Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Immunity ; 56(1): 180-192.e11, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36563676

ABSTRACT

The reinvigoration of anti-tumor T cells in response to immune checkpoint blockade (ICB) therapy is well established. Whether and how ICB therapy manipulates antibody-mediated immune response in cancer environments, however, remains elusive. Using tandem mass spectrometric analysis of modification of immunoglobulin G (IgG) from hepatoma tissues, we identified a role of ICB therapy in catalyzing IgG sialylation in the Fc region. Effector T cells triggered sialylation of IgG via an interferon (IFN)-γ-ST6Gal-I-dependent pathway. DC-SIGN+ macrophages represented the main target cells of sialylated IgG. Upon interacting with sialylated IgG, DC-SIGN stimulated Raf-1-elicited elevation of ATF3, which inactivated cGAS-STING pathway and eliminated subsequent type-I-IFN-triggered antitumorigenic immunity. Although enhanced IgG sialylation in tumors predicted improved therapeutic outcomes for patients receiving ICB therapy, impeding IgG sialylation augmented antitumorigenic T cell immunity after ICB therapy. Thus, targeting antibody-based negative feedback action of ICB therapy has potential for improving efficacy of cancer immunotherapies.


Subject(s)
Carcinoma, Hepatocellular , Interferon Type I , Liver Neoplasms , Humans , Immunoglobulin G , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Immunotherapy/methods
2.
Mol Cell ; 83(1): 90-104.e4, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36521492

ABSTRACT

RIG-I is essential for host defense against viral pathogens, as it triggers the release of type I interferons upon encounter with viral RNA molecules. In this study, we show that RIG-I is rapidly and efficiently activated by small quantities of incoming viral RNA and that it relies exclusively on the constitutively expressed resident pool of RIG-I receptors for a strong antiviral response. Live-cell imaging of RIG-I following stimulation with viral or synthetic dsRNA reveals that RIG-I signaling occurs without mass aggregation at the mitochondrial membrane. By contrast, interferon-induced RIG-I protein becomes embedded in cytosolic aggregates that are functionally unrelated to signaling. These findings suggest that endogenous RIG-I efficiently recognizes viral RNA and rapidly relays an antiviral signal to MAVS via a transient signaling complex and that cellular aggregates of RIG-I have a function that is distinct from signaling.


Subject(s)
Interferon Type I , Signal Transduction , Signal Transduction/genetics , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Antiviral Agents/pharmacology , Interferon Type I/genetics , RNA, Double-Stranded/genetics , RNA, Viral/genetics , Immunity, Innate
3.
Mol Cell ; 83(23): 4370-4385.e9, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-38016475

ABSTRACT

Targeting epigenetic regulators to potentiate anti-PD-1 immunotherapy converges on the activation of type I interferon (IFN-I) response, mimicking cellular response to viral infection, but how its strength and duration are regulated to impact combination therapy efficacy remains largely unknown. Here, we show that mitochondrial CPT1A downregulation following viral infection restrains, while its induction by epigenetic perturbations sustains, a double-stranded RNA-activated IFN-I response. Mechanistically, CPT1A recruits the endoplasmic reticulum-localized ZDHHC4 to catalyze MAVS Cys79-palmitoylation, which promotes MAVS stabilization and activation by inhibiting K48- but facilitating K63-linked ubiquitination. Further elevation of CPT1A incrementally increases MAVS palmitoylation and amplifies the IFN-I response, which enhances control of viral infection and epigenetic perturbation-induced antitumor immunity. Moreover, CPT1A chemical inducers augment the therapeutic effect of combined epigenetic treatment with PD-1 blockade in refractory tumors. Our study identifies CPT1A as a stabilizer of MAVS activation, and its link to epigenetic perturbation can be exploited for cancer immunotherapy.


