Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters

Publication year range
1.
J Virol ; 97(3): e0197722, 2023 03 30.
Article in English | MEDLINE | ID: mdl-36815839

ABSTRACT

African swine fever (ASF) is an acute and severe infectious disease caused by the ASF virus (ASFV). The mortality rate of ASF in pigs can reach 100%, causing huge economic losses to the pig industry. Here, we found that ASFV protein MGF505-7R inhibited the beta interferon (IFN-ß)-mediated Janus-activated kinase-signal transducer and activation of transcription (JAK-STAT) signaling. Our results demonstrate that MGF505-7R inhibited interferon-stimulated gene factor 3 (ISGF3)-mediated IFN-stimulated response element (ISRE) promoter activity. Importantly, we observed that MGF505-7R inhibits ISGF3 heterotrimer formation by interacting with interferon regulatory factor 9 (IRF9) and inhibits the nuclear translocation of ISGF3. Moreover, to demonstrate the role of MGF505-7R in IFN-I signal transduction during ASFV infection, we constructed and evaluated ASFV-ΔMGF505-7R recombinant viruses. ASFV-ΔMGF505-7R restored STAT2 and STAT1 phosphorylation, alleviated the inhibition of ISGF3 nuclear translocation, and showed increased susceptibility to IFN-ß, unlike the parental GZ201801 strain. In conclusion, our study shows that ASFV protein MGF505-7R plays a key role in evading IFN-I-mediated innate immunity, revealing a new mode of evasion for ASFV. IMPORTANCE ASF, caused by ASFV, is currently prevalent in Eurasia, with mortality rates reaching 100% in pigs. At present, there are no safe or effective vaccines against ASFV. In this study, we found that the ASFV protein MGF505-7R hinders IFN-ß signaling by interacting with IRF9 and inhibiting the formation of ISGF3 heterotrimers. Of note, we demonstrated that MGF505-7R plays a role in the immune evasion of ASFV in infected hosts and that recombinant viruses alleviated the effect on type I IFN (IFN-I) signaling and exhibited increased susceptibility to IFN-ß. This study provides a theoretical basis for developing vaccines against ASFV using strains with MGF505-7R gene deletions.


Subject(s)
African Swine Fever Virus , African Swine Fever , Interferon Type I , Interferon-Stimulated Gene Factor 3, gamma Subunit , Virus Replication , Animals , African Swine Fever/immunology , African Swine Fever/virology , African Swine Fever Virus/genetics , African Swine Fever Virus/immunology , Immunity, Innate , Interferon Type I/immunology , Interferon-Stimulated Gene Factor 3, gamma Subunit/immunology , Signal Transduction , Swine , Viral Proteins/genetics , Viral Proteins/immunology , Virus Replication/physiology , Active Transport, Cell Nucleus/genetics , Immune Evasion/genetics
2.
J Virol ; 97(10): e0072723, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37819133

ABSTRACT

IMPORTANCE: Type I interferon (IFN) signaling plays a principal role in host innate immune responses against invading viruses. Viruses have evolved diverse mechanisms that target the Janus kinase-signal transducer and activator of transcription (STAT) signaling pathway to modulate IFN response negatively. Seneca Valley virus (SVV), an emerging porcine picornavirus, has received great interest recently because it poses a great threat to the global pork industry. However, the molecular mechanism by which SVV evades host innate immunity remains incompletely clear. Our results revealed that SVV proteinase (3Cpro) antagonizes IFN signaling by degrading STAT1, STAT2, and IRF9, and cleaving STAT2 to escape host immunity. SVV 3Cpro also degrades karyopherin 1 to block IFN-stimulated gene factor 3 nuclear translocation. Our results reveal a novel molecular mechanism by which SVV 3Cpro antagonizes the type I IFN response pathway by targeting STAT1-STAT2-IRF9 and karyopherin α1 signals, which has important implications for our understanding of SVV-evaded host innate immune responses.


Subject(s)
3C Viral Proteases , Interferon Type I , Picornaviridae , Animals , Host-Pathogen Interactions , Interferon Type I/metabolism , Karyopherins , Picornaviridae/metabolism , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , Swine , 3C Viral Proteases/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , alpha Karyopherins/metabolism , Signal Transduction
3.
Fish Shellfish Immunol ; 153: 109857, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39182707

ABSTRACT

The major histocompatibility complex class II (MHCII) molecules are crucial elements of the adaptive immune system, essential for orchestrating immune responses against foreign pathogens. However, excessive expression of MHCII can disrupt normal physiological functions. Therefore, the host employs various mechanisms to regulate MHCII expression and maintain immune homeostasis. Despite this importance, limited studies have explored the negative regulation of MHCII transcription in bony fish. In this study, we found that interferon h (IFNh), a subtype of type I IFN in sea perch Lateolabrax japonicus, could inhibit the activation of IFNγ induced-MHCII expression by modulating the transcription of the class II major histocompatibility complex transactivator (CIITA). Transcriptome analysis revealed 57 up-regulated and 69 down-regulated genes in cells treated with both IFNγ and IFNh compared to those treated with IFNγ alone. To maintain cellular homeostasis, interferon regulatory factor 9 (IRF9) was up-regulated following IFNγ stimulation, thereby preventing MHCII overexpression. Mechanistically, IRF9 bound to the CIITA promoter and suppressed its expression activated by IRF1. Furthermore, IRF9 inhibited the promoter activity of both MHCII-α and MHCII-ß induced by CIITA. Our findings highlight the roles of IFNh and IRF9 as suppressors regulating MHCII expression at different hierarchical levels. This study provides insights into the intricate regulation of antigen presentation and the foundation for further exploration of the interaction mechanisms between aquatic virus and fish.


Subject(s)
Fish Proteins , Interferon-gamma , Animals , Fish Proteins/genetics , Fish Proteins/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Gene Expression Regulation/immunology , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Interferon-Stimulated Gene Factor 3, gamma Subunit/immunology , Immunity, Innate/genetics , Gene Expression Profiling/veterinary , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Nuclear Proteins , Trans-Activators
4.
Eur J Immunol ; 52(1): 138-148, 2022 01.
Article in English | MEDLINE | ID: mdl-34676541

ABSTRACT

The interferon pathway, a key antiviral defense mechanism, is being considered as a therapeutic target in COVID-19. Both, substitution of interferon and JAK/STAT inhibition to limit cytokine storms have been proposed. However, little is known about possible abnormalities in STAT signaling in immune cells during SARS-CoV-2 infection. We investigated downstream targets of interferon signaling, including STAT1, STAT2, pSTAT1 and 2, and IRF1, 7 and 9 by flow cytometry in 30 patients with COVID-19, 17 with mild, and 13 with severe infection. We report upregulation of STAT1 and IRF9 in mild and severe COVID-19 cases, which correlated with the IFN-signature assessed by Siglec-1 (CD169) expression on peripheral monocytes. Interestingly, Siglec-1 and STAT1 in CD14+ monocytes and plasmablasts showed lower expression among severe cases compared to mild cases. Contrary to the baseline STAT1 expression, the phosphorylation of STAT1 was enhanced in severe COVID-19 cases, indicating a dysbalanced JAK/STAT signaling that fails to induce transcription of interferon stimulated response elements (ISRE). This abnormality persisted after IFN-α and IFN-γ stimulation of PBMCs from patients with severe COVID-19. Data suggest impaired STAT1 transcriptional upregulation among severely infected patients may represent a potential predictive biomarker and would allow stratification of patients for certain interferon-pathway targeted treatments.


Subject(s)
COVID-19/immunology , Monocytes/immunology , SARS-CoV-2/immunology , STAT1 Transcription Factor/immunology , Signal Transduction/immunology , Up-Regulation/immunology , Adult , Aged , Female , Humans , Interferon Regulatory Factors/immunology , Male , Middle Aged , Patient Acuity , Phosphorylation/immunology
5.
J Virol ; 96(13): e0217121, 2022 07 13.
Article in English | MEDLINE | ID: mdl-35708311

ABSTRACT

The alphaherpesvirus pseudorabies virus (PRV) is the etiologic agent of swine Aujeszky's disease, which can cause huge economic losses to the pig industry. PRV can overcome a type I interferon (IFN)-induced antiviral state in host cells through its encoded EP0 protein. However, the exact role of EP0 in this process is poorly defined. Here, we report that EP0 transcriptionally represses IFN regulatory factor 9 (IRF9), a critical component in the IFN signaling pathway, thereby reducing the cellular levels of IRF9 and inhibiting IFN-induced gene transcription. This activity of EP0 is mediated by its C-terminal region independently of the RING domain. Moreover, compared with EP0 wild-type PRV, EP0-deficient PRV loses the ability to efficiently decrease cellular IRF9, while reintroducing the C-terminal region of EP0 back into the EP0-deficient virus restores the activity. Together, these results suggest that EP0 can transcriptionally modulate IRF9-mediated antiviral pathways through its C-terminal region, contributing to PRV innate immune evasion. IMPORTANCE Alphaherpesviruses can establish lifelong infections and cause many diseases in humans and animals. Pseudorabies virus (PRV) is a swine alphaherpesvirus that threatens pig production. Using PRV as a model, we found that alphaherpesvirus can utilize its encoded early protein EP0 to inhibit the IFN-induced upregulation of antiviral proteins by reducing the basal expression levels of IRF9 through repressing its transcription. Our findings reveal a mechanism employed by alphaherpesvirus to evade the immune response and indicate that EP0 is an important viral protein in pathogenesis and a potential target for antiviral drug development.


Subject(s)
Herpesvirus 1, Suid , Interferon Type I , Interferon-Stimulated Gene Factor 3, gamma Subunit , Pseudorabies , Swine Diseases , Animals , Antiviral Agents/pharmacology , Gene Expression Regulation/immunology , Herpesvirus 1, Suid/immunology , Herpesvirus 1, Suid/metabolism , Host Microbial Interactions/immunology , Interferon Type I/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Pseudorabies/immunology , Pseudorabies/virology , Swine , Swine Diseases/immunology , Swine Diseases/virology , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/metabolism
6.
J Virol ; 96(17): e0094422, 2022 09 14.
Article in English | MEDLINE | ID: mdl-35972295

ABSTRACT

African swine fever virus (ASFV) is the etiological agent of a highly lethal hemorrhagic disease in domestic pigs and wild boars that has significant economic consequences for the pig industry. The type I interferon (IFN) signaling pathway is a pivotal component of the innate antiviral response, and ASFV has evolved multiple mechanisms to antagonize this pathway and facilitate infection. Here, we reported a novel function of ASFV pI215L in inhibiting type I IFN signaling. Our results showed that ASFV pI215L inhibited IFN-stimulated response element (ISRE) promoter activity and subsequent transcription of IFN-stimulated genes (ISGs) by triggering interferon regulatory factor 9 (IRF9) degradation. Additionally, we found that catalytically inactive pI215L mutations retained the ability to block type I IFN signaling, indicating that this only known viral E2 ubiquitin-conjugating enzyme mediates IFR9 degradation in a ubiquitin-conjugating activity-independent manner. By coimmunoprecipitation, confocal immunofluorescence, and subcellular fractionation approaches, we demonstrated that pI215L interacted with IRF9 and impaired the formation and nuclear translocation of IFN-stimulated gene factor 3 (ISGF3). Moreover, further mechanism studies supported that pI215L induced IRF9 degradation through the autophagy-lysosome pathway in both pI215L-overexpressed and ASFV-infected cells. These findings reveal a new immune evasion strategy evolved by ASFV in which pI215L acts to degrade host IRF9 via the autophagic pathway, thus inhibiting the type I IFN signaling and counteracting the host innate immune response. IMPORTANCE African swine fever virus (ASFV) causes a highly contagious and lethal disease in pigs and wild boars that is currently present in many countries, severely affecting the global pig industry. Despite extensive research, effective vaccines and antiviral strategies are still lacking, and many fundamental questions regarding the molecular mechanisms underlying host innate immunity escape remain unclear. In this study, we identified ASFV pI215L, the only known viral E2 ubiquitin-conjugating enzyme, which is involved in antagonizing the type I interferon signaling. Mechanistically, pI215L interacted with interferon regulatory factor 9 for autophagic degradation, and this degradation was independent of its ubiquitin-conjugating activity. These results increase the current knowledge regarding ASFV evasion of innate immunity, which may instruct future research on antiviral strategies and dissection of ASFV pathogenesis.


Subject(s)
African Swine Fever , Autophagy , Interferon Type I , Interferon-Stimulated Gene Factor 3, gamma Subunit , African Swine Fever/immunology , African Swine Fever Virus , Animals , Immunity, Innate , Interferon Type I/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Signal Transduction , Sus scrofa , Swine , Ubiquitin-Conjugating Enzymes/metabolism
7.
J Allergy Clin Immunol ; 150(4): 955-964.e16, 2022 10.
Article in English | MEDLINE | ID: mdl-35182547

ABSTRACT

BACKGROUND: Inflammatory phenomena such as hyperinflammation or hemophagocytic lymphohistiocytosis are a frequent yet paradoxical accompaniment to virus susceptibility in patients with impairment of type I interferon (IFN-I) signaling caused by deficiency of signal transducer and activator of transcription 2 (STAT2) or IFN regulatory factor 9 (IRF9). OBJECTIVE: We hypothesized that altered and/or prolonged IFN-I signaling contributes to inflammatory complications in these patients. METHODS: We explored the signaling kinetics and residual transcriptional responses of IFN-stimulated primary cells from individuals with complete loss of one of STAT1, STAT2, or IRF9 as well as gene-edited induced pluripotent stem cell-derived macrophages. RESULTS: Deficiency of any IFN-stimulated gene factor 3 component suppressed but did not abrogate IFN-I receptor signaling, which was abnormally prolonged, in keeping with insufficient induction of negative regulators such as ubiquitin-specific peptidase 18 (USP18). In cells lacking either STAT2 or IRF9, this late transcriptional response to IFN-α2b mimicked the effect of IFN-γ. CONCLUSION: Our data suggest a model wherein the failure of negative feedback of IFN-I signaling in STAT2 and IRF9 deficiency leads to immune dysregulation. Aberrant IFN-α receptor signaling in STAT2- and IRF9-deficient cells switches the transcriptional output to a prolonged, IFN-γ-like response and likely contributes to clinically overt inflammation in these individuals.


Subject(s)
Interferon Type I , Factor IX , Humans , Interferon Type I/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Interferon-alpha , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/genetics , Ubiquitin Thiolesterase , Ubiquitin-Specific Proteases
8.
Int J Mol Sci ; 24(24)2023 Dec 18.
Article in English | MEDLINE | ID: mdl-38139463

ABSTRACT

In addition to the canonical ISGF3 and non-canonical STAT2/IRF9 complexes, evidence is emerging of the role of their unphosphorylated counterparts in IFN-dependent and -independent ISG transcription. To better understand the relation between ISGF3 and U-ISGF3 and STAT2/IRF9 and U-STAT2/IRF9 in IFN-I-stimulated transcriptional responses, we performed RNA-Seq and ChIP-Seq, in combination with phosphorylation inhibition and antiviral experiments. First, we identified a group of ISRE-containing ISGs that were commonly regulated in IFNα-treated WT and STAT1-KO cells. Thus, in 2fTGH and Huh7.5 WT cells, early and long-term IFNα-inducible transcription and antiviral activity relied on the DNA recruitment of the ISGF3 components STAT1, STAT2 and IRF9 in a phosphorylation- and time-dependent manner. Likewise, in ST2-U3C and Huh-STAT1KO cells lacking STAT1, delayed IFN responses correlated with DNA binding of phosphorylated STAT2/IRF9 but not U-STAT2/IRF9. In addition, comparative experiments in U3C (STAT1-KO) cells overexpressing all the ISGF3 components (ST1-ST2-IRF9-U3C) revealed U-ISGF3 (and possibly U-STAT2/IRF9) chromatin interactions to correlate with phosphorylation-independent ISG transcription and antiviral activity. Together, our data point to the dominant role of the canonical ISGF3 and non-canonical STAT2/IRF9, without a shift to U-ISGF3 or U-STAT2/IRF9, in the regulation of early and prolonged ISG expression and viral protection. At the same time, they suggest the threshold-dependent role of U-ISFG3, and potentially U-STAT2/IRF9, in the regulation of constitutive and possibly long-term IFNα-dependent responses.


Subject(s)
Interferon Type I , Interferon-Stimulated Gene Factor 3 , Interleukin-1 Receptor-Like 1 Protein , STAT2 Transcription Factor , Antiviral Agents/pharmacology , DNA/pharmacology , Immunoglobulins/metabolism , Interferon Type I/metabolism , Interleukin-1 Receptor-Like 1 Protein/metabolism , Signal Transduction , STAT1 Transcription Factor/metabolism , Interferon-Stimulated Gene Factor 3/metabolism , STAT2 Transcription Factor/metabolism , Humans
9.
Proc Natl Acad Sci U S A ; 115(15): 3906-3911, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29581268

ABSTRACT

In response to IFNß, the IL6 gene is activated, modestly at early times by ISGF3 (IRF9 plus tyrosine-phosphorylated STATs 1 and 2), and strongly at late times by U-ISGF3 (IRF9 plus U-STATs 1 and 2, lacking tyrosine phosphorylation). A classical IFN-stimulated response element (ISRE) at -1,513 to -1,526 in the human IL6 promoter is required. Pretreating cells with IFNß or increasing the expression of U-STAT2 and IRF9 exogenously greatly enhances IL6 expression in response to the classical NF-κB activators IL1, TNF, and LPS. U-STAT2 binds tightly to IRF9, the DNA binding subunit of ISGF3, and also to the p65 subunit of NF-κB. Therefore, as shown by ChIP analyses, U-STAT2 can bridge the ISRE and κB elements in the IL6 promoter. In some cancer cells, the protumorigenic activation of STAT3 will be enhanced by the increased synthesis of IL6 that is facilitated by high expression of U-STAT2 and IRF9.


Subject(s)
Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interleukin-6/genetics , NF-kappa B/metabolism , STAT2 Transcription Factor/metabolism , Gene Expression Regulation , Humans , Interferon-beta/genetics , Interferon-beta/metabolism , Interleukin-6/metabolism , NF-kappa B/genetics , Phosphorylation , Promoter Regions, Genetic , Response Elements , STAT2 Transcription Factor/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Up-Regulation
10.
Cells Tissues Organs ; 209(2-3): 110-119, 2020.
Article in English | MEDLINE | ID: mdl-32772027

ABSTRACT

OBJECTIVE: To discuss how IRF9 affects the fibroblast-like synoviocytes (FLS) in TNF-induced rheumatoid arthritis (RA) via the SIRT-1/NF-κB signaling pathway. METHODS: RA-FLS were isolated and divided into control, sh-IRF9, TNF, TNF + sh-Ctrl, TNF + sh-IRF9, TNF + sh-SIRT1, and TNF + sh-IRF9 + sh-SIRT1 groups. Biological features of FLS were evaluated by MTT, wound healing, and Transwell assays, respectively. Cell apoptosis and cycle were assessed flow cytometrically. Inflammatory cytokines were determined through enzyme-linked immunosorbent assay (ELISA), while IRF9 expression and SIRT1/NF-κB signaling pathway activity were measured by Western blotting. RESULTS: TNF increased IRF9 expression as well as NF-κB signaling activity and down-regulated SIRT1 of RA-FLS. Silencing IRF9 resulted in up-regulation of SIRT1 and blocked NF-κB signaling, with significant decreases in TNF-induced cell viability, migration, and invasion, prominent enhancement in apoptosis and the proportion of cells in G0/G1 phase, but a decrease in the proportion of cells in S and G2/M phases, and reduced levels of inflammatory cytokines. However, these changes were totally abolished after silencing SIRT1, i.e., the IRF9 shRNA-induced inhibitory effect on the growth of RA-FLS was reversed. CONCLUSION: Silencing IRF9 curbs the activity of the NF-κB signaling pathway via up-regulating SIRT-1, to further suppress TNF-induced changes in the malignant features of RA-FLS, and the secretion of inflammatory cytokines, with the promoted apoptosis.


Subject(s)
Arthritis, Rheumatoid/pathology , Fibroblasts/pathology , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , NF-kappa B/metabolism , Signal Transduction , Sirtuin 1/metabolism , Synoviocytes/pathology , Tumor Necrosis Factor-alpha/adverse effects , Cell Cycle , Cell Movement , Female , Humans , Inflammation Mediators/metabolism , Male , Middle Aged , Phenotype
11.
Mol Cell Biochem ; 472(1-2): 125-134, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32577948

ABSTRACT

Acute pancreatitis (AP) is an inflammatory disease caused by the abnormal activation of pancreatic enzymes in the pancreas, with a considerably high morbidity and mortality. However, the etiological factor and pathogenesis of AP are still unclear. This study was aimed to explore the role and mechanism of interferon regulatory factor 9 (IRF9) in the occurrence of AP and to provide experimental and theoretical foundation for AP diagnosis and treatment. AP model in vitro was established by caerulein-induced group. Small interfering RNA (siRNA) was designed and constructed to silence IRF9 gene. After siRNA transfected and caerulein treated successfully, the expression levels of IRF9, SIRT1, and acetylated p53 (Ac-p53) were determined by qRT-PCR and Western blot. The apoptosis, proliferation, and migration of AR42J cells were checked by flow cytometry, MTT, and transwell assay. Dual-luciferase reporter assay was implemented to validate the regulatory effect of IRF9 on SIRT1. Here, our study showed that the expression of IRF9 and Ac-p53 was increased, SIRT1 was decreased, and cell apoptosis, proliferation, and migration of AR42J cells were increased after caerulein induced. IRF9 gene silencing upregulated SIRT1, downregulated Ac-p53, and inhibited cell apoptosis, proliferation, and migration. Dual-Luciferase reporter assay showed that IRF9 could negatively regulate SIRT1. The potential mechanism was that IRF9 could modulate cell apoptosis, proliferation, migration, and bind the promoter of SIRT1 to repress SIRT1-p53. It hinted that IRF9 showed a novel function in AP by modulating cell apoptosis, proliferation, migration, and suppressing SIRT1-p53. IRF9 might be a good potential treatment target for AP.


Subject(s)
Apoptosis , Cell Movement , Cell Proliferation , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Pancreatitis/pathology , Sirtuin 1/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Biomarkers/metabolism , Cells, Cultured , Gene Expression Regulation , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Pancreatitis/genetics , Pancreatitis/metabolism , Rats , Sirtuin 1/genetics , Tumor Suppressor Protein p53/genetics
12.
Fish Shellfish Immunol ; 103: 220-228, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32439513

ABSTRACT

As a NAD+-dependent deacetylase, SIRT1 is widely involved in apoptosis and cellular inflammation via multiple pathways such as p53, NF-кB and STAT. More and more studies have shown that p53 is the first non-histone deacetylation target of SIRT1. SIRT1-p53 axis thus plays an important role in mammalian cells. IRF9 is an important member of interferon regulator factor family and performs an important role in innate immunity against foreign virus invasion. More importantly, human IRF9 can suppress the SIRT1-p53 axis. However, the functions and relationship between IRF9 and SIRT1-p53 axis are rarely studied in fish. To this end, we made a preliminary research on the functions of grass carp (Ctenopharyngodon idella) IRF9, SIRT1 and p53 in apoptosis and innate immunity. Firstly, we cloned and identified the ORF of SIRT1 (named CiSIRT1, MN125614) from C. idella and demonstrated that CiIRF9 promoted apoptosis, while CiSIRT1 inhibited apoptosis by flow cytometry and TUNEL experiments. Next, we found the interaction between CiSIRT1 and Cip53 in vivo by co-immunoprecipitation experiments. Moreover, the colocalization analysis also showed CiSIRT1 and Cip53 were mainly distributed in nucleus. Thirdly, we got a conclusion that CiIRF9 can repress the expression of CiSIRT1, implying that CiIRF9 regulates CiSIRT1-p53 axis. Finally, CiSIRT1 mRNA level was significantly up-regulated and the expression reached the highest level at 24 h post poly (I:C) stimulation in CIK cells. So, CiSIRT1 may exert an important function in innate immunity. Furthermore, we found CiSIRT1 down-regulated the expression of CiIFN1. In summary, CiIRF9 promotes apoptosis and innate immunity by inhibiting SIRT1-p53 axis. These findings will provide a new theoretical basis for the research on teleost innate immunity.


Subject(s)
Apoptosis/genetics , Carps/immunology , Fish Proteins/immunology , Immunity, Innate/genetics , Interferon-Stimulated Gene Factor 3, gamma Subunit/immunology , Sirtuin 1/immunology , Tumor Suppressor Protein p53/immunology , Animals , Carps/genetics , Fish Proteins/genetics , Gene Expression Regulation/immunology , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Sirtuin 1/genetics , Tumor Suppressor Protein p53/genetics
13.
Int J Mol Sci ; 21(17)2020 Aug 27.
Article in English | MEDLINE | ID: mdl-32867271

ABSTRACT

ADAR (adenosine deaminase acting on RNA) catalyzes the deamination of adenosine to generate inosine, through its binding to double-stranded RNA (dsRNA), a phenomenon known as RNA editing. One of the functions of ADAR1 is suppressing the type I interferon (IFN) response, but its mechanism in gastric cancer is not clearly understood. We analyzed changes in RNA editing and IFN signaling in ADAR1-depleted gastric cancer cells, to clarify how ADAR1 regulates IFN signaling. Interestingly, we observed a dramatic increase in the protein level of signal transducer and activator of transcription 1 (STAT1) and interferon regulatory factor 9 (IRF9) upon ADAR1 knockdown, in the absence of type I or type II IFN treatment. However, there were no changes in protein expression or localization of the mitochondrial antiviral signaling protein (MAVS) and interferon alpha and beta-receptor subunit 2 (IFNAR2), the two known mediators of IFN production. Instead, we found that miR-302a-3p binds to the untranslated region (UTR) of IRF9 and regulate its expression. The treatment of ADAR1-depleted AGS cells with an miR-302a mimic successfully restored IRF9 as well as STAT1 protein level. Hence, our results suggest that ADAR1 regulates IFN signaling in gastric cancer through the suppression of STAT1 and IRF9 via miR-302a, which is independent from the RNA editing of known IFN production pathway.


Subject(s)
Adenosine Deaminase/genetics , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interferons/metabolism , MicroRNAs/genetics , RNA-Binding Proteins/genetics , STAT2 Transcription Factor/metabolism , Stomach Neoplasms/genetics , 3' Untranslated Regions , Adaptor Proteins, Signal Transducing/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HEK293 Cells , Humans , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , RNA Editing , Receptor, Interferon alpha-beta/metabolism , Signal Transduction , Stomach Neoplasms/metabolism
14.
J Virol ; 92(21)2018 11 01.
Article in English | MEDLINE | ID: mdl-30089701

ABSTRACT

Both type I and type II interferons (IFNs) have been implicated in the host defense against varicella-zoster virus (VZV), a common human herpesvirus that causes varicella and zoster. The purpose of this study was to compare their contributions to the control of VZV replication, to identify the signaling pathways that are critical for mediating their antiviral activity, and to define the mechanisms by which the virus counteracts their effects. Gamma interferon (IFN-γ) was much more potent than IFN-α in blocking VZV infection, which was associated with a differential induction of the interferon regulatory factor (IRF) proteins IRF1 and IRF9, respectively. These observations account for the clinical experience that while the formation of VZV skin lesions is initially controlled by local immunity, adaptive virus-specific T cell responses are required to prevent life-threatening VZV infections.IMPORTANCE While both type I and type II IFNs are involved in the control of herpesvirus infections in the human host, to our knowledge, their relative contributions to the restriction of viral replication and spread have not been assessed. We report that IFN-γ has more potent activity than IFN-α against VZV. Findings from this comparative analysis show that the IFN-α-IRF9 axis functions as a first line of defense to delay the onset of viral replication and spread, whereas the IFN-γ-IRF1 axis has the capacity to block the infectious process. Our findings underscore the importance of IRFs in IFN regulation of herpesvirus infection and account for the clinical experience of the initial control of VZV skin infection attributable to IFN-α production, together with the requirement for induction of adaptive IFN-γ-producing VZV-specific T cells to resolve the infection.


Subject(s)
Herpesvirus 3, Human/immunology , Interferon Regulatory Factor-1/immunology , Interferon-Stimulated Gene Factor 3, gamma Subunit/immunology , Interferon-alpha/immunology , Interferon-gamma/immunology , Varicella Zoster Virus Infection/immunology , Cell Line, Tumor , HEK293 Cells , Humans , STAT1 Transcription Factor/immunology , STAT2 Transcription Factor/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Varicella Zoster Virus Infection/virology , Virus Replication/immunology
15.
Fish Shellfish Immunol ; 88: 606-618, 2019 May.
Article in English | MEDLINE | ID: mdl-30885743

ABSTRACT

SAMHD1 is an innate immunity restriction factor that inhibits virus infection through IRF3-mediated antiviral and apoptotic responses. Fish SAMHD1 shares some similar properties with those in mammals. In this study, a SAMHD1 orthologue from grass carp (Ctenopharyngodon idellus) was cloned and characterized. The full-length cDNA of CiSAMHD1 is 2792 bp with an ORF of 1884 bp encoding a polypeptide of 627 amino acids. Multiple alignments showed that SAMHD1 is highly conserved among different species. Phylogenetic tree analysis revealed that CiSAMHD1 shared a high degree of homology with Sinocyclocheilus rhinocerous SAMHD1. Expression analysis indicated that CiSAMHD1 was widely expressed in all tissues tested including the brain, eyes, spleen, gill, intestine, liver, heart and kidney. It was significantly up-regulated in spleen, liver and intestines after treatment with poly I:C. Also, CiSAMHD1 can be induced following stimulation with recombinant IFN in CIK cells. The promoter sequence of CiSAMHD1 was identified to explore the mechanism underlying the transcriptional regulation of CiSAMHD1. The promoter sequence of CiSAMHD1 (1370 bp) consists of IRF1, IRF3, IRF9 and p65 binding elements. Gel mobility shift assay also showed that IRF1, IRF3, IRF9 and p65 prokaryotic proteins can separately interact with CiSAMHD1 promoter. Dual luciferase assay and q-PCR suggested that the promoter of CiSAMHD1 can be activated by the overexpression of CiIRF3 and CiIRF9, but cannot be triggered by CiIRF1 and Cip65. In contrast, knockdown of CiIRF3 or CiIRF9 inhibits the transcription of CiSAMHD1. Intriguingly, CCK assay suggested that CiSAMHD1 decreased cell viability. TUNEL apoptosis assay and Hoechst 33258 staining assay indicated that apoptosis is induced by the overexpression of CiSAMHD1. Crystal violet staining, detection of two GCRV genes (vp3 and vp5) and viral titration showed that CiSAMHD1 can suppress the proliferation of grass carp reovirus (GCRV) in CIK cells.


Subject(s)
Apoptosis , Carps/genetics , Fish Proteins/genetics , Orthoreovirus/physiology , SAM Domain and HD Domain-Containing Protein 1/genetics , Virus Replication , Animals , Cell Proliferation , Cloning, Molecular , DNA, Complementary/genetics , Fish Proteins/metabolism , Gene Expression Regulation , Immunity, Innate , Interferon Regulatory Factor-3/genetics , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Promoter Regions, Genetic , Reoviridae Infections , SAM Domain and HD Domain-Containing Protein 1/metabolism
16.
Exp Cell Res ; 365(2): 185-193, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29501566

ABSTRACT

Acute myeloid leukemia (AML) is a highly heterogeneous disease, with biologically and prognostically different subtypes. Although a growing number of distinct AML subsets have been increasingly characterized, patient management has remained disappointingly uniform. The molecular mechanism underlying AML needs to be further investigated. Here we identify IRF9 as a negative regulator of human AML. We show that IRF9 mRNA and protein levels are down-regulated in human AML samples compared with samples from healthy donors. IRF9 knockdown promotes proliferation, colony formation and survival of OCI/AML-2 and OCI/AML-3 cells, whereas IRF9 overexpression obtains oppose results. Mechanism analysis shows that IRF9 binds SIRT1 promoter and represses SIRT1 expression in OCI/AML-2 and OCI/AML-3 cells. In AML samples, the expression of SIRT1 is up-regulated and negatively correlated with IRF9 level. IRF9 also increases the acetylation of p53, a deacetylation substrate of SIRT1, and promotes the expression of p53 target genes. Knockdown of p53 blocks the effects of IRF9 on cell survival and growth in vitro. These findings provide evidence that IRF9 serves as an important regulator in human AML by repressing SIRT1-p53 pathway and that IRF9 may be a potential target for AML treatment.


Subject(s)
Cell Proliferation , Interferon-Stimulated Gene Factor 3, gamma Subunit/physiology , Leukemia, Myeloid, Acute/pathology , Sirtuin 1/physiology , Tumor Suppressor Protein p53/physiology , Case-Control Studies , Cell Proliferation/genetics , Cell Survival/genetics , Cells, Cultured , Gene Expression Regulation, Leukemic , HEK293 Cells , Humans , Leukemia, Myeloid, Acute/genetics , Signal Transduction/physiology
17.
Biochem Biophys Res Commun ; 507(1-4): 330-336, 2018 12 09.
Article in English | MEDLINE | ID: mdl-30446226

ABSTRACT

The programmed death ligand-1 (PD-L1) (also called B7-H1 and CD274) belonging to the CD28 family of co-stimulatory molecules is ectopically expressed on the surface of various cancer cells. PD-L1 interacts with programmed death-1 (PD-1) on T cells to trigger an inhibitory signal that suppresses anti-tumor T cell responses as an important mechanism of tumor escape from anti-tumor immune response. Recent development of PD-1/PD-L1 blockades has provided novel immunotherapy strategies for cancers including non-small cell lung cancer (NSCLC). Although the therapy is quite effective for some patients with NSCLC, others are resistant to the treatment, so that regulatory mechanisms of PD-L1 in lung cancer cells need to be understood in detail. Here we analyzed effect of interferon-ß (IFN-ß) that can be produced in cancer microenvironment on PD-L1 expression in lung tumor cells. An addition of IFN-ß elevated PD-L1 expression in mouse and human lung cancer cell lines in culture. This phenomenon was totally dependent on JAK signaling molecules, while IRF9 deficiency in murine lung cancer cells partially attenuated the IFN-ß-induced increase in PD-L1. mTOR may not be significantly involved in the regulation of PD-L1, whereas PI3-K pathway played differential roles on PD-L1 mRNA and cell-surface PD-L1 expression, in the cells treated with IFN-ß. These results strongly suggest that the type I IFN receptor signal elicits an increase in PD-L1 expression in lung cancer cells through IRF9-dependent and independent pathways.


Subject(s)
B7-H1 Antigen/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interferon-beta/metabolism , Lung Neoplasms/metabolism , Signal Transduction , Up-Regulation , Animals , Cell Line, Tumor , HEK293 Cells , Humans , Mice
18.
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
19.
Fish Shellfish Immunol ; 66: 524-530, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28546020

ABSTRACT

Interferon regulatory factors (IRFs) are transcription factors which play important roles in regulating the expression of type I interferons (IFNs) and IFN-stimulated genes. IRF9 is one of the IRF family gene members which belongs to the IRF4 subfamily. Mammalian IRF9 has been known to be involved in antiviral responses as the DNA sequence recognition subunit of IFN-stimulated gene factor 3 (ISGF3) complex. In fish, only a few studies investigated the characteristics of IRF9 and the role in IFN signaling. In this study, we identified the IRF9 gene from miiuy croaker (mmiIRF9) and studied its feature and function. Sequence analysis showed the similarity of mmiIRF9 and other fish IRF9 genes. Structural and syntenic analysis showed the conservatism in fish IRF9 genes. The result of expression analysis in normal tissues and infected tissues and macrophages showed that mmiIRF9 expressed in all tested normal tissues and up-regulated expression in liver, kidney and macrophages after stimulated with poly(I:C). Luciferase reporter assays demonstrated the mmiIRF9 can induced IFNα and IFNß luciferase reporters and the cellular localization of mmiIRF9 was mainly distributed in the cytoplasm in Hela cells. Furthermore, the evolutionary analysis of IRF4 subfamily showed the IRF4 and IRF8 may be the most ancient and conservative genes in the evolution of this subfamily.


Subject(s)
Evolution, Molecular , Fish Proteins/genetics , Immunity, Innate , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Perciformes/genetics , Amino Acid Sequence , Animals , DNA, Complementary/genetics , DNA, Complementary/metabolism , Fish Proteins/chemistry , Fish Proteins/immunology , Gene Expression Regulation , Interferon-Stimulated Gene Factor 3, gamma Subunit/chemistry , Interferon-Stimulated Gene Factor 3, gamma Subunit/immunology , Perciformes/classification , Perciformes/immunology , Perciformes/metabolism , Phylogeny , Poly I-C/pharmacology , Sequence Alignment/veterinary , Tissue Distribution
20.
Immunogenetics ; 68(1): 67-76, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26518782

ABSTRACT

The interferon regulatory factor 9 (IRF9) gene is a member of the IRF family and has been shown to play functionally diverse roles in the regulation of the immune system. Previous study revealed the IRF9 gene resides within the reported quantitative trait locus (QTLs) for cytokine levels. The aims of this study were to identify genomic variants in IRF9 and to test the association between the variants and cytokine levels in pig. A synonymous single-nucleotide polymorphism (c.459A>G) was identified in exon 4 of the IRF9 gene. Association analysis in 300 piglets (Landrace, n=68; large white, n=158; and Songliao black, n=74) showed that this variant was significantly associated with the level of interferon (IFN)-γ and the ratio of IFN-γ to IL-10 in serum (P<0.05). Relative quantification of messenger RNA (mRNA) revealed that spleen had the highest expression level and individuals with genotype AA had higher expression than those with genotype AG. Transfection-based mRNA stability assay analysis further showed that the mutant allele G could reduce the RNA stability of IRF9. These findings suggest that the SNP (c.459A>G) could be a causative mutation for the association between IRF9 and the serum cytokine levels in swine.


Subject(s)
Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Interferon-gamma/blood , Interleukin-10/blood , Polymorphism, Single Nucleotide , Sus scrofa/genetics , Alternative Splicing , Animals , Classical Swine Fever , Gene Expression Regulation , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interferon-gamma/genetics , Interleukin-10/genetics , RNA Stability , Sus scrofa/blood , Swine/immunology , Viral Vaccines/immunology , Viral Vaccines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL