Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
1.
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
2.
Viruses ; 14(10)2022 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-36298850

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is associated with vascular endothelial cell tumor, Kaposi's sarcoma (KS) and lymphoproliferative disorder, multicentric Castleman's disease (MCD), primary effusion lymphoma (PEL) and KSHV inflammatory cytokine syndrome (KICS). Dysregulation of proinflammatory cytokines is found in most KSHV associated diseases. However, little is known about the role of host microenvironment in the regulation of KSHV establishment in B cells. In the present study, we demonstrated that IFN-γ has a strong inhibitory effect on KSHV infection but only in a subset of tonsil-derived lymphocyte samples that are intrinsically more susceptible to infection, contain higher proportions of naïve B cells, and display increased levels of IRF1 and STAT1-pY701. The effect of IFN-γ in responsive samples was associated with increased frequencies of germinal center B cells (GCB) and decreased infection of plasma cells, suggesting that IFN-γ-mediated modulation of viral dynamics in GC can inhibit the establishment of KSHV infection.


Asunto(s)
Linfocitos B , Infecciones por Herpesviridae , Herpesvirus Humano 8 , Interferón gamma , Humanos , Síndrome de Inmunodeficiencia Adquirida/inmunología , Linfocitos B/inmunología , Enfermedad de Castleman/inmunología , Citocinas/inmunología , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/inmunología , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/inmunología , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/inmunología , Interferón gamma/inmunología , Sarcoma de Kaposi/inmunología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Susceptibilidad a Enfermedades/inmunología
3.
J Virol ; 96(17): e0077422, 2022 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-35972291

RESUMEN

XIAP-associated factor 1 (XAF1) is an interferon (IFN)-stimulated gene (ISG) that enhances IFN-induced apoptosis. However, it is unexplored whether XAF1 is essential for the host fighting against invaded viruses. Here, we find that XAF1 is significantly upregulated in the host cells infected with emerging RNA viruses, including influenza, Zika virus (ZIKV), and SARS-CoV-2. IFN regulatory factor 1 (IRF1), a key transcription factor in immune cells, determines the induction of XAF1 during antiviral immunity. Ectopic expression of XAF1 protects host cells against various RNA viruses independent of apoptosis. Knockout of XAF1 attenuates host antiviral innate immunity in vitro and in vivo, which leads to more severe lung injuries and higher mortality in the influenza infection mouse model. XAF1 stabilizes IRF1 protein by antagonizing the CHIP-mediated degradation of IRF1, thus inducing more antiviral IRF1 target genes, including DDX58, DDX60, MX1, and OAS2. Our study has described a protective role of XAF1 in the host antiviral innate immunity against RNA viruses. We have also elucidated the molecular mechanism that IRF1 and XAF1 form a positive feedback loop to induce rapid and robust antiviral immunity. IMPORTANCE Rapid and robust induction of antiviral genes is essential for the host to clear the invaded viruses. In addition to the IRF3/7-IFN-I-STAT1 signaling axis, the XAF1-IRF1 positive feedback loop synergistically or independently drives the transcription of antiviral genes. Moreover, XAF1 is a sensitive and reliable gene that positively correlates with the viral infection, suggesting that XAF1 is a potential diagnostic marker for viral infectious diseases. In addition to the antitumor role, our study has shown that XAF1 is essential for antiviral immunity. XAF1 is not only a proapoptotic ISG, but it also stabilizes the master transcription factor IRF1 to induce antiviral genes. IRF1 directly binds to the IRF-Es of its target gene promoters and drives their transcriptions, which suggests a unique role of the XAF1-IRF1 loop in antiviral innate immunity, particularly in the host defect of IFN-I signaling such as invertebrates.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Reguladoras de la Apoptosis , Factor 1 Regulador del Interferón , Infecciones por Virus ARN , Virus ARN , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/inmunología , Humanos , Inmunidad Innata , Factor 1 Regulador del Interferón/inmunología , Ratones , Ratones Noqueados , Infecciones por Virus ARN/inmunología , Replicación Viral
4.
Cells ; 11(3)2022 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-35159296

RESUMEN

The global outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is still ongoing, as is research on the molecular mechanisms underlying cellular infection by coronaviruses, with the hope of developing therapeutic agents against this pandemic. Other important respiratory viruses such as 2009 pandemic H1N1 and H7N9 avian influenza virus (AIV), influenza A viruses, are also responsible for a possible outbreak due to their respiratory susceptibility. However, the interaction of these viruses with host cells and the regulation of post-transcriptional genes remains unclear. In this study, we detected and analyzed the comparative transcriptome profiling of SARS-CoV-2, panH1N1 (A/California/07/2009), and H7N9 (A/Shanghai/1/2013) infected cells. The results showed that the commonly upregulated genes among the three groups were mainly involved in autophagy, pertussis, and tuberculosis, which indicated that autophagy plays an important role in viral pathogenicity. There are three groups of commonly downregulated genes involved in metabolic pathways. Notably, unlike panH1N1 and H7N9, SARS-CoV-2 infection can inhibit the m-TOR pathway and activate the p53 signaling pathway, which may be responsible for unique autophagy induction and cell apoptosis. Particularly, upregulated expression of IRF1 was found in SARS-CoV-2, panH1N1, and H7N9 infection. Further analysis showed SARS-CoV-2, panH1N1, and H7N9 infection-induced upregulation of lncRNA-34087.27 could serve as a competitive endogenous RNA to stabilize IRF1 mRNA by competitively binding with miR-302b-3p. This study provides new insights into the molecular mechanisms of influenza A virus and SARS-CoV-2 infection.


Asunto(s)
COVID-19/inmunología , Inmunidad/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H7N9 del Virus de la Influenza A/inmunología , Gripe Humana/inmunología , ARN/inmunología , Transcriptoma/inmunología , Células A549 , Animales , COVID-19/genética , COVID-19/virología , Células HEK293 , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad/genética , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H7N9 del Virus de la Influenza A/fisiología , Gripe Humana/genética , Gripe Humana/virología , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/inmunología , Factor 1 Regulador del Interferón/metabolismo , MicroARNs/genética , MicroARNs/inmunología , MicroARNs/metabolismo , Pandemias/prevención & control , ARN/genética , ARN/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/inmunología , ARN Largo no Codificante/metabolismo , ARN Mensajero/genética , ARN Mensajero/inmunología , ARN Mensajero/metabolismo , RNA-Seq/métodos , SARS-CoV-2/fisiología , Transducción de Señal/genética , Transducción de Señal/inmunología , Transcriptoma/genética
5.
Inflammation ; 45(1): 308-330, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34536158

RESUMEN

Immunosuppression developed by cancer cells remains a leading cause of treating failure of immunotherapies. This study aimed to explore the function of human endogenous retrovirus-H long terminal repeat-associating 2 (HHLA2), an immune checkpoint molecule from the B7 family, in the immune escape in hepatocellular carcinoma (HCC). Mouse models with primary HCC or with xenograft tumors were established. The portion of tumor-associated macrophages (TAMs) and the level of PD-L1 in the tumor tissues were examined. THP-1 cells were treated with PMA to obtain a macrophage-like phenotype. The PMA-treated THP-1 cells were co-cultured with the HCC cells in Transwell chambers to examine the function of HHLA2 in chemotactic migration and polarization of macrophages. HHLA2 expression was correlated with infiltration of immune cells, especially macrophages, and was linked to poor prognosis of patients with HCC. HHLA2 knockdown reduced incidence rate of primary HCC in mice. It also reduced tumor metastasis, the portion of M2 macrophages, and the expression of PD-L1 in primary and xenograft tumors. In vitro, HHLA2 upregulation increased expression of PD-L1 in HCC cells indirectly by inducing M2 polarization and chemotactic migration of macrophages. Interferon gamma (IFNG) enhanced expression of interferon regulatory factor 1 (IFR1) in HCC cells, and IFR1 bound to the promoter region of HHLA2 to activate HHLA2 expression. This study suggested that the IFNG/IFR1/HHLA2 axis in HCC induces M2 polarization and chemotactic migration of macrophages, which leads to immune escape and development of HCC.


Asunto(s)
Carcinoma Hepatocelular/inmunología , Inmunoglobulinas/inmunología , Factor 1 Regulador del Interferón/inmunología , Interferón gamma/inmunología , Neoplasias Hepáticas/inmunología , Escape del Tumor/inmunología , Animales , Antígeno B7-H1/inmunología , Antígeno B7-H1/metabolismo , Carcinoma Hepatocelular/metabolismo , Humanos , Inmunoglobulinas/metabolismo , Factor 1 Regulador del Interferón/metabolismo , Interferón gamma/metabolismo , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Células THP-1 , Activación Transcripcional/inmunología
6.
mBio ; 12(5): e0212121, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34607464

RESUMEN

Macrophages sense and respond to pathogens by induction of antimicrobial and inflammatory programs to alert other immune cells and eliminate the infectious threat. We have previously identified the transcription factor IRF1 to be consistently activated in macrophages during Mycobacterium avium infection, but its precise role during infection is not clear. Here, we show that tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6) autocrine/paracrine signaling contributes to controlling the intracellular growth of M. avium in human primary macrophages through activation of IRF1 nuclear translocation and expression of IRG1, a mitochondrial enzyme that produces the antimicrobial metabolite itaconate. Small interfering RNA (siRNA)-mediated knockdown of IRF1 or IRG1 increased the mycobacterial load, whereas exogenously provided itaconate was bacteriostatic at high concentrations. While the overall level of endogenous itaconate was low in M. avium-infected macrophages, the repositioning of mitochondria to M. avium phagosomes suggests a mechanism by which itaconate can be delivered directly to M. avium phagosomes in sufficient quantities to inhibit growth. Using mRNA hybridization, we further show that uninfected bystander cells actively contribute to the resolution of infection by producing IL-6 and TNF-α, which, via paracrine signaling, activate IRF1/IRG1 and strengthen the antimicrobial activity of infected macrophages. This mechanism contributes to the understanding of why patients on anti-inflammatory treatment, e.g., with tocilizumab or infliximab, can be more susceptible to mycobacterial disease. IMPORTANCE The prevalence of lung diseases caused by nontuberculous mycobacteria, such as Mycobacterium avium, is increasing in countries where tuberculosis is not endemic, most likely because of an aging population that is immunocompromised from underlying disease or immunosuppressive therapy. Our study contributes to the understanding of mycobacterial survival and killing in human macrophages and, more broadly, to the impact of immunometabolism during infection. We show evidence of an antimicrobial program in human primary macrophages where activation of the transcription factor IRF1 and expression of the mitochondrial enzyme IRG1 restrict the intracellular growth of M. avium, possibly by directed delivery of itaconate to M. avium phagosomes. The study also sheds light on why patients on immunosuppressive therapy are more susceptible to mycobacterial infections, since TNF-α and IL-6 contribute to driving the described antimycobacterial program.


Asunto(s)
Carboxiliasas/inmunología , Factor 1 Regulador del Interferón/inmunología , Interleucina-6/inmunología , Macrófagos/inmunología , Mycobacterium avium/fisiología , Tuberculosis/inmunología , Tuberculosis/microbiología , Factor de Necrosis Tumoral alfa/inmunología , Carboxiliasas/genética , Células Cultivadas , Humanos , Inmunidad Innata , Factor 1 Regulador del Interferón/genética , Macrófagos/microbiología , Mycobacterium avium/genética , Comunicación Paracrina , Tuberculosis/genética , Factor de Necrosis Tumoral alfa/genética
7.
Sci Immunol ; 6(61)2021 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-34244313

RESUMEN

Conventional type 1 dendritic cells (cDC1s) are critical for antitumor immunity. They acquire antigens from dying tumor cells and cross-present them to CD8+ T cells, promoting the expansion of tumor-specific cytotoxic T cells. However, the signaling pathways that govern the antitumor functions of cDC1s in immunogenic tumors are poorly understood. Using single-cell transcriptomics to examine the molecular pathways regulating intratumoral cDC1 maturation, we found nuclear factor κB (NF-κB) and interferon (IFN) pathways to be highly enriched in a subset of functionally mature cDC1s. We identified an NF-κB-dependent and IFN-γ-regulated gene network in cDC1s, including cytokines and chemokines specialized in the recruitment and activation of cytotoxic T cells. By mapping the trajectory of intratumoral cDC1 maturation, we demonstrated the dynamic reprogramming of tumor-infiltrating cDC1s by NF-κB and IFN signaling pathways. This maturation process was perturbed by specific inactivation of either NF-κB or IFN regulatory factor 1 (IRF1) in cDC1s, resulting in impaired expression of IFN-γ-responsive genes and consequently a failure to efficiently recruit and activate antitumoral CD8+ T cells. Last, we demonstrate the relevance of these findings to patients with melanoma, showing that activation of the NF-κB/IRF1 axis in association with cDC1s is linked with improved clinical outcome. The NF-κB/IRF1 axis in cDC1s may therefore represent an important focal point for the development of new diagnostic and therapeutic approaches to improve cancer immunotherapy.


Asunto(s)
Células Dendríticas/inmunología , Factor 1 Regulador del Interferón/inmunología , Melanoma/inmunología , FN-kappa B/inmunología , Neoplasias Cutáneas/inmunología , Animales , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Factor 1 Regulador del Interferón/genética , Interferón gamma/inmunología , Estimación de Kaplan-Meier , Masculino , Melanoma/genética , Melanoma/mortalidad , Ratones Transgénicos , FN-kappa B/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/mortalidad
8.
Front Immunol ; 12: 563336, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34248923

RESUMEN

The innate immune and host-protective responses to viruses, such as the airway pathogen human metapneumovirus (HMPV), depend on interferons (IFNs) that is induced through TANK-binding kinase 1 (TBK1) and IFN regulatory factors (IRFs). The transcription factor IRF1 is important for host resistance against several viruses and has a key role in induction of IFN-λ at mucosal surfaces. In most cell types IRF1 is expressed at very low levels, but its mRNA is rapidly induced when the demand for IRF1 activity arises. Despite general recognition of the importance of IRF1 to antiviral responses, the molecular mechanisms by which IRF1 is regulated during viral infections are not well understood. Here we identify the serine/threonine kinase TBK1 and IFN-ß as critical regulators of IRF1 mRNA and protein levels in human monocyte-derived macrophages. We find that inhibition of TBK1 activity either by the semi-selective TBK1/IKKε inhibitor BX795 or by siRNA-mediated knockdown abrogates HMPV-induced expression of IRF1. Moreover, we show that canonical NF-κB signaling is involved in IRF1 induction and that the TBK1/IKKε inhibitor BX795, but not siTBK1 treatment, impairs HMPV-induced phosphorylation of the NF-κB subunit p65. At later time-points of the infection, IRF1 expression depended heavily on IFN-ß-mediated signaling via the IFNAR-STAT1 pathway. Hence, our results suggest that TBK1 activation and TBK1/IKKε-mediated phosphorylation of the NF-κB subunit p65 control transcription of IRF1. Our study identifies a novel mechanism for IRF1 induction in response to viral infection of human macrophages that could be relevant not only to defense against HMPV, but also to other viral, bacterial and fungal pathogens.


Asunto(s)
Inmunidad Innata , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/inmunología , Interferón Tipo I/inmunología , Metapneumovirus/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Línea Celular , Células Cultivadas , Humanos , Interferón Tipo I/genética , Metapneumovirus/genética , Monocitos/inmunología , Monocitos/virología , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal
9.
Viruses ; 13(4)2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33807175

RESUMEN

Type I interferons (IFNs) are produced by most cells in response to virus infection and stimulate a program of anti-viral gene expression in neighboring cells to suppress virus replication. Type III IFNs have similar properties, however their effects are limited to epithelial cells at mucosal surfaces due to restricted expression of the type III IFN receptor. Rotavirus (RV) replicates in intestinal epithelial cells that respond predominantly to type III IFNs, and it has been shown that type III rather than type I IFNs are important for controlling RV infections in vivo. The RV NSP1 protein antagonizes the host type I IFN response by targeting IRF-3, IRF-5, IRF-7, or ß-TrCP for proteasome-mediated degradation in a strain-specific manner. Here we provide the first demonstration that NSP1 proteins from several human and animal RV strains antagonize type III as well as type I IFN induction. We also show that NSP1 is a potent inhibitor of IRF-1, a previously undescribed property of NSP1 which is conserved among human and animal RVs. Interestingly, all NSP1 proteins were substantially more effective inhibitors of IRF-1 than either IRF-3 or IRF-7 which has significance for evasion of basal anti-viral immunity and type III IFN induction in the intestinal epithelium.


Asunto(s)
Células Epiteliales/virología , Interferón Tipo I/antagonistas & inhibidores , Interferones/antagonistas & inhibidores , Rotavirus/inmunología , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Animales , Células Epiteliales/inmunología , Células HEK293 , Humanos , Factor 1 Regulador del Interferón/antagonistas & inhibidores , Factor 1 Regulador del Interferón/inmunología , Interferón Tipo I/inmunología , Interferones/inmunología , Intestinos/citología , Rotavirus/química , Rotavirus/aislamiento & purificación
10.
PLoS Pathog ; 17(1): e1009220, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33476326

RESUMEN

The eponymous member of the interferon regulatory factor (IRF) family, IRF1, was originally identified as a nuclear factor that binds and activates the promoters of type I interferon genes. However, subsequent studies using genetic knockouts or RNAi-mediated depletion of IRF1 provide a much broader view, linking IRF1 to a wide range of functions in protection against invading pathogens. Conserved throughout vertebrate evolution, IRF1 has been shown in recent years to mediate constitutive as well as inducible host defenses against a variety of viruses. Fine-tuning of these ancient IRF1-mediated host defenses, and countering strategies by pathogens to disarm IRF1, play crucial roles in pathogenesis and determining the outcome of infection.


Asunto(s)
Enfermedades Transmisibles/inmunología , Enfermedades Transmisibles/terapia , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/inmunología , Factor 1 Regulador del Interferón/metabolismo , Animales , Enfermedades Transmisibles/metabolismo , Humanos , Factor 1 Regulador del Interferón/inmunología
11.
Cell Rep ; 33(5): 108345, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33147460

RESUMEN

Bat cells and tissue have elevated basal expression levels of antiviral genes commonly associated with interferon alpha (IFNα) signaling. Here, we show Interferon Regulatory Factor 1 (IRF1), 3, and 7 levels are elevated in most bat tissues and that, basally, IRFs contribute to the expression of type I IFN ligands and high expression of interferon regulated genes (IRGs). CRISPR knockout (KO) of IRF 1/3/7 in cells reveals distinct subsets of genes affected by each IRF in an IFN-ligand signaling-dependent and largely independent manner. As the master regulators of innate immunity, the IRFs control the kinetics and maintenance of the IRG response and play essential roles in response to influenza A virus (IAV), herpes simplex virus 1 (HSV-1), Melaka virus/Pteropine orthoreovirus 3 Melaka (PRV3M), and Middle East respiratory syndrome-related coronavirus (MERS-CoV) infection. With its differential expression in bats compared to that in humans, this highlights a critical role for basal IRF expression in viral responses and potentially immune cell development in bats with relevance for IRF function in human biology.


Asunto(s)
Quirópteros/inmunología , Regulación de la Expresión Génica/inmunología , Factor 1 Regulador del Interferón/inmunología , Factor 7 Regulador del Interferón/inmunología , Virosis/inmunología , Animales , Herpesvirus Humano 1/inmunología , Virus de la Influenza A/inmunología , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , Orthoreovirus/inmunología
12.
Mol Immunol ; 128: 33-40, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33053462

RESUMEN

The bovine viral diarrhea virus (BVDV-1) is a pathogen with the capacity to modulate the interferon type I system. To further investigate the effects of BVDV-1 on the production of the immune response, the Madin-Darby bovine kidney cell line was infected with the cytopathic CH001 field isolate of BVDV-1, and the IFNbeta expression profiles were analyzed. The results showed that cpBVDV-1 was able to induce the production of IFNbeta in a way similar to polyinosinic-polycytidylic acid, but with less intensity. Interestingly, all cpBVDV-1 activities were blocked by pharmacological inhibitors of the IRF-1, IRF-7, and NF-κB signaling pathway, and the level of IFNbeta decreased at the level of transcript and protein. These results, together with in silico analyses showing the presence of several regulatory consensus target motifs, suggest that cpBVDV-1 regulates IFNbeta expression in bovines through the activation of several key transcription factors. Collectively, the results suggest that during cpBVDV-1 infection, cross talk is evident between various signaling pathways involved in transcriptional activation of IFNbeta in cattle.


Asunto(s)
Diarrea Mucosa Bovina Viral/genética , Virus de la Diarrea Viral Bovina Tipo 1/inmunología , Regulación de la Expresión Génica/genética , Expresión Génica/genética , Factor 1 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/genética , FN-kappa B/genética , Animales , Diarrea Mucosa Bovina Viral/virología , Bovinos , Línea Celular , Células Epiteliales/inmunología , Células Epiteliales/virología , Expresión Génica/inmunología , Regulación de la Expresión Génica/inmunología , Factor 1 Regulador del Interferón/inmunología , Factor 7 Regulador del Interferón/inmunología , FN-kappa B/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología
13.
J Virol ; 94(22)2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-32878885

RESUMEN

Innate immunity is an essential way for host cells to resist viral infection through the production of interferons (IFNs) and proinflammatory cytokines. Interferon regulatory factor 3 (IRF3) plays a critical role in the innate immune response to viral infection. However, the role of IRF1 in innate immunity remains largely unknown. In this study, we found that IRF1 is upregulated through the IFN/JAK/STAT signaling pathway upon viral infection. The silencing of IRF1 attenuates the innate immune response to viral infection. IRF1 interacts with IRF3 and augments the activation of IRF3 by blocking the interaction between IRF3 and protein phosphatase 2A (PP2A). The DNA binding domain (DBD) of IRF1 is the key functional domain for its interaction with IRF3. Overall, our study reveals a novel mechanism by which IRF1 promotes the innate immune response to viral infection by enhancing the activation of IRF3, thereby inhibiting viral infection.IMPORTANCE The activation of innate immunity is essential for host cells to restrict the spread of invading viruses and other pathogens. IRF3 plays a critical role in the innate immune response to RNA viral infection. However, whether IRF1 plays a role in innate immunity is unclear. In this study, we demonstrated that IRF1 promotes the innate immune response to viral infection. IRF1 is induced by viral infection. Notably, IRF1 targets and augments the phosphorylation of IRF3 by blocking the interaction between IRF3 and PP2A, leading to the upregulation of innate immunity. Collectively, the results of our study provide new insight into the regulatory mechanism of IFN signaling and uncover the role of IRF1 in the positive regulation of the innate immune response to viral infection.


Asunto(s)
Inmunidad Innata/inmunología , Factor 1 Regulador del Interferón/inmunología , Factor 3 Regulador del Interferón/metabolismo , Virosis/inmunología , Línea Celular , Proteínas de Unión al ADN , Células HEK293 , Humanos , Factor 1 Regulador del Interferón/metabolismo , Fosforilación , Infecciones por Virus ARN/inmunología , Virus ARN , Transducción de Señal/inmunología
14.
Medicine (Baltimore) ; 99(37): e22170, 2020 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-32925784

RESUMEN

BACKGROUND: Evidence suggests that metastasis is chiefly responsible for the poor prognosis of colon adenocarcinoma (COAD). The tumor microenvironment plays a vital role in regulating this biological process. However, the mechanisms involved remain unclear. The aim of this study was to identify crucial metastasis-related biomarkers in the tumor microenvironment and investigate its association with tumor-infiltrating immune cells. METHODS: We obtained gene expression profiles and clinical information from The Cancer Genome Atlas database. According to the "Estimation of STromal and Immune cells in MAlignant Tumor tissue using Expression data" algorithm, each sample generated the immune and stromal scores. Following correlation analysis, the metastasis-related gene was identified in The Cancer Genome Atlas database and validated in the GSE40967 dataset from Gene Expression Omnibus. The correlation between metastasis-related gene and infiltrating immune cells was assessed using the Tumor IMmune Estimation Resource database. RESULTS: The analysis included 332 patients; the metastatic COAD samples showed a low immune score. Correlation analysis results showed that interferon regulatory factor 1 (IRF1) was associated with tumor stage, lymph node metastasis, and distant metastasis. Furthermore, significant associations between IRF1 and CD8+ T cells, T cell (general), dendritic cells, T-helper 1 cells, and T cell exhaustion were demonstrated by Spearmans correlation coefficients and P values. CONCLUSIONS: The present findings suggest that IRF1 is associated with metastasis and the degree of immune infiltration of CD8+ T cells (general), dendritic cells, T-helper 1 cells, and T cell exhaustion in COAD. These results may provide information for immunotherapy in colon cancer.


Asunto(s)
Adenocarcinoma/inmunología , Adenocarcinoma/patología , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Factor 1 Regulador del Interferón/inmunología , Linfocitos T/inmunología , Biomarcadores de Tumor , Linfocitos T CD8-positivos/metabolismo , Bases de Datos Genéticas , Células Dendríticas/metabolismo , Perfilación de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Metástasis Linfática/inmunología , Estadificación de Neoplasias , Pronóstico , Microambiente Tumoral
15.
Biochem Biophys Res Commun ; 533(3): 313-318, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-32958247

RESUMEN

Formyl peptide receptors (FPRs) are mainly expressed on leucocytes and sense microbe-associated molecular pattern (MAMP) molecules, thereby regulating leukocyte chemotaxis and activation. The formyl peptide receptor 2 (FPR2) selective agonist WKYMVm (Trp-Lys-Met-Val-D-Met) has shown potent pro-angiogenic, anti-inflammatory, and anti-apoptotic properties. In this study, we investigated whether WKYMVm exhibits bactericidal activity during neutrophil accumulation in acute lung injury (ALI) in mice and determined its cellular signaling pathways in HL-60 neutrophil-like cells. A daily intraperitoneal treatment of ALI mice with WKYMVm (2.5- and 5 mg/kg/d) daily over four days decreased the levels of proinflammatory cytokines TNF-α, IL-6, and IL-1ß, while it increased the MPO and NO release by differentiated HL-60 neutrophil-like cells. The IRF1 level and STAT1 phosphorylation at S727 were increased in the lungs of mice with ALI treated with WKYMVm. Lung histology induced by ALI was unaffected by treatment with WKYMVm. In vitro, WKYMVm increased MPO, NO, and SOD activity, as well as IRF1 and STAT1 phosphorylation at Ser727. Taken together, our data suggest therapeutic potential of WKYMVm, via FPR2-dependent regulation of STAT1/IRF1, in ALI.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Antiinflamatorios no Esteroideos/farmacología , Péptidos Catiónicos Antimicrobianos/inmunología , Factor 1 Regulador del Interferón/genética , Oligopéptidos/farmacología , Factor de Transcripción STAT1/genética , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/inmunología , Animales , Péptidos Catiónicos Antimicrobianos/metabolismo , Regulación de la Expresión Génica , Células HL-60 , Humanos , Factor 1 Regulador del Interferón/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/farmacología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Neutrófilos/metabolismo , Peroxidasa/genética , Peroxidasa/inmunología , Fosforilación , Factor de Transcripción STAT1/inmunología , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
16.
Vet Microbiol ; 247: 108785, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32768229

RESUMEN

Porcine deltacoronavirus (PDCoV) is a novel swine enteropathogenic coronavirus that causes watery diarrhea, vomiting and mortality in nursing piglets. Type III interferons (IFN-λs) are the major antiviral cytokines in intestinal epithelial cells, the target cells in vivo for PDCoV. In this study, we found that PDCoV infection remarkably inhibited Sendai virus-induced IFN-λ1 production by suppressing transcription factors IRF and NF-κB in IPI-2I cells, a line of porcine intestinal mucosal epithelial cells. We also confirmed that PDCoV infection impeded the activation of IFN-λ1 promoter stimulated by RIG-I, MDA5 and MAVS, but not by TBK1 and IRF1. Although the expression levels of IRF1 and MAVS were not changed, PDCoV infection resulted in reduction of the number of peroxisomes, the platform for MAVS to activate IRF1, and subsequent type III IFN production. Taken together, our study demonstrates that PDCoV suppresses type III IFN responses to circumvent the host's antiviral immunity.


Asunto(s)
Infecciones por Coronavirus/veterinaria , Células Epiteliales/inmunología , Células Epiteliales/virología , Interacciones Huésped-Patógeno/inmunología , Interferones/antagonistas & inhibidores , Animales , Línea Celular , Coronavirus , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Factor 1 Regulador del Interferón/antagonistas & inhibidores , Factor 1 Regulador del Interferón/inmunología , Interferones/inmunología , Intestinos/citología , Intestinos/virología , Riñón/citología , Riñón/virología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/inmunología , Virus Sendai/inmunología , Transducción de Señal/inmunología , Porcinos/virología , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/virología , Interferón lambda
17.
Elife ; 92020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32539931

RESUMEN

Counteracting innate immunity is essential for successful viral replication. Host cyclophilins (Cyps) have been implicated in viral evasion of host antiviral responses, although the mechanisms are still unclear. Here, we show that hepatitis C virus (HCV) co-opts the host protein CypA to aid evasion of antiviral responses dependent on the effector protein kinase R (PKR). Pharmacological inhibition of CypA rescues PKR from antagonism by HCV NS5A, leading to activation of an interferon regulatory factor-1 (IRF1)-driven cell intrinsic antiviral program that inhibits viral replication. These findings further the understanding of the complexity of Cyp-virus interactions, provide mechanistic insight into the remarkably broad antiviral spectrum of Cyp inhibitors, and uncover novel aspects of PKR activity and regulation. Collectively, our study identifies a novel antiviral mechanism that harnesses cellular antiviral immunity to suppress viral replication.


Asunto(s)
Ciclofilina A/antagonistas & inhibidores , Hepacivirus/fisiología , Factor 1 Regulador del Interferón/inmunología , Proteínas no Estructurales Virales/metabolismo , Replicación Viral , eIF-2 Quinasa/genética , Línea Celular Tumoral , Ciclofilina A/inmunología , Humanos , eIF-2 Quinasa/inmunología
18.
Eur J Immunol ; 50(9): 1321-1334, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32270872

RESUMEN

SAMHD1 is an enzyme with phosphohydrolase activity. Mutations in SAMHD1 have been linked to the development of Aicardi-Goutières syndrome in humans. This enzyme also has the capacity to restrict HIV virus replication in macrophages. Here, we report that Samhd1 is highly expressed in murine macrophages and is regulated by proinflammatory (IFN-γ and LPS) but not by anti-inflammatory (IL-4 or IL-10) activators. The induction of Samhd1 follows the pattern of an intermediate gene that requires protein synthesis. In transient transfection experiments using the Samhd1 promoter, we found that a fragment of 27 bps of this gene, falling between -937 and -910 bps relative to the transcription start site, is required for IFN-γ-dependent activation. Using EMSAs, we determined that IFN-γ treatment led to the elimination of a protein complex. Chromatin immunoprecipitation assays and siRNA experiments revealed that IRF1 is required for IFN-γ- or LPS-induced Samhd1 expression. Therefore, our results indicate that Samhd1 is stimulated by proinflammatory agents IFN-γ and LPS. Moreover, they reveal that these two agents, via IRF1, eliminate a protein complex that may be related to a repressor, thereby, triggering Samhd1 expression.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Factor 1 Regulador del Interferón/metabolismo , Interferón gamma/inmunología , Macrófagos/inmunología , Proteína 1 que Contiene Dominios SAM y HD/metabolismo , Animales , Factor 1 Regulador del Interferón/inmunología , Interferón gamma/farmacología , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteína 1 que Contiene Dominios SAM y HD/inmunología
19.
PLoS Pathog ; 16(3): e1008435, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32210480

RESUMEN

A striking feature of human visceral leishmaniasis (VL) is chronic inflammation in the spleen and liver, and VL patients present increased production levels of multiple inflammatory mediators, which contribute to tissue damage and disease severity. Here, we combined an experimental model with the transcriptional profile of human VL to demonstrate that the TLR4-IFN-ß pathway regulates the chronic inflammatory process and is associated with the asymptomatic form of the disease. Tlr4-deficient mice harbored fewer parasites in their spleen and liver than wild-type mice. TLR4 deficiency enhanced the Th1 immune response against the parasite, which was correlated with an increased activation of dendritic cells (DCs). Gene expression analyses demonstrated that IRF1 and IFN-ß were expressed downstream of TLR4 after infection. Accordingly, IRF1- and IFNAR-deficient mice harbored fewer parasites in the target organs than wild-type mice due to having an increased Th1 immune response. However, the absence of TLR4 or IFNAR increased the serum transaminase levels in infected mice, indicating the presence of liver damage in these animals. In addition, IFN-ß limits IFN-γ production by acting directly on Th1 cells. Using RNA sequencing analysis of human samples, we demonstrated that the transcriptional signature for the TLR4 and type I IFN (IFN-I) pathways was positively modulated in asymptomatic subjects compared with VL patients and thus provide direct evidence demonstrating that the TLR4-IFN-I pathway is related to the nondevelopment of the disease. In conclusion, our results demonstrate that the TLR4-IRF1 pathway culminates in IFN-ß production as a mechanism for dampening the chronic inflammatory process and preventing immunopathology development.


Asunto(s)
Factor 1 Regulador del Interferón/inmunología , Interferón beta/inmunología , Leishmania infantum/inmunología , Leishmaniasis Visceral/inmunología , Células TH1/inmunología , Receptor Toll-Like 4/inmunología , Animales , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Factor 1 Regulador del Interferón/genética , Interferón beta/genética , Leishmaniasis Visceral/genética , Leishmaniasis Visceral/patología , Ratones , Ratones Noqueados , Células TH1/patología , Receptor Toll-Like 4/genética
20.
J Virol ; 94(11)2020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32213613

RESUMEN

Interferon alpha (IFN-α) and IFN-ß are type I IFNs that are induced by virus infection and are important in the host's innate antiviral response. EBV infection activates multiple cell signaling pathways, resulting in the production of type I IFN which inhibits EBV infection and virus-induced B-cell transformation. We reported previously that EBV tegument protein BGLF2 activates p38 and enhances EBV reactivation. To further understand the role of BGLF2 in EBV infection, we used mass spectrometry to identify cellular proteins that interact with BGLF2. We found that BGLF2 binds to Tyk2 and confirmed this interaction by coimmunoprecipitation. BGLF2 blocked type I IFN-induced Tyk2, STAT1, and STAT3 phosphorylation and the expression of IFN-stimulated genes (ISGs) IRF1, IRF7, and MxA. In contrast, BGLF2 did not inhibit STAT1 phosphorylation induced by IFN-γ. Deletion of the carboxyl-terminal 66 amino acids of BGLF2 reduced the ability of the protein to repress type I IFN signaling. Treatment of gastric carcinoma and Raji cells with IFN-α blocked BZLF1 expression and EBV reactivation; however, expression of BGLF2 reduced the ability of IFN-α to inhibit BZLF1 expression and enhanced EBV reactivation. In summary, EBV BGLF2 interacts with Tyk2, inhibiting Tyk2, STAT1, and STAT3 phosphorylation and impairs type I IFN signaling; BGLF2 also counteracts the ability of IFN-α to suppress EBV reactivation.IMPORTANCE Type I interferons are important for controlling virus infection. We have found that the Epstein-Barr virus (EBV) BGLF2 tegument protein binds to a protein in the type I interferon signaling pathway Tyk2 and inhibits the expression of genes induced by type I interferons. Treatment of EBV-infected cells with type I interferon inhibits reactivation of the virus, while expression of EBV BGLF2 reduces the ability of type I interferon to inhibit virus reactivation. Thus, a tegument protein delivered to cells during virus infection inhibits the host's antiviral response and promotes virus reactivation of latently infected cells. Therefore, EBV BGLF2 might protect virus-infected cells from the type I interferon response in cells undergoing lytic virus replication.


Asunto(s)
Infecciones por Virus de Epstein-Barr/inmunología , Herpesvirus Humano 4/fisiología , Interferón Tipo I/inmunología , Transducción de Señal/inmunología , Proteínas Virales de Fusión/inmunología , Activación Viral/inmunología , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/patología , Células HEK293 , Humanos , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/inmunología , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/inmunología , Interferón Tipo I/genética , Interferón gamma/genética , Interferón gamma/inmunología , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología , Transducción de Señal/genética , TYK2 Quinasa/genética , TYK2 Quinasa/inmunología , Proteínas Virales de Fusión/genética , Activación Viral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...