Subject(s)
Interferon Type I , Virus Diseases , Humans , Signal Transduction , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Lipoylation , Epigenesis, Genetic , Immunity, Innate
4.
J Infect Dis ; 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38195164

ABSTRACT

The varicella-zoster virus (VZV) infects over 95% of the population. VZV reactivation causes herpes zoster (HZ), known as shingles, primarily affecting the elderly and immunocompromised individuals. However, HZ can also occur in otherwise healthy individuals. We analyzed the immune signature and risk profile in HZ patients using a genome-wide association study across different UK Biobank HZ cohorts. Additionally, we conducted one of the largest HZ HLA association studies to date, coupled with transcriptomic analysis of pathways underlying HZ susceptibility. Our findings highlight the significance of the MHC locus for HZ development, identifying five protective and four risk HLA alleles. This demonstrates that HZ susceptibility is largely governed by variations in the MHC. Furthermore, functional analyses revealed the upregulation of type I interferon and adaptive immune responses. These findings provide fresh molecular insights into the pathophysiology and the activation of innate and adaptive immune responses triggered by symptomatic VZV reactivation.

5.
Mol Cell ; 64(2): 267-281, 2016 10 20.
Article in English | MEDLINE | ID: mdl-27692986

ABSTRACT

TBK1 is a component of the type I interferon (IFN) signaling pathway, yet the mechanisms controlling its activity and degradation remain poorly understood. Here we report that USP38 negatively regulates type I IFN signaling by targeting the active form of TBK1 for degradation in vitro and in vivo. USP38 specifically cleaves K33-linked poly-ubiquitin chains from TBK1 at Lys670, and it allows for subsequent K48-linked ubiquitination at the same position mediated by DTX4 and TRIP. Knockdown or knockout of USP38 increases K33-linked ubiquitination, but it abrogates K48-linked ubiquitination and degradation of TBK1, thus enhancing type I IFN signaling. Our findings identify an essential role for USP38 in negatively regulating type I IFN signaling, and they provide insights into the mechanisms by which USP38 regulates TBK1 ubiquitination through the NLRP4 signalosome.


Subject(s)
Immunity, Innate , Interferon Type I/metabolism , Macrophages/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteins/metabolism , Signal Transduction/immunology , Ubiquitin-Specific Proteases/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/virology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/immunology , Eukaryotic Initiation Factors/metabolism , Gene Expression Regulation , Herpesvirus 1, Human/growth & development , Herpesvirus 1, Human/immunology , Host-Pathogen Interactions , Interferon Type I/genetics , Interferon Type I/immunology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/immunology , Macrophages/virology , Mice , Mice, Knockout , Phosphorylation , Polyubiquitin/genetics , Polyubiquitin/immunology , Polyubiquitin/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Proteins/genetics , Proteins/immunology , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/immunology , Ubiquitination , Vesiculovirus/growth & development , Vesiculovirus/immunology
6.
Int J Mol Sci ; 25(4)2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38397027

ABSTRACT

A feature of the SARS-CoV-2 Omicron subvariants BF.5 and BF.7 that recently circulated mainly in China and Japan was the high prevalence of the ORF7a: H47Y mutation, in which the 47th residue of ORF7a has been mutated from a histidine (H) to a tyrosine (Y). Here, we evaluated the effect of this mutation on the three main functions ascribed to the SARS-CoV-2 ORF7a protein. Our findings show that H47Y mutation impairs the ability of SARS-CoV-2 ORF7a to antagonize the type I interferon (IFN-I) response and to downregulate major histocompatibility complex I (MHC-I) cell surface levels, but had no effect in its anti-SERINC5 function. Overall, our results suggest that the H47Y mutation of ORF7a affects important functions of this protein, resulting in changes in virus pathogenesis.


Subject(s)
COVID-19 , Interferon Type I , Humans , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , COVID-19/genetics , Interferon Type I/metabolism , Mutation , China
7.
Int J Mol Sci ; 24(17)2023 Aug 24.
Article in English | MEDLINE | ID: mdl-37685953

ABSTRACT

The innate immune system is the first line of defense against pathogens such as the acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The type I-interferon (IFN) response activation during the initial steps of infection is essential to prevent viral replication and tissue damage. SARS-CoV and SARS-CoV-2 can inhibit this activation, and individuals with a dysregulated IFN-I response are more likely to develop severe disease. Several mutations in different variants of SARS-CoV-2 have shown the potential to interfere with the immune system. Here, we evaluated the buffy coat transcriptome of individuals infected with Gamma or Delta variants of SARS-CoV-2. The Delta transcriptome presents more genes enriched in the innate immune response and Gamma in the adaptive immune response. Interactome and enriched promoter analysis showed that Delta could activate the INF-I response more effectively than Gamma. Two mutations in the N protein and one in the nsp6 protein found exclusively in Gamma have already been described as inhibitors of the interferon response pathway. This indicates that the Gamma variant evolved to evade the IFN-I response. Accordingly, in this work, we showed one of the mechanisms that variants of SARS-CoV-2 can use to avoid or interfere with the host Immune system.


Subject(s)
COVID-19 , Interferon Type I , Severe acute respiratory syndrome-related coronavirus , Humans , Interferon Type I/genetics , SARS-CoV-2 , Transcriptome , COVID-19/genetics
8.
Int J Mol Sci ; 24(21)2023 Oct 24.
Article in English | MEDLINE | ID: mdl-37958498

ABSTRACT

Bioactive lipids are involved in cellular signalling events with links to human disease. Many of these are involved in inflammation under normal and pathological conditions. Despite being attractive molecules from a pharmacological point of view, the detection and quantification of lipids has been a major challenge. Here, we have optimised a liquid chromatography-dynamic multiple reaction monitoring-targeted mass spectrometry (LC-dMRM-MS) approach to profile eicosanoids and fatty acids in biological samples. In particular, by applying this analytic workflow to study a cellular model of chronic myeloid leukaemia (CML), we found that the levels of intra- and extracellular 2-Arachidonoylglycerol (2-AG), intracellular Arachidonic Acid (AA), extracellular Prostaglandin F2α (PGF2α), extracellular 5-Hydroxyeicosatetraenoic acid (5-HETE), extracellular Palmitic acid (PA, C16:0) and extracellular Stearic acid (SA, C18:0), were altered in response to immunomodulation by type I interferon (IFN-I), a currently approved treatment for CML. Our observations indicate changes in eicosanoid and fatty acid metabolism, with potential relevance in the context of cancer inflammation and CML.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Leukemia, Myeloid , Humans , Fatty Acids , Interferons , Tandem Mass Spectrometry/methods , Eicosanoids/metabolism , Inflammation
9.
J Biol Chem ; 296: 100683, 2021.
Article in English | MEDLINE | ID: mdl-33887325

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) is a devastating pathogen in the swine industry worldwide. miRNAs are reported to be involved in virus-host interaction. Here, we used high-throughput sequencing and miRNA inhibitors to screen possible miRNAs that can inhibit PRRSV infection on its target cell, porcine alveolar macrophages. We observed that miR-218 was downregulated upon virus infection, and knockdown of miR-218 significantly enhanced PRRSV replication. Overexpression of miR-218 resulted in a decrease in PRRSV replication, and this overexpression did not alter viral genomic RNA levels, but rather increased antiviral interferon signaling. Further analysis revealed that miR-218 regulated PRRSV replication by directly targeting porcine suppressor of cytokine signaling 3 (SOCS3), a JAK2 kinase inhibitor. Knockdown of the endogenous SOCS3 expression led to augmentation of type I interferon genes and resulted in decreased PRRSV replication, and vice versa. During PRRSV infection in vivo and in vitro, cellular miR-218 expression was downregulated and SOCS3 expression was upregulated, further supporting the inverse correlation between miR-218 and SOCS3 expression. The data on SOCS3 depletion in combination with miR-218 inhibition suggested that the antiviral activity of miR-218 required the SOCS3-mediated signaling pathway. Similarly, miR-218 negatively regulated PRRSV replication in Marc-145 cells, as well as the replication of porcine epidemic diarrhea virus and transmissible gastroenteritis virus in Vero and ST cells respectively. Taken together, these results demonstrate that PRRSV-induced miR-218 downregulation serves to inhibit the type I interferon response and may provide a novel therapeutic target for treatment of PRRSV and other viral infections.


Subject(s)
Down-Regulation , Interferon Type I/metabolism , Macrophages, Alveolar/virology , MicroRNAs/genetics , Porcine respiratory and reproductive syndrome virus/physiology , Virus Replication , Animals , Cell Line , Macrophages, Alveolar/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism
10.
Eur J Clin Invest ; 52(10): e13818, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35598178

ABSTRACT

BACKGROUND: SARS-CoV-2 infection triggers different auto-antibodies, including anti-apolipoprotein A-1 IgGs (AAA1), which could be of concern as mediators of persistent symptoms. We determined the kinetics of AAA1 response over after COVID-19 and the impact of AAA1 on the inflammatory response and symptoms persistence. METHODS: All serologies were assessed at one, three, six and twelve months in 193 hospital employees with COVID-19. ROC curve analyses and logistic regression models (LRM) were used to determine the prognostic accuracy of AAA1 and their association with patient-reported COVID-19 symptoms persistence at 12 months. Interferon (IFN)-α and-γ production by AAA1-stimulated human monocyte-derived macrophages (HMDM) was assessed in vitro. RESULTS: AAA1 seropositivity was 93% at one month and declined to 15% at 12 months after COVID-19. Persistent symptoms at 12 months were observed in 45.1% of participants, with a predominance of neurological (28.5%), followed by general (15%) and respiratory symptoms (9.3%). Over time, strength of correlations between AAA1 and anti-SARS-COV2 serologies decreased, but remained significant. From the 3rd month on, AAA1 levels predicted persistent respiratory symptoms (area under the curves 0.72-0.74; p < 0.001), independently of disease severity, age and gender (adjusted odds ratios 4.81-4.94; p = 0.02), while anti-SARS-CoV-2 serologies did not. AAA1 increased IFN-α production by HMDMs (p = 0.03), without affecting the IFN-γ response. CONCLUSION: COVID-19 induces a marked though transient AAA1 response, independently predicting one-year persistence of respiratory symptoms. By increasing IFN-α response, AAA1 may contribute to persistent symptoms. If and how AAA1 levels assessment could be of use for COVID-19 risk stratification remains to be determined.


Subject(s)
COVID-19 , Antibodies, Viral , Antiviral Agents , Apolipoprotein A-I , Autoantibodies , Humans , SARS-CoV-2
11.
Vet Res ; 51(1): 145, 2020 Dec 09.
Article in English | MEDLINE | ID: mdl-33298177

ABSTRACT

This study focuses on the immunoregulatory effects of chicken TRIM25 on the replication of subgroup A of avian leukosis virus (ALV-A) and the MDA5-mediated type I interferon response. The ALV-A-SDAU09C1 strain was inoculated into DF1 cells and 1-day-old SPF chickens, and the expression of TRIM25 was detected at different time points after inoculation. A recombinant overexpression plasmid containing the chicken TRIM25 gene (TRIM25-GFP) was constructed and transfected into DF1 cells to analyse the effects of the overexpression of chicken TRIM25 on the replication of ALV-A and the expression of MDA5, MAVS and IFN-ß. A small interfering RNA targeting chicken TRIM25 (TRIM25-siRNA) was prepared and transfected into DF1 cells to assess the effects of the knockdown of chicken TRIM25 on the replication of ALV-A and the expression of MDA5, MAVS and IFN-ß. The results showed that chicken TRIM25 was significantly upregulated at all time points both in ALV-A-infected cells and in ALV-A-infected chickens. Overexpression of chicken TRIM25 in DF1 cells dramatically decreased the antigenic titres of ALV-A in the cell supernatant and upregulated the relative expression of MDA5, MAVS and IFN-ß induced by ALV-A or by poly(I:C); in contrast, knockdown of chicken TRIM25 significantly increased the antigenic titres of ALV-A and downregulated the relative expression of MDA5, MAVS and IFN-ß. It can be concluded that chicken TRIM25 can inhibit the replication of ALV-A and upregulate the MDA5 receptor-mediated type I interferon response in chickens. This study can help improve the understanding of the antiviral activities of chicken TRIM25 and enrich the knowledge of antiviral responses in chickens.


Subject(s)
Avian Leukosis Virus/physiology , Chickens , Interferon-Induced Helicase, IFIH1/metabolism , Transcription Factors/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Avian Leukosis Virus/classification , Cell Line , Gene Expression Regulation/immunology , Gene Knockdown Techniques , Interferon-Induced Helicase, IFIH1/genetics , Interferon-beta/genetics , Interferon-beta/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/genetics , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Up-Regulation , Virus Replication
12.
Fish Shellfish Immunol ; 87: 721-729, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30753916

ABSTRACT

Salmonella is a facultative intracellular pathogen that can cause significant morbidity and mortality in humans and animals. Salmonella plasmid virulence (spv) gene sequence is a highly conserved 6.8 kb region which exists in the plasmid of most pathogenic Salmonella. Autophagy is a degradation process of unnecessary and dysfunctional cytoplasm components to maintain cellular homeostasis, which could affect host inflammatory responses, such as type I interferon response. Type I interferon response can promote the antibacterial activity of macrophage as well as the secretion of cytokines and neutrophil chemokines. We previously reported that spv locus could suppress autophagy and the aggregation of neutrophils in zebrafish larvae. To explore the influence of spv locus on Salmonella escaping from the innate immune responses and the underlying mechanism, the models of Salmonella enterica serovar Typhimurium infected macrophages in vitro and zebrafish larvae in vivo were used in this study. The interactions among spv locus, autophagy, type I interferon response and the chemotaxis of neutrophils were investigated. Western blot was used to detect the expression levels of autophagy related proteins and RT-qPCR was used to measure the mRNA levels of type I interferon response and the neutrophil chemokines. The chemotaxis of neutrophils were observed by Laser Scanning confocal microscopy. Autophagy agonist Torin 1 was also involved to interfere the autophagy influx. Results showed that spv locus could restrain type I interferon response and the chemotaxis of neutrophils via suppressing autophagy, which provided substantial foundation to study the mechanism of Salmonella escaping the innate immunity.


Subject(s)
Autophagy , Immunity, Innate/physiology , Interferon Type I/metabolism , Neutrophils/immunology , Salmonella typhimurium/physiology , Salmonella typhimurium/pathogenicity , Zebrafish/physiology , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Chemotaxis , Fish Diseases/immunology , Fish Proteins/metabolism , Plasmids/physiology , Random Allocation , Salmonella Infections, Animal/immunology , Virulence , Virulence Factors/genetics , Virulence Factors/immunology , Zebrafish/genetics , Zebrafish/immunology
13.
J Biol Chem ; 292(50): 20379-20393, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29046350

ABSTRACT

Promyelocytic leukemia nuclear bodies (PML-NB) are sub-nuclear organelles that are the hub of numerous proteins. DNA/RNA viruses often hijack the cellular factors resident in PML-NBs to promote their proliferation in host cells. Hepatitis B virus (HBV), belonging to Hepadnaviridae family, remains undetected in early infection as it does not induce the innate immune response and is known to be the cause of several hepatic diseases leading to cirrhosis and hepatocellular carcinoma. The association of PML-NB proteins and HBV is being addressed in a number of recent studies. Here, we report that the PML-NB protein Speckled 110 kDa (Sp110) is SUMO1-modified and undergoes a deSUMOylation-driven release from the PML-NB in the presence of HBV. Intriguingly, Sp110 knockdown significantly reduced viral DNA load in the culture supernatant by activation of the type I interferon-response pathway. Furthermore, we found that Sp110 differentially regulates several direct target genes of hepatitis B virus protein X (HBx), a viral co-factor. Subsequently, we identified Sp110 as a novel interactor of HBx and found this association to be essential for the exit of Sp110 from the PML-NB during HBV infection and HBx recruitment on the promoter of these genes. HBx, in turn, modulates the recruitment of its associated transcription cofactors p300/HDAC1 to these co-regulated genes, thereby altering the host gene expression program in favor of viral persistence. Thus, we report a mechanism by which HBV can evade host immune response by hijacking the PML-NB protein Sp110, and therefore, we propose it to be a novel target for antiviral therapy.


Subject(s)
Hepatitis B virus/metabolism , Hepatitis B, Chronic/metabolism , Hepatocytes/metabolism , Inclusion Bodies, Viral/metabolism , Minor Histocompatibility Antigens/metabolism , Nuclear Proteins/metabolism , Sumoylation , Trans-Activators/physiology , Apoptosis , Biomarkers/blood , Biomarkers/metabolism , DNA, Viral/metabolism , Gene Expression Regulation, Bacterial , Hep G2 Cells , Hepatitis B virus/growth & development , Hepatitis B virus/immunology , Hepatitis B virus/isolation & purification , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/pathology , Hepatitis B, Chronic/virology , Hepatocytes/immunology , Hepatocytes/pathology , Hepatocytes/virology , Host-Pathogen Interactions , Humans , Immunity, Innate , Inclusion Bodies, Viral/pathology , Inclusion Bodies, Viral/virology , Minor Histocompatibility Antigens/blood , Minor Histocompatibility Antigens/genetics , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/blood , Nuclear Proteins/genetics , Promoter Regions, Genetic , Protein Transport , RNA Interference , Viral Load , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Regulatory and Accessory Proteins , Virus Physiological Phenomena , Virus Replication
14.
Fish Shellfish Immunol ; 83: 443-448, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30244086

ABSTRACT

Viral hemorrhagic septicemia virus (VHSV) has been a notorious pathogen in freshwater and marine fish. Due to the lack of effective treatment measures against VHSV disease, the development of prophylactic vaccines has been required, and methods that can produce high-titered viruses would be advantageous in producing cost-effective vaccines. Type I interferon (IFN) responses are the key elements of vertebrates' antiviral activities, and IFN-stimulated gene factor 3 (ISGF3) complex formed through type I IFNs up-regulates the expression of IFN-stimulated genes (ISGs). IFN regulatory factor 9 (IRF9) is a key component of ISGF3, so the inhibition of IRF9 would compromise host's type I IFN responses, which would weaken host antiviral activity. In this study, to increase the replication of VHSV, we generated IRF9 knockout Epithelioma papulosum cyprini (EPC) cells using a CRISPR/Cas9 vector that contains an EPC cell's U6 promoter-driven guide RNA cassette (targeting IRF9 gene) and a Cas9 expressing cassette. In the clones of IRF9 knockout EPC cells, there were no increase in ISG15 gene by poly I:C, and in Mx1 gene by both poly I:C and VHSV. Interestingly, although the increased folds were conspicuously lower than control EPC cells, the expression of ISG 15 gene in all the IRF9 knockout clones was significantly increased by VHSV infection. Control EPC cells pre-treated with poly I:C did not show any CPE when infected with VHSV, however, IRF9 knockout EPC cells showed CPE by VHSV infection in spite of being pretreated with poly I:C. The replication of VHSV in IRF9 knockout EPC cells was significantly faster and higher than that in control EPC cells indicating that the IRF9 knockout-mediated decrease of type I IFN responses allowed VHSV to replicate efficiently. Considering an economical aspect for the production of fish vaccines, the present IRF9 knockout EPC cells can be used to get higher-titered VHSV.


Subject(s)
Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Novirhabdovirus/physiology , Animals , CRISPR-Cas Systems , Cell Line , Fishes , Gene Editing , Gene Knockout Techniques , Interferon Type I/immunology , Poly I-C/pharmacology , Virus Replication
15.
Fish Shellfish Immunol ; 58: 318-322, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27663852

ABSTRACT

Vaccines based on inactivated or attenuated viruses can be a way to prevent viral hemorrhagic septicemia virus (VHSV) disease, and the efficiency of viral production is a critical factor that can determine the practical use of developed vaccines in aquaculture farms. To know the effects of epithelioma papulosum cyprini (EPC) cells over-subculture on VHSV replication, the VHSV titer produced from high-passage EPC cells (subcultured more than 200 times in our laboratory) was compared to the titer produced from low-passage EPC cells (subcultured 5-15 times). Furthermore, to know whether immune factors are involved in VHSV titers, differences not only in the expression of Mx1 and ISG15 genes but also in the apoptosis progression by VHSV infection between high- and low-passage EPC cells were analyzed. The VHSV titers from high-passage EPC cells were significantly higher than titers from low-passage EPC cells, suggesting that the changed properties of EPC cells by over-subculture were favorable for VHSV proliferation. The DNA laddering of high-passage EPC cells by VHSV infection took a longer time than that of low-passage EPC cells, suggesting that over-subculture might delay apoptosis in VHSV infected EPC cells, and the delay of apoptosis by over-subculture can be thought as one of the factors that increased VHSV titers in high-passage EPC cells. The increased folds of Mx1 and ISG15 genes in high-passage EPC cells were significantly lower than those in low-passage EPC cells when exposed to either poly (I:C) or VHSV. However, the expression levels of Mx1 and ISG15 genes of high-passage EPC cells that were not stimulated with poly I:C or VHSV were almost equal to or higher than the expression levels of low-passage EPC cells that were exposed to poly (I:C) or VHSV. This result suggests that high-passage EPC cells were already in an excited state in type I interferon responses without any stimulants. The full open reading frame (ORF) sequences of Mx1 gene between high- and low-passage EPC cells were completely same. However, there were some differences in the amino acids sequences of ISG15 gene between high- and low-passage EPC cells, suggesting that ISG15-mediated pathways might be different between high- and low-passage EPC cells, which might influence on the replication of VHSV. The present results showed that the changed properties of EPC cells by over-subculture were favorable for VHSV proliferation.


Subject(s)
Apoptosis , Cell Culture Techniques/veterinary , Cytokines/genetics , Myxovirus Resistance Proteins/genetics , Novirhabdovirus/physiology , Virus Replication , Animals , Cell Line, Tumor , Cyprinidae , Cytokines/metabolism , Myxovirus Resistance Proteins/metabolism , Poly I-C/pharmacology
16.
Int J Cancer ; 135(6): 1381-9, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24347514

ABSTRACT

Glioblastoma is a deadly malignant brain tumor and one of the most incurable forms of cancer in need of new therapeutic targets. As some cancers are known to be caused by a virus, the discovery of viruses could open the possibility to treat, and perhaps prevent, such a disease. Although an association with viruses such as cytomegalovirus or Simian virus 40 has been strongly suggested, involvement of these and other viruses in the initiation and/or propagation of glioblastoma remains vague, controversial and warrants elucidation. To exhaustively address the association of virus and glioblastoma, we developed and validated a robust metagenomic approach to analyze patient biopsies via high-throughput sequencing, a sensitive tool for virus screening. In addition to traditional clinical diagnostics, glioblastoma biopsies were deep-sequenced and analyzed with a multistage computational pipeline to identify known or potentially discover unknown viruses. In contrast to the studies reporting the presence of viral signatures in glioblastoma, no common or recurring active viruses were detected, despite finding an antiviral-like type I interferon response in some specimens. Our findings highlight a discrete and non-specific viral signature and uncharacterized short RNA sequences in glioblastoma. This study provides new insights into glioblastoma pathogenesis and defines a general methodology that can be used for high-resolution virus screening and discovery in human cancers.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/virology , Cytomegalovirus/immunology , Glioblastoma/genetics , Glioblastoma/virology , Interferon Type I/immunology , Antibodies, Viral/blood , Brain Neoplasms/immunology , High-Throughput Nucleotide Sequencing , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Metagenomics
17.
Cell Rep Med ; 5(4): 101503, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38593810

ABSTRACT

In monogenic autoinflammatory diseases, mutations in genes regulating innate immune responses often lead to uncontrolled activation of inflammasome pathways or the type I interferon (IFN-I) response. We describe a mechanism of autoinflammation potentially predisposing patients to life-threatening necrotizing soft tissue inflammation. Six unrelated families are identified in which affected members present with necrotizing fasciitis or severe soft tissue inflammations. Exome sequencing reveals truncating monoallelic loss-of-function variants of nuclear factor κ light-chain enhancer of activated B cells (NFKB1) in affected patients. In patients' macrophages and in NFKB1-variant-bearing THP-1 cells, activation increases both interleukin (IL)-1ß secretion and IFN-I signaling. Truncation of NF-κB1 impairs autophagy, accompanied by the accumulation of reactive oxygen species and reduced degradation of inflammasome receptor nucleotide-binding oligomerization domain, leucine-rich repeat-containing protein 3 (NLRP3), and Toll/IL-1 receptor domain-containing adaptor protein inducing IFN-ß (TRIF), thus leading to combined excessive inflammasome and IFN-I activity. Many of the patients respond to anti-inflammatory treatment, and targeting IL-1ß and/or IFN-I signaling could represent a therapeutic approach for these patients.


Subject(s)
Fasciitis, Necrotizing , Interferon Type I , Humans , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Immunity, Innate , Inflammation/metabolism , NF-kappa B p50 Subunit
18.
Biomed Pharmacother ; 169: 115883, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37979373

ABSTRACT

The stimulator of the interferon gene (STING) signaling pathway acts as a primary defense system against DNA pathogens. Because of the crucial role of STING in type I interferon (IFN) response and innate immunity, extensive research has been conducted to elucidate the roles of various effector molecules involved in STING-mediated signal transduction. However, despite the substantial contribution of microtubules to the immune system, the association between the STING signaling pathway and microtubules remains unclear. In this study, we revealed that the modulation of STING via microtubule-destabilizing agents (MDAs) specifically induced type I IFN responses rather than inflammatory responses in human monocytes. Co-treatment of MDAs with STING agonists induced the elevation of phospho-TANK-binding kinase 1 (TBK1), amplifying the innate immune response. However, during the deficiency of TBK1, the non-canonical signaling pathway through nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) contributed to MDA-induced STING activation in type I IFN response which suggested the versatile regulation of MDA in STING-mediated immunity.


Subject(s)
Interferon Type I , Monocytes , Humans , Immunity, Innate/physiology , Interferon Type I/metabolism , Membrane Proteins/metabolism , Monocytes/metabolism , NF-kappa B/metabolism , Signal Transduction/physiology
19.
mBio ; 14(5): e0195023, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37773002

ABSTRACT

IMPORTANCE: In order to efficiently produce infectious viral particles, HIV must counter several restrictions exerted by host cell antiviral proteins. MARCH1 is a member of the MARCH protein family that restricts HIV infection by limiting the incorporation of viral envelope glycoproteins into nascent virions. Here, we identified two regulatory RNAs, microRNAs-25 and -93, induced by the HIV-1 accessory protein Vpu, that downregulate MARCH1 mRNA. We also show that Vpu induces these cellular microRNAs in macrophages by hijacking the cellular ß-catenin pathway. The notion that HIV-1 has evolved a mechanism to counteract MARCH1 restriction on viral infectivity underlines the importance of MARCH1 in the host antiviral response.


Subject(s)
HIV Infections , HIV Seropositivity , HIV-1 , MicroRNAs , Humans , HIV Infections/metabolism , HIV-1/physiology , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism , Human Immunodeficiency Virus Proteins/genetics , Antiviral Agents/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Macrophages/metabolism , GPI-Linked Proteins/metabolism
20.
Front Immunol ; 14: 1146699, 2023.
Article in English | MEDLINE | ID: mdl-37275914

ABSTRACT

Introduction: Transcriptional activation depends on the interplay of chromatin modifiers to establish a permissive epigenetic landscape. While histone 3 lysine 9 (H3K9) methylation has long been associated with gene repression, there is limited evidence to support a role for H3K9 demethylases in gene activation. Methods: We leveraged knockdown and overexpression of JMJD2d / Kdm4d in mouse embryonic fibroblasts, coupled with extensive epigenomic analysesm to decipher the role of histone 3 lysine 9 demethylases in the innate immune response. Results: Here we describe the H3K9 demethylase Kdm4d/JMJD2d as a positive regulator of type I interferon responses. In mouse embryonic fibroblasts (MEFs), depletion of JMJD2d attenuates the transcriptional response, conferring increased viral susceptibility, while overexpression of the demethylase results in more robust IFN activation. We find that the underlying mechanism of JMJD2d in type I interferon responses consists of an effect both on the transcription of enhancer RNAs (eRNAs) and on dynamic H3K9me2 at associated promoters. In support of these findings, we establish that JMJD2d is associated with enhancer regions throughout the genome prior to stimulation but is redistributed to inducible promoters in conjunction with transcriptional activation. Discussion: Taken together, our data reveal JMJD2d as a chromatin modifier that connects enhancer transcription with promoter demethylation to modulate transcriptional responses.


Subject(s)
Histone Demethylases , Interferon Type I , Animals , Mice , Histone Demethylases/genetics , Histone Demethylases/metabolism , Histones/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Interferon Type I/genetics , Lysine/genetics , Fibroblasts/metabolism , Chromatin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL