Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 866
Filtrar
1.
Front Biosci (Landmark Ed) ; 29(7): 247, 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-39082331

RESUMEN

BACKGROUND: Retinal pigment epithelial (RPE) cells have a pivotal function in preserving the equilibrium of the retina and moderating the immunological interaction between the choroid and the retina. This study primarily focuses on delineating the protective effect offered by Kaempferol (Kae) against RPE cell damage. METHODS: Bioinformatics analysis was performed on the GSE30719 dataset to identify hub genes associated with RPE. Subsequently, we analyzed the impact of Kae on RPE apoptosis, cell viability, and inflammatory response through cell experiments, and explored the interaction between hub genes and Kae. RESULTS: Based on the GSE30719 dataset, nine hub genes (ISG15, IFIT1, IFIT3, STAT1, OASL, RSAD2, IRF7, MX2, and MX1) were identified, all of which were highly expressed in the GSE30719 case group. Kae could boost the proliferative activity of RPE cells caused by lipopolysaccharide (LPS), as well as reduce apoptosis and the generation of inflammatory factors (tumor necrosis factor receptor (TNFR), interleukin-1beta (IL-1ß)) and cytokines (IL-1, IL-6, IL-12). STAT1 was shown to inhibit cell proliferation, promote apoptosis, and secrete IL-1/IL-6/IL-12 in LPS-induced RPE cells. Moreover, IRF7 was found to interact with STAT1 in LPS-induced RPE cells, and STAT1 could maintain IRF7 levels through deubiquitination. In addition, we also found that the protective effect of Kae on LPS-induced RPE cell injury was mediated through STAT1/IRF7 axis. CONCLUSION: This study provided evidence that Kae protects RPE cells via regulating the STAT1/IRF7 signaling pathways, indicating its potential therapeutic relevance in the diagnosis and management of retinal disorders linked with RPE cell damage.


Asunto(s)
Apoptosis , Factor 7 Regulador del Interferón , Quempferoles , Epitelio Pigmentado de la Retina , Factor de Transcripción STAT1 , Ubiquitinación , Humanos , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/efectos de los fármacos , Factor de Transcripción STAT1/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/genética , Ubiquitinación/efectos de los fármacos , Apoptosis/efectos de los fármacos , Quempferoles/farmacología , Línea Celular , Proliferación Celular/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Lipopolisacáridos , Proteolisis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Citocinas/metabolismo , Citocinas/genética
2.
Biomed Pharmacother ; 177: 116885, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38878633

RESUMEN

Sjögren's syndrome (SS) is an autoimmune disease in which the salivary glands (SGs) and the lacrimal glands (LGs) are affected by lymphocytic infiltration and inflammation. It has been reported that interferon-α (IFN-α) released by plasmacytoid dendritic cells (pDCs) contribute to the pathology of SS, and ART has been shown to effectively ameliorates SS. Despite the current research endeavors, the mechanism of how ART works in the treatment of SS remains to be fully elucidated. Whether ART can treat SS by inhibiting IFN-α remains unclear. This hypothesis was tested both in vivo and in vitro settings during the study. The SS model mice, which were treated with ART, showed amelioration in symptoms related to dryness. RNA-seq analysis revealed strong anti-IFN-α signaling response upon ART treatment. Additional in vitro studies provided further confirmation that the application of ART inhibits the MyD88 protein expression and the nuclear translocation of IRF7. This suggests that the intervention of ART in the TLR-MyD88-IRF7 pathway plays a role in the therapeutic approach for SS. In summary, this study highlighted the therapeutic potential of ART in SS and ART inhibited the IFN-α signaling in pDCs via the TLR-MyD88-IRF7 pathway.


Asunto(s)
Artesunato , Células Dendríticas , Factor 7 Regulador del Interferón , Interferón-alfa , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide , Transducción de Señal , Síndrome de Sjögren , Animales , Síndrome de Sjögren/tratamiento farmacológico , Síndrome de Sjögren/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Interferón-alfa/metabolismo , Interferón-alfa/farmacología , Transducción de Señal/efectos de los fármacos , Factor 7 Regulador del Interferón/metabolismo , Ratones , Artesunato/farmacología , Artesunato/uso terapéutico , Receptores Toll-Like/metabolismo , Femenino , Modelos Animales de Enfermedad
3.
J Immunol ; 212(12): 1932-1944, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38709167

RESUMEN

IFN regulatory factor 7 (IRF7) exerts anti-infective effects by promoting the production of IFNs in various bacterial and viral infections, but its role in highly morbid and fatal Candida albicans infections is unknown. We unexpectedly found that Irf7 gene expression levels were significantly upregulated in tissues or cells after C. albicans infection in humans and mice and that IRF7 actually exacerbates C. albicans infection in mice independent of its classical function in inducing IFNs production. Compared to controls, Irf7-/- mice showed stronger phagocytosis of fungus, upregulation of C-type lectin receptor CD209 expression, and enhanced P53-AMPK-mTOR-mediated autophagic signaling in macrophages after C. albicans infection. The administration of the CD209-neutralizing Ab significantly hindered the phagocytosis of Irf7-/- mouse macrophages, whereas the inhibition of p53 or autophagy impaired the killing function of these macrophages. Thus, IRF7 exacerbates C. albicans infection by compromising the phagocytosis and killing capacity of macrophages via regulating CD209 expression and p53-AMPK-mTOR-mediated autophagy, respectively. This finding reveals a novel function of IRF7 independent of its canonical IFNs production and its unexpected role in enhancing fungal infections, thus providing more specific and effective targets for antifungal therapy.


Asunto(s)
Autofagia , Candida albicans , Candidiasis , Factor 7 Regulador del Interferón , Lectinas Tipo C , Macrófagos , Ratones Noqueados , Fagocitosis , Receptores de Superficie Celular , Serina-Treonina Quinasas TOR , Animales , Ratones , Fagocitosis/inmunología , Autofagia/inmunología , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Candidiasis/inmunología , Candida albicans/inmunología , Candida albicans/fisiología , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/inmunología , Macrófagos/inmunología , Humanos , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Ratones Endogámicos C57BL , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Transducción de Señal/inmunología
4.
Exp Dermatol ; 33(5): e15083, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38794808

RESUMEN

Interferons (IFNs) are signalling proteins primarily involved in initiating innate immune responses against pathogens and promoting the maturation of immune cells. Interferon Regulatory Factor 7 (IRF7) plays a pivotal role in the IFNs signalling pathway. The activation process of IRF7 is incited by exogenous or abnormal nucleic acids, which is followed by the identification via pattern recognition receptors (PRRs) and the ensuing signalling cascades. Upon activation, IRF7 modulates the expression of both IFNs and inflammatory gene regulation. As a multifunctional transcription factor, IRF7 is mainly expressed in immune cells, yet its presence is also detected in keratinocytes, fibroblasts, and various dermal cell types. In these cells, IRF7 is critical for skin immunity, inflammation, and fibrosis. IRF7 dysregulation may lead to autoimmune and inflammatory skin conditions, including systemic scleroderma (SSc), systemic lupus erythematosus (SLE), Atopic dermatitis (AD) and Psoriasis. This comprehensive review aims to extensively elucidate the role of IRF7 and its signalling pathways in immune cells and keratinocytes, highlighting its significance in skin-related and connective tissue diseases.


Asunto(s)
Enfermedades del Tejido Conjuntivo , Factor 7 Regulador del Interferón , Queratinocitos , Transducción de Señal , Enfermedades de la Piel , Humanos , Factor 7 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/genética , Enfermedades de la Piel/inmunología , Enfermedades de la Piel/metabolismo , Queratinocitos/metabolismo , Queratinocitos/inmunología , Enfermedades del Tejido Conjuntivo/metabolismo , Enfermedades del Tejido Conjuntivo/inmunología , Psoriasis/inmunología , Psoriasis/metabolismo , Animales , Piel/metabolismo , Piel/inmunología , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/inmunología , Esclerodermia Sistémica/genética , Inmunidad Innata
5.
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
6.
Cancer Lett ; 596: 216988, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38797234

RESUMEN

Type I interferons exhibit anti-proliferative and anti-cancer activities, but their detailed regulatory mechanisms in cancer have not been fully elucidated yet. RNA binding proteins are master orchestrators of gene regulation, which are closely related to tumor progression. Here we show that the upregulated RNA binding protein RBM45 correlates with poor prognosis in breast cancer. Depletion of RBM45 suppresses breast cancer progression both in cultured cells and xenograft mouse models. Mechanistically, RBM45 ablation inhibits breast cancer progression through regulating type I interferon signaling, particularly by elevating IFN-ß production. Importantly, RBM45 recruits TRIM28 to IRF7 and stimulates its SUMOylation, thereby repressing IFNB1 transcription. Loss of RBM45 reduced the SUMOylation of IRF7 by reducing the interaction between TRIM28 and IRF7 to promote IFNB1 transcription, leading to the inhibition of breast cancer progression. Taken together, our finding uncovers a vital role of RBM45 in modulating type I interferon signaling and cancer aggressive progression, implicating RBM45 as a potential therapeutic target in breast cancer.


Asunto(s)
Neoplasias de la Mama , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Factor 7 Regulador del Interferón , Proteínas de Unión al ARN , Sumoilación , Humanos , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Femenino , Animales , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/metabolismo , Ratones , Transcripción Genética , Línea Celular Tumoral , Interferón beta/metabolismo , Interferón beta/genética , Transducción de Señal , Ratones Desnudos , Proliferación Celular , Proteína 28 que Contiene Motivos Tripartito/metabolismo , Proteína 28 que Contiene Motivos Tripartito/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Endogámicos BALB C
7.
Brain Behav Immun ; 120: 10-20, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38777286

RESUMEN

BACKGROUND: It is known that nerve signals arising from sites of inflammation lead to persistent changes in the spinal cord and contribute to the amplification and persistence of pain. Nevertheless, the underlying mechanisms have not yet been completely elucidated. We identified differentially expressed genes in the lumbar (L4-L6) segment of the spinal cord from complete Freund's adjuvant (CFA) rats compared to control animals via high throughput sequencing. Based on differential gene expression analysis, we selected interferon regulatory factor 7 (IRF7) for follow-up experiments to explore its antinociceptive potential. METHODS: An animal model of inflammatory pain was induced by intraplantar injection of CFA. We evaluated the effects of adeno-associated viral (AAV)-mediated overexpression of IRF7 in the spinal cord on pain-related behavior after CFA injection. Moreover, the activation of the nuclear factor-κB (NF-κB) and the expression of inflammatory cytokines were investigated to understand the underlying mechanisms related to the contribution of IRF7 to inflammatory pain. RESULTS: CFA intraplantar injection caused a significant decrease in the level of spinal IRF7, which is mainly expressed in the dorsal horn neurons and astrocytes. Moreover, IRF7 overexpression significantly attenuated pain-related behaviors, as well as the activity of NF-κB/p65 and the production of interleukin-1ß (IL-1ß), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α) in the spinal cord of CFA rats. CONCLUSIONS: Our data indicated that spinal IRF7 plays an important role in the regulation of inflammatory pain. Thus, IRF7 overexpression at the spinal cord level might represent a potential target for the treatment of inflammatory pain.


Asunto(s)
Citocinas , Adyuvante de Freund , Inflamación , Factor 7 Regulador del Interferón , FN-kappa B , Dolor , Ratas Sprague-Dawley , Médula Espinal , Animales , Ratas , Factor 7 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/genética , Citocinas/metabolismo , Inflamación/metabolismo , Masculino , FN-kappa B/metabolismo , Médula Espinal/metabolismo , Dolor/metabolismo , Modelos Animales de Enfermedad
8.
Dev Comp Immunol ; 156: 105181, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38636698

RESUMEN

Interferon regulatory factor 7 (IRF7) is considered the master regulator of virus-induced interferon (IFN) production. However, to avoid an autoimmune response, the expression of IRF7 must be tightly controlled. In this study, we report that zebrafish ubiquitin-specific protease 8 (USP8) promotes IRF7 degradation through an autophagy-lysosome-dependent pathway to inhibit IFN production. First, zebrafish usp8 is induced upon spring viremia of carp virus (SVCV) infection and polyinosinic/polycytidylic acid (poly I:C) stimulation. Second, overexpression of USP8 suppresses SVCV or poly I:C-mediated IFN expression. Mechanistically, USP8 interacts with IRF7 and promotes its degradation via an autophagy-lysosome-dependent pathway. Finally, USP8 significantly suppresses cellular antiviral responses and enhances SVCV proliferation. In summary, our discoveries offer a perspective on the role of zebrafish USP8 and provide additional understanding of the regulation of IRF7 in host antiviral immune response.


Asunto(s)
Autofagia , Factor 7 Regulador del Interferón , Factores Reguladores del Interferón , Lisosomas , Rhabdoviridae , Proteínas de Pez Cebra , Pez Cebra , Animales , Pez Cebra/inmunología , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Autofagia/inmunología , Lisosomas/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/genética , Rhabdoviridae/fisiología , Rhabdoviridae/inmunología , Interferones/metabolismo , Poli I-C/inmunología , Infecciones por Rhabdoviridae/inmunología , Proteolisis , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/virología , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina Tiolesterasa/genética , Humanos , Inmunidad Innata
9.
J Infect ; 88(5): 106151, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38582127

RESUMEN

BACKGROUND: Immunological non-responders (INRs) among people living with HIV have inherently higher mortality and morbidity rates. The underlying immunological mechanisms whereby failure of immune reconstitution occurs in INRs require elucidation. METHOD: HIV-1 DNA and HIV-1 cell-associated RNA (CA-HIV RNA) quantifications were conducted via RT-qPCR. Transcriptome sequencing (RNA-seq), bioinformatics, and biological verifications were performed to discern the crosstalk between host and viral factors. Flow cytometry was employed to analyze cellular activation, proliferation, and death. RESULTS: HIV-1 DNA and CA-HIV RNA levels were observed to be significantly higher in INRs compared to immunological responders (IRs). Evaluation of CD4/CD8 ratios showed a significantly negative correlation with HIV-1 DNA in IRs, but not in INRs. Bioinformatics analyses and biological verifications showed IRF7/INF-α regulated antiviral response was intensified in INRs. PBMCs of INRs expressed significantly more HIV integrase-mRNA (p31) than IRs. Resting (CD4+CD69- T-cells) and activated (CD4+CD69+ T-cells) HIV-1 reservoir harboring cells were significantly higher in INRs, with the co-occurrence of significantly higher cellular proliferation and cell death in CD4+ T-cells of INRs. CONCLUSION: In INRs, the systematic crosstalk between the HIV-1 reservoir and host cells tends to maintain a persistent antiviral response-associated inflammatory environment, which drives aberrant cellular activation, proliferation, and death of CD4+ T-cells.


Asunto(s)
Proliferación Celular , Infecciones por VIH , VIH-1 , Factor 7 Regulador del Interferón , Humanos , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/genética , VIH-1/fisiología , Transcriptoma , Muerte Celular , Masculino , ARN Viral , Homeostasis , Adulto , ADN Viral/genética , Femenino , Linfocitos T CD4-Positivos/inmunología , Persona de Mediana Edad , Linfocitos T/inmunología , Activación de Linfocitos , Relación CD4-CD8 , Carga Viral
10.
Biochem Biophys Res Commun ; 712-713: 149915, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38663038

RESUMEN

Viral infections pose a significant threat to public health, and the production of interferons represents one of the most critical antiviral innate immune responses of the host. Consequently, the screening and identification of compounds or reagents that induce interferon production are of paramount importance. This study commenced with the cultivation of host bacterium 15,597, followed by the infection of Escherichia coli with the MS2 bacteriophage. Utilizing the J2 capture technique, a class of dsRNA mixtures (MS2+15,597) was isolated from the E. coli infected with the MS2 bacteriophage. Subsequent investigations were conducted on the immunostimulatory activity of the MS2+15,597 mixture. The results indicated that the dsRNA mixtures (MS2+15,597) extracted from E. coli infected with the MS2 bacteriophage possess the capability to activate innate immunity, thereby inducing the production of interferon-ß. These dsRNA mixtures can activate the RIG-I and TLR3 pattern recognition receptors, stimulating the expression of interferon stimulatory factors 3/7, which in turn triggers the NF-κB signaling pathway, culminating in the cellular production of interferon-ß to achieve antiviral effects. This study offers novel insights and strategies for the development of broad-spectrum antiviral drugs, potentially providing new modalities for future antiviral therapies.


Asunto(s)
Escherichia coli , Levivirus , ARN Bicatenario , Escherichia coli/virología , Escherichia coli/genética , Escherichia coli/metabolismo , ARN Bicatenario/metabolismo , Humanos , Levivirus/genética , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 3/genética , Inmunidad Innata , Interferón beta/metabolismo , Interferón beta/genética , FN-kappa B/metabolismo , Proteína 58 DEAD Box/metabolismo , Proteína 58 DEAD Box/genética , Transducción de Señal , Factor 7 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/genética , Receptores Inmunológicos , Factor 3 Regulador del Interferón/metabolismo , Factor 3 Regulador del Interferón/genética
11.
Cell Mol Biol Lett ; 29(1): 61, 2024 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-38671352

RESUMEN

BACKGROUND: Macrophage proinflammatory activation contributes to the pathology of severe acute pancreatitis (SAP) and, simultaneously, macrophage functional changes, and increased pyroptosis/necrosis can further exacerbate the cellular immune suppression during the process of SAP, where cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) plays an important role. However, the function and mechanism of cGAS-STING in SAP-induced lung injury (LI) remains unknown. METHODS: Lipopolysaccharide (LPS) was combined with caerulein-induced SAP in wild type, cGAS -/- and sting -/- mice. Primary macrophages were extracted via bronchoalveolar lavage and peritoneal lavage. Ana-1 cells were pretreated with LPS and stimulated with nigericin sodium salt to induce pyroptosis in vitro. RESULTS: SAP triggered NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome activation-mediated pyroptosis of alveolar and peritoneal macrophages in mouse model. Knockout of cGAS/STING could ameliorate NLRP3 activation and macrophage pyroptosis. In addition, mitochondrial (mt)DNA released from damaged mitochondria further induced macrophage STING activation in a cGAS- and dose-dependent manner. Upregulated STING signal can promote NLRP3 inflammasome-mediated macrophage pyroptosis and increase serum interleukin (IL)-6, IL-1ß, and tumor necrosis factor (TNF)-α levels and, thus, exacerbate SAP-associated LI (SAP-ALI). Downstream molecules of STING, IRF7, and IRF3 connect the mtDNA-cGAS-STING axis and the NLRP3-pyroptosis axis. CONCLUSIONS: Negative regulation of any molecule in the mtDNA-cGAS-STING-IRF7/IRF3 pathway can affect the activation of NLRP3 inflammasomes, thereby reducing macrophage pyroptosis and improving SAP-ALI in mouse model.


Asunto(s)
ADN Mitocondrial , Factor 3 Regulador del Interferón , Lesión Pulmonar , Macrófagos , Proteínas de la Membrana , Nucleotidiltransferasas , Pancreatitis , Piroptosis , Transducción de Señal , Animales , Piroptosis/genética , Factor 3 Regulador del Interferón/metabolismo , Factor 3 Regulador del Interferón/genética , Ratones , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/genética , Pancreatitis/metabolismo , Pancreatitis/genética , Pancreatitis/patología , Pancreatitis/inducido químicamente , Macrófagos/metabolismo , Lesión Pulmonar/patología , Lesión Pulmonar/genética , Lesión Pulmonar/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Inflamasomas/metabolismo , Lipopolisacáridos , Masculino , Modelos Animales de Enfermedad
12.
J Innate Immun ; 16(1): 226-247, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38527452

RESUMEN

INTRODUCTION: While TLR ligands derived from microbial flora and pathogens are important activators of the innate immune system, a variety of factors such as intracellular bacteria, viruses, and parasites can induce a state of hyperreactivity, causing a dysregulated and potentially life-threatening cytokine over-response upon TLR ligand exposure. Type I interferon (IFN-αß) is a central mediator in the induction of hypersensitivity and is strongly expressed in splenic conventional dendritic cells (cDC) and marginal zone macrophages (MZM) when mice are infected with adenovirus. This study investigates the ability of adenoviral infection to influence the activation state of the immune system and underlines the importance of considering this state when planning the treatment of patients. METHODS: Infection with adenovirus-based vectors (Ad) or pretreatment with recombinant IFN-ß was used as a model to study hypersensitivity to lipopolysaccharide (LPS) in mice, murine macrophages, and human blood samples. The TNF-α, IL-6, IFN-αß, and IL-10 responses induced by LPS after pretreatment were measured. Mouse knockout models for MARCO, IFN-αßR, CD14, IRF3, and IRF7 were used to probe the mechanisms of the hypersensitive reaction. RESULTS: We show that, similar to TNF-α and IL-6 but not IL-10, the induction of IFN-αß by LPS increases strongly after Ad infection. This is true both in mice and in human blood samples ex vivo, suggesting that the regulatory mechanisms seen in the mouse are also present in humans. In mice, the scavenger receptor MARCO on IFN-αß-producing cDC and splenic marginal zone macrophages is important for Ad uptake and subsequent cytokine overproduction by LPS. Interestingly, not all IFN-αß-pretreated macrophage types exposed to LPS exhibit an enhanced TNF-α and IL-6 response. Pretreated alveolar macrophages and alveolar macrophage-like murine cell lines (MPI cells) show enhanced responses, while bone marrow-derived and peritoneal macrophages show a weaker response. This correlates with the respective absence or presence of the anti-inflammatory IL-10 response in these different macrophage types. In contrast, Ad or IFN-ß pretreatment enhances the subsequent induction of IFN-αß in all macrophage types. IRF3 is dispensable for the LPS-induced IFN-αß overproduction in infected MPI cells and partly dispensable in infected mice, while IRF7 is required. The expression of the LPS co-receptor CD14 is important but not absolutely required for the elicitation of a TNF-α over-response to LPS in Ad-infected mice. CONCLUSION: Viral infections or application of virus-based vaccines induces type I interferon and can tip the balance of the innate immune system in the direction of hyperreactivity to a subsequent exposure to TLR ligands. The adenoviral model presented here is one example of how multiple factors, both environmental and genetic, affect the physiological responses to pathogens. Being able to measure the current reactivity state of the immune system would have important benefits for infection-specific therapies and for the prevention of vaccination-elicited adverse effects.


Asunto(s)
Adenoviridae , Citocinas , Factor 3 Regulador del Interferón , Lipopolisacáridos , Macrófagos , Ratones Noqueados , Animales , Ratones , Lipopolisacáridos/inmunología , Humanos , Factor 3 Regulador del Interferón/metabolismo , Factor 3 Regulador del Interferón/genética , Macrófagos/inmunología , Citocinas/metabolismo , Ratones Endogámicos C57BL , Factor 7 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/genética , Vectores Genéticos , Infecciones por Adenoviridae/inmunología , Interferón Tipo I/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Células Cultivadas , Células Dendríticas/inmunología , Interferón beta/metabolismo
13.
J Biol Chem ; 300(4): 107200, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38508315

RESUMEN

Interferon (IFN) regulatory factors (IRF) are key transcription factors in cellular antiviral responses. IRF7, a virus-inducible IRF, expressed primarily in myeloid cells, is required for transcriptional induction of interferon α and antiviral genes. IRF7 is activated by virus-induced phosphorylation in the cytoplasm, leading to its translocation to the nucleus for transcriptional activity. Here, we revealed a nontranscriptional activity of IRF7 contributing to its antiviral functions. IRF7 interacted with the pro-inflammatory transcription factor NF-κB-p65 and inhibited the induction of inflammatory target genes. Using knockdown, knockout, and overexpression strategies, we demonstrated that IRF7 inhibited NF-κB-dependent inflammatory target genes, induced by virus infection or toll-like receptor stimulation. A mutant IRF7, defective in transcriptional activity, interacted with NF-κB-p65 and suppressed NF-κB-induced gene expression. A single-action IRF7 mutant, active in anti-inflammatory function, but defective in transcriptional activity, efficiently suppressed Sendai virus and murine hepatitis virus replication. We, therefore, uncovered an anti-inflammatory function for IRF7, independent of transcriptional activity, contributing to the antiviral response of IRF7.


Asunto(s)
Factor 7 Regulador del Interferón , FN-kappa B , Animales , Humanos , Ratones , Células HEK293 , Inflamación/genética , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/inmunología , FN-kappa B/genética , FN-kappa B/inmunología , Virus Sendai/fisiología , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/inmunología , Replicación Viral , Mutación , Regulación de la Expresión Génica/genética , Virus de la Hepatitis Murina/fisiología , Infecciones por Coronavirus/inmunología , Infecciones por Respirovirus/inmunología
14.
Environ Int ; 184: 108480, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38341879

RESUMEN

1,2-Dichloroethane (1,2-DCE) is a prevalent environmental contaminant, and our study revealed its induction of testicular toxicity in mice upon subacute exposure. Melatonin, a prominent secretory product of the pineal gland, has been shown to offer protection against pyroptosis in male reproductive toxicity. However, the exact mechanism underlying 1,2-DCE-induced testicular toxicity and the comprehensive extent of melatonin's protective effects in this regard remain largely unexplored. Therefore, we sequenced testis piRNAs in mice exposed to environmentally relevant concentrations of 1,2-DCE by 28-day dynamic inhalation, and investigated the role of key piRNAs using GC-2 spd cells. Our results showed that 1,2-DCE induced mouse testicular damage and GC-2 spd cell pyroptosis. 1,2-DCE upregulated the expression of pyroptosis-correlated proteins in both mouse testes and GC-2 spd cells. 1,2-DCE exposure caused pore formation on cellular membranes and lactate dehydrogenase leakage in GC-2 spd cells. Additionally, we identified three upregulated piRNAs in 1,2-DCE-exposed mouse testes, among which piR-mmu-1019957 induced pyroptosis in GC-2 spd cells, and its inhibition alleviated 1,2-DCE-induced pyroptosis. PiR-mmu-1019957 mimic and 1,2-DCE treatment activated the expression of interferon regulatory factor 7 (IRF7) in GC-2 spd cells. IRF7 knockdown reversed 1,2-DCE-induced cellular pyroptosis, and overexpression of piR-mmu-1019957 did not promote pyroptosis when IRF7 was inhibited. Notably, melatonin reversed 1,2-DCE-caused testicular toxicity, cellular pyroptosis, and upregulated piR-mmu-1019957 and IRF7. Collectively, our findings indicated that melatonin mitigates this effect, suggesting its potential as a therapeutic intervention against 1,2-DCE-induced male reproductive toxicity in clinical practice.


Asunto(s)
Dicloruros de Etileno , Melatonina , Testículo , Masculino , Ratones , Animales , Piroptosis , Melatonina/farmacología , Melatonina/metabolismo , ARN de Interacción con Piwi , Factor 7 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/farmacología
15.
Eur J Pharmacol ; 968: 176382, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38311277

RESUMEN

Psoriasis is a chronic, recurrent, inflammatory dermatosis accompanied by excessive activation of dendritic cells (DCs), which are primarily responsible for initiating an immune response. The bromodomain and extraterminal domain (BET) family plays a pivotal role in the transcriptional regulation of inflammation and its inhibitors can downregulate DCs maturation and activation. Here we investigated the effect of NHWD-870, a potent BET inhibitor, on inflammation in an imiquimod (IMQ)-induced psoriasis-like mouse model and murine bone marrow-derived dendritic cells (BMDCs) stimulated by lipopolysaccharide (LPS) and IMQ. Application of NHWD-870 significantly ameliorated IMQ-triggered skin inflammation in mice, and markers associated with DC maturation (CD40, CD80 and CD86) were decreased in skin lesions, spleen and lymph nodes. Additionally, NHWD-870 reduced LPS or IMQ induced DCs maturation and activation in vitro, with lower expression of inflammatory cytokines [interleukin (IL)-12, IL-23, tumor necrosis factor-α, IL-6, IL-1ß, chemokine (C-X-C motif) ligand (CXCL)9 and CXCL10]. In addition, we found that interferon regulatory factor 7 (IRF7) significantly increased during DCs maturation, and inhibition of IRF7 could impair BMDCs maturation and activation. What's more, IRF7 was highly expressed in both psoriatic patients and IMQ-induced psoriasis-like mice. Single-cell RNA sequencing of normal and psoriatic skin demonstrated that IRF7 expression was increased in DCs of psoriatic skin. While NHWD-870 could inhibit IRF7 and phosphorylated-IRF7 expression in vivo and in vitro. These results indicate that NHWD-870 suppresses the maturation and activation of DCs by decreasing IRF7 proteins which finally alleviates psoriasis-like skin lesions, and NHWD-870 may be a potent therapeutic drug for psoriasis.


Asunto(s)
Dermatitis , Psoriasis , Humanos , Animales , Ratones , Imiquimod/efectos adversos , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/farmacología , Lipopolisacáridos/metabolismo , Psoriasis/inducido químicamente , Psoriasis/tratamiento farmacológico , Piel , Dermatitis/patología , Inflamación/patología , Células Dendríticas , Transducción de Señal , Modelos Animales de Enfermedad , Ratones Endogámicos BALB C
16.
Adv Sci (Weinh) ; 11(13): e2304991, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38286661

RESUMEN

Radiotherapy (RT) can induce tumor regression outside the irradiation field, known as the abscopal effect. However, the detailed underlying mechanisms remain largely unknown. A tumor-bearing mouse model is successfully constructed by inducing both subcutaneous tumors and lung metastases. Single-cell RNA sequencing, immunofluorescence, and flow cytometry are performed to explore the regulation of tumor microenvironment (TME) by RT. A series of in vitro assays, including luciferase reporter, RNA Pulldown, and fluorescent in situ hybridization (FISH) assays, are performed to evaluate the detailed mechanism of the abscopal effect. In addition, in vivo assays are performed to investigate combination therapy strategies for enhancing the abscopal effect. The results showed that RT significantly inhibited localized tumor and lung metastasis progression and improved the TME. Mechanistically, RT promoted the release of tumor-derived exosomes carrying circPIK3R3, which is taken up by macrophages. circPIK3R3 promoted Type I interferon (I-IFN) secretion and M1 polarization via the miR-872-3p/IRF7 axis. Secreted I-IFN activated the JAK/STAT signaling pathway in CD8+ T cells, and promoted IFN-γ and GZMB secretion. Together, the study shows that tumor-derived exosomes promote I-IFN secretion via the circPIK3R3/miR-872-3p/IRF7 axis in macrophages and enhance the anti-tumor immune response of CD8+ T cells.


Asunto(s)
Exosomas , Neoplasias Pulmonares , Melanoma , MicroARNs , Animales , Ratones , Anticuerpos , Linfocitos T CD8-positivos , Exosomas/efectos de la radiación , Hibridación Fluorescente in Situ , Interferones , Neoplasias Pulmonares/radioterapia , Macrófagos/efectos de la radiación , Melanoma/radioterapia , MicroARNs/genética , Microambiente Tumoral , Factor 7 Regulador del Interferón/inmunología , Factor 7 Regulador del Interferón/efectos de la radiación
17.
J Affect Disord ; 349: 297-309, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38211750

RESUMEN

BACKGROUND: Postoperative neurocognitive disorder (PND) is a common central nervous system complication after undergoing surgery and anesthesia especially in elderly patients, while the therapeutic options are very limited. This study was carried out to investigate the beneficial effects of transcranial near infrared light (NIRL) which was employed to the treatment of PND and propose the involved mechanisms. METHODS: The PND mice were established through left carotid artery exposure under isoflurane anesthesia and received transcranial NIRL treatment. Behavioral testing was performed to evaluate the cognitive function of PND mice after transcranial NIRL therapy. Changes in the transcriptomic profiles of prefrontal cortex (PFC) and hippocampus (HP) were identified by next generation sequencing (NGS), and the molecular mechanisms involved were examined by both in vivo mouse model and in vitro cell culture studies. RESULTS: We found that transcranial NIRL therapy effectively ameliorated learning and memory deficit induced by anesthesia and surgery in aged mice. Specifically, we identified down-regulation of interferon regulatory factor 7 (IRF7) in the brains of PND mice that was mechanistically associated with increased pro-inflammatory M1 phenotype of microglia and elevated neuroinflammatory. NIRL treatment produced protective effects through the upregulation of IRF7 expression and reversing microglial phenotypes from pro-inflammatory to neuroprotective, resulting in reduced brain damage and improved cognitive function in PND mice. CONCLUSION: Our results indicate that transcranial NIRL is an effective and safe therapy for PND via alleviating neuroinflammation, and IRF7 plays a key transcription factor in regulating the M1-to-M2 switch of microglia.


Asunto(s)
Factor 7 Regulador del Interferón , Fármacos Neuroprotectores , Anciano , Animales , Humanos , Ratones , Encéfalo/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Ratones Endogámicos C57BL , Trastornos Neurocognitivos , Fototerapia
18.
Mol Biol Rep ; 51(1): 114, 2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38227268

RESUMEN

BACKGROUND: The production of interferons (IFNs) is essential for the control of viral infections, and interferon regulatory factor 7 (IRF7) is considered as a vital regulator for the transcription of type I IFNs. Amphibians appear to possess a highly expanded type I IFN repertoire, consisting of intron-containing genes as observed in fish, and intronless genes as in other higher vertebrates. However, the knowledge on transcriptional regulatory mechanism of these two types of type I IFN genes is rather scarce in amphibians. METHODS AND RESULTS: A IRF7 gene named as Np-IRF7 was identified in Tibetan frog (Nanorana parkeri), and bioinformatic analysis revealed that the predicted protein of Np-IRF7 contains several important structural features known in IRF7. Expression analysis showed that Np-IRF7 gene was widely expressed and rapidly induced by poly(I:C) in different organs/tissues. Interestingly, luciferase reporter assay revealed that intronless IFN promoters were more effectively activated than intron-containing IFN promoter in Np-IRF7-transfected cells. Moreover, the overexpression of Np-IRF7 could induce the expression of ISGs and suppress the replication of FV3 in A6 cells. CONCLUSION: Np-IRF7 is indeed the ortholog of known IRF7, and IRF7 is structurally conserved in different lineages of vertebrates. Np-IRF7 played distinct roles in the activation of intron-containing and intronless type I IFN promoters, thus inducing the expression of interferon-stimulated antiviral effectors and providing a protection against ranavirus infection. The present research thus contributes to a better understanding of regulatory function of IRF7 in the IFN-mediated antiviral response of anuran amphibians.


Asunto(s)
Factor 7 Regulador del Interferón , Interferón Tipo I , Animales , Humanos , Factor 7 Regulador del Interferón/genética , Tibet , Anuros/genética , Intrones/genética , Interferón Tipo I/genética
19.
Inflammation ; 47(1): 404-420, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37930487

RESUMEN

In the realm of Parkinson's disease (PD) research, NLRP3 inflammasome-mediated pyroptosis has recently garnered significant attention as a potential novel form of dopaminergic neuronal death. Our previous research revealed the activation of innate immune-related genes, such as the TLR4 signaling pathway and interferon regulatory factor 7 (IRF7), although the specific mechanism remains unclear. Our current study shed light on whether the TLR4 signaling pathway and IRF7 can affect the pyroptosis of dopaminergic nerve cells and thus participate in the pathogenesis of PD. The PD model was constructed by MPP+ treatment of PC12 cells or stereotactic injection of the striatum of SD rats, and the expression of genes were detected by RT-qPCR and Western Blotting. Lentivirus, siRNA and (5Z)-7-Oxozeaenol were used to validate the regulation of this pathway on pyroptosis. The expression of TLR4, TAK1, IRF7 and pyroptosis molecular markers was upregulated after MPP+ treatment. IRF7 could affect dopaminergic neural cells pyroptosis by targeted regulation of NLRP3. Furthermore, inhibition of the TLR4/TAK1 signaling pathway led to a decrease in the expression of both IRF7 and NLRP3, while overexpression of IRF7 reversed the reduction in pyroptosis and increase in TH expression. TLR4/TAK1/IRF7 axis can promote PD by influencing pyroptosis through NLRP3.


Asunto(s)
Proteína con Dominio Pirina 3 de la Familia NLR , Enfermedad de Parkinson , Ratas , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis , Enfermedad de Parkinson/metabolismo , Receptor Toll-Like 4/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Ratas Sprague-Dawley , Inflamasomas/metabolismo
20.
J Biol Chem ; 300(1): 105525, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38043800

RESUMEN

The innate antiviral response to RNA viruses is initiated by sensing of viral RNAs by RIG-I-like receptors and elicits type I interferon (IFN) production, which stimulates the expression of IFN-stimulated genes that orchestrate the antiviral response to prevent systemic infection. Negative regulation of type I IFN and its master regulator, transcription factor IRF7, is essential to maintain immune homeostasis. We previously demonstrated that AIP (aryl hydrocarbon receptor interacting protein) functions as a negative regulator of the innate antiviral immune response by binding to and sequestering IRF7 in the cytoplasm, thereby preventing IRF7 transcriptional activation and type I IFN production. However, it remains unknown how AIP inhibition of IRF7 is regulated. We show here that the kinase TBK1 phosphorylates AIP and Thr40 serves as the primary target for TBK1 phosphorylation. AIP Thr40 plays critical roles in regulating AIP stability and mediating its interaction with IRF7. The AIP phosphomimetic T40E exhibited increased proteasomal degradation and enhanced interaction with IRF7 compared with wildtype AIP. AIP T40E also blocked IRF7 nuclear translocation, which resulted in reduced type I IFN production and increased viral replication. In sharp contrast, AIP phosphonull mutant T40A had impaired IRF7 binding, and stable expression of AIP T40A in AIP-deficient mouse embryonic fibroblasts elicited a heightened type I IFN response and diminished RNA virus replication. Taken together, these results demonstrate that TBK1-mediated phosphorylation of AIP at Thr40 functions as a molecular switch that enables AIP to interact with and inhibit IRF7, thus preventing overactivation of type I IFN genes by IRF7.


Asunto(s)
Inmunidad Innata , Factor 7 Regulador del Interferón , Interferón Tipo I , Proteínas Serina-Treonina Quinasas , Infecciones por Virus ARN , Virus ARN , Receptores de Hidrocarburo de Aril , Animales , Ratones , Fibroblastos , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Virus ARN/inmunología , Infecciones por Virus ARN/inmunología , Humanos , Células HEK293
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA