inmunología , Factor de Transcripción STAT2/metabolismo , Factor de Transcripción STAT2/genética , Factor de Transcripción STAT2/inmunología , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/genética , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/inmunología , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/inmunología , Subunidad beta del Receptor de Interleucina-10/genética , Subunidad beta del Receptor de Interleucina-10/inmunología , Subunidad beta del Receptor de Interleucina-10/metabolismo
16.
J Immunol ; 212(12): 1945-1957, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38700419

RESUMEN

The cytosolic detection of pathogen-derived nucleic acids has evolved as an essential strategy for host innate immune defense in mammals. One crucial component in this process is the stimulator of IFN genes (STING), which acts as a vital signaling adaptor, connecting the cytosolic detection of DNA by cyclic GMP-AMP (cGAMP) synthase (cGAS) to the downstream type I IFN signaling pathway. However, this process remains elusive in invertebrates. In this study, we present evidence demonstrating that STING, an ortholog found in a marine invertebrate (shrimp) called Litopenaeus vannamei, can directly detect DNA and initiate an IFN-like antiviral response. Unlike its homologs in other eukaryotic organisms, which exclusively function as sensors for cyclic dinucleotides, shrimp STING has the ability to bind to both double-stranded DNA and cyclic dinucleotides, including 2'3'-cGAMP. In vivo, shrimp STING can directly sense DNA nucleic acids from an infected virus, accelerate IFN regulatory factor dimerization and nuclear translocation, induce the expression of an IFN functional analog protein (Vago4), and finally establish an antiviral state. Taken together, our findings unveil a novel double-stranded DNA-STING-IKKε-IRF-Vago antiviral axis in an arthropod, providing valuable insights into the functional origins of DNA-sensing pathways in evolution.


Asunto(s)
Proteínas de la Membrana , Animales , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/inmunología , Penaeidae/inmunología , Penaeidae/virología , Inmunidad Innata/inmunología , Transducción de Señal/inmunología , Interferones/metabolismo , Interferones/inmunología , Nucleótidos Cíclicos/metabolismo , Nucleótidos Cíclicos/inmunología
17.
Fish Shellfish Immunol ; 150: 109611, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38734119

RESUMEN

During virus-host co-evolution, viruses have developed multiple strategies to dampen IFN response and prevent its antiviral activity in host cells. To date, the interactions between host IFN response and the immune evasion strategies exploited by fish iridoviruses still remain largely uncertain. Here, a potential immune evasion protein candidate of Singapore grouper iridovirus (SGIV), VP82 (encoded by SGIV ORF82) was screened and its roles during viral replication were investigated in detail. Firstly, VP82 overexpression dramatically decreased IFN or ISRE promoter activity and the transcription levels of IFN stimulated genes (ISGs) stimulated by grouper cyclic GMP-AMP synthase (EccGAS)/stimulator of interferon genes (EcSTING), TANK-binding kinase 1 (EcTBK1), IFN regulatory factor 3 (EcIRF3)and EcIRF7. Secondly, Co-IP assays indicated that VP82 interacted with EcIRF3 and EcIRF7, but not EcSTING and EcTBK1, which was consistent with the co-localization between VP82 and EcIRF3 or EcIRF7. Furthermore, VP82 promoted the degradation of EcIRF3 and EcIRF7 in a dose-dependent manner via the autophagy pathway. Finally, VP82 overexpression accelerated SGIV replication, evidenced by the increased transcriptions of viral core genes and viral production. Moreover, the antiviral action of EcIRF3 or EcIRF7 was significantly depressed in VP82 overexpressed cells. Together, VP82 was speculated to exert crucial roles for SGIV replication by inhibiting the IFN response via the degradation of IRF3 and IRF7. Our findings provided new insights into understanding the immune evasion strategies utilized by fish iridovirus through IFN regulation.


Asunto(s)
Infecciones por Virus ADN , Enfermedades de los Peces , Proteínas de Peces , Factor 3 Regulador del Interferón , Factor 7 Regulador del Interferón , Ranavirus , Proteínas Virales , Animales , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/inmunología , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Proteínas de Peces/metabolismo , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/virología , Infecciones por Virus ADN/inmunología , Infecciones por Virus ADN/veterinaria , Ranavirus/fisiología , Proteínas Virales/genética , Proteínas Virales/metabolismo , Inmunidad Innata/genética , Interferones/genética , Interferones/inmunología , Interferones/metabolismo , Evasión Inmune , Lubina/inmunología , Lubina/genética , Replicación Viral , Proteínas de Pez Cebra , Factores Reguladores del Interferón
18.
Curr Opin Immunol ; 87: 102425, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38763032

RESUMEN

Interferons (IFNs) are an integral component of the host innate immune response during viral infection. Recent advances in the study of type I and III IFNs suggest that though both types counteract viral infection, type III IFNs act predominantly at epithelial barrier sites, while type I IFNs drive systemic responses. The dynamics and specific roles of type I versus III IFNs have been studied in the context of infection by a variety of enteric pathogens, including reovirus, rotavirus, norovirus, astrovirus, and intestinal severe acute respiratory syndrome coronavirus 2, revealing shared patterns of regulatory influence. An important role for the gut microbiota, including the virome, in regulating homeostasis and priming of intestinal IFN responses has also recently emerged.


Asunto(s)
Tracto Gastrointestinal , Interacciones Huésped-Patógeno , Interferón lambda , Interferón Tipo I , Interferones , Humanos , Interferón Tipo I/metabolismo , Interferón Tipo I/inmunología , Interacciones Huésped-Patógeno/inmunología , Interferones/metabolismo , Interferones/inmunología , Animales , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/virología , Tracto Gastrointestinal/microbiología , Inmunidad Innata , Microbioma Gastrointestinal/inmunología , Virosis/inmunología
19.
Curr Opin Immunol ; 87: 102424, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38761566

RESUMEN

Type I and III interferons (IFN-I and IFN-III) have a central role in the early antimicrobial response against invading pathogens. Induction of IFN-Is and IFN-IIIs arises due to the sensing by pattern recognition receptors of pathogen-associated molecular patterns (from micro-organisms) or of damage-associated molecular patterns (DAMPs; produced by host cells). Here, we review recent developments on how IFN-I and IFN-III expression is stimulated by different pathogens and how the signalling pathways leading to IFN induction are tightly regulated. We also summarise the growing knowledge of the sensing pathways that lead to IFN-I and IFN-III induction in response to severe acute respiratory syndrome coronavirus 2.


Asunto(s)
COVID-19 , Interferón lambda , Interferón Tipo I , Interferones , SARS-CoV-2 , Transducción de Señal , Humanos , Interferón Tipo I/metabolismo , Interferón Tipo I/inmunología , Animales , Transducción de Señal/inmunología , SARS-CoV-2/inmunología , Interferones/metabolismo , Interferones/inmunología , COVID-19/inmunología , COVID-19/virología , Interacciones Huésped-Patógeno/inmunología , Receptores de Reconocimiento de Patrones/metabolismo , Receptores de Reconocimiento de Patrones/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Moléculas de Patrón Molecular Asociado a Patógenos/metabolismo
20.
Curr Opin Immunol ; 87: 102423, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38776716

RESUMEN

The host immune response against severe acute respiratory syndrome coronavirus 2 includes the induction of a group of natural antiviral cytokines called interferons (IFNs). Although originally recognized for their ability to potently counteract infections, the mechanistic functions of IFNs in patients with varying severities of coronavirus disease 2019 (COVID-19) have highlighted a more complex scenario. Cellular and molecular analyses have revealed that timing, location, and subtypes of IFNs produced during severe acute respiratory syndrome coronavirus 2 infection play a major role in determining disease progression and severity. In this review, we summarize what the COVID-19 pandemic has taught us about the protective and detrimental roles of IFNs during the inflammatory response elicited against a new respiratory virus across different ages and its longitudinal consequences in driving the development of long COVID-19.


Asunto(s)
COVID-19 , Interferones , SARS-CoV-2 , Humanos , COVID-19/inmunología , Interferones/inmunología , Interferones/metabolismo , SARS-CoV-2/inmunología , Pandemias , Animales , Yin-Yang
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